51
|
Dauda B, Molina SJ, Allen DS, Fuentes A, Ghosh N, Mauro M, Neale BM, Panofsky A, Sohail M, Zhang SR, Lewis ACF. Ancestry: How researchers use it and what they mean by it. Front Genet 2023; 14:1044555. [PMID: 36755575 PMCID: PMC9900027 DOI: 10.3389/fgene.2023.1044555] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/10/2023] [Indexed: 01/24/2023] Open
Abstract
Background: Ancestry is often viewed as a more objective and less objectionable population descriptor than race or ethnicity. Perhaps reflecting this, usage of the term "ancestry" is rapidly growing in genetics research, with ancestry groups referenced in many situations. The appropriate usage of population descriptors in genetics research is an ongoing source of debate. Sound normative guidance should rest on an empirical understanding of current usage; in the case of ancestry, questions about how researchers use the concept, and what they mean by it, remain unanswered. Methods: Systematic literature analysis of 205 articles at least tangentially related to human health from diverse disciplines that use the concept of ancestry, and semi-structured interviews with 44 lead authors of some of those articles. Results: Ancestry is relied on to structure research questions and key methodological approaches. Yet researchers struggle to define it, and/or offer diverse definitions. For some ancestry is a genetic concept, but for many-including geneticists-ancestry is only tangentially related to genetics. For some interviewees, ancestry is explicitly equated to ethnicity; for others it is explicitly distanced from it. Ancestry is operationalized using multiple data types (including genetic variation and self-reported identities), though for a large fraction of articles (26%) it is impossible to tell which data types were used. Across the literature and interviews there is no consistent understanding of how ancestry relates to genetic concepts (including genetic ancestry and population structure), nor how these genetic concepts relate to each other. Beyond this conceptual confusion, practices related to summarizing patterns of genetic variation often rest on uninterrogated conventions. Continental labels are by far the most common type of label applied to ancestry groups. We observed many instances of slippage between reference to ancestry groups and racial groups. Conclusion: Ancestry is in practice a highly ambiguous concept, and far from an objective counterpart to race or ethnicity. It is not uniquely a "biological" construct, and it does not represent a "safe haven" for researchers seeking to avoid evoking race or ethnicity in their work. Distinguishing genetic ancestry from ancestry more broadly will be a necessary part of providing conceptual clarity.
Collapse
Affiliation(s)
- Bege Dauda
- Center for Global Genomics and Health Equity, University of Pennsylvania, Philadelphia, PA, United States
| | - Santiago J. Molina
- Department of Sociology, Northwestern University, Evanston, IL, United States
| | - Danielle S. Allen
- Edmond & Lily Safra Center for Ethics, Harvard University, Cambridge, MA, United States
| | - Agustin Fuentes
- Department of Anthropology, Princeton University, Princeton, NJ, United States
| | - Nayanika Ghosh
- Department of the History of Science, Harvard University, Cambridge, MA, United States
| | - Madelyn Mauro
- Edmond & Lily Safra Center for Ethics, Harvard University, Cambridge, MA, United States
| | - Benjamin M. Neale
- Broad Institute of Harvard and MIT, Cambridge, MA, United States
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, United States
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Aaron Panofsky
- Institute for Society & Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Public Policy, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Sociology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mashaal Sohail
- Centro de Ciencias Genomicas (CCG), Universidad Nacional Autonoma de Mexico (UNAM), Cuernavaca, Morelos, Mexico
| | - Sarah R. Zhang
- University of California, Berkeley, Berkeley, CA, United States
| | - Anna C. F. Lewis
- Edmond & Lily Safra Center for Ethics, Harvard University, Cambridge, MA, United States
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
52
|
De Oliveira TC, Secolin R, Lopes-Cendes I. A review of ancestrality and admixture in Latin America and the caribbean focusing on native American and African descendant populations. Front Genet 2023; 14:1091269. [PMID: 36741309 PMCID: PMC9893294 DOI: 10.3389/fgene.2023.1091269] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/09/2023] [Indexed: 01/21/2023] Open
Abstract
Genomics can reveal essential features about the demographic evolution of a population that may not be apparent from historical elements. In recent years, there has been a significant increase in the number of studies applying genomic epidemiological approaches to understand the genetic structure and diversity of human populations in the context of demographic history and for implementing precision medicine. These efforts have traditionally been applied predominantly to populations of European origin. More recently, initiatives in the United States and Africa are including more diverse populations, establishing new horizons for research in human populations with African and/or Native ancestries. Still, even in the most recent projects, the under-representation of genomic data from Latin America and the Caribbean (LAC) is remarkable. In addition, because the region presents the most recent global miscegenation, genomics data from LAC may add relevant information to understand population admixture better. Admixture in LAC started during the colonial period, in the 15th century, with intense miscegenation between European settlers, mainly from Portugal and Spain, with local indigenous and sub-Saharan Africans brought through the slave trade. Since, there are descendants of formerly enslaved and Native American populations in the LAC territory; they are considered vulnerable populations because of their history and current living conditions. In this context, studying LAC Native American and African descendant populations is important for several reasons. First, studying human populations from different origins makes it possible to understand the diversity of the human genome better. Second, it also has an immediate application to these populations, such as empowering communities with the knowledge of their ancestral origins. Furthermore, because knowledge of the population genomic structure is an essential requirement for implementing genomic medicine and precision health practices, population genomics studies may ensure that these communities have access to genomic information for risk assessment, prevention, and the delivery of optimized treatment; thus, helping to reduce inequalities in the Western Hemisphere. Hoping to set the stage for future studies, we review different aspects related to genetic and genomic research in vulnerable populations from LAC countries.
Collapse
Affiliation(s)
- Thais C. De Oliveira
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- The Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Rodrigo Secolin
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- The Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Iscia Lopes-Cendes
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- The Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| |
Collapse
|
53
|
Soremekun O, Dib MJ, Rajasundaram S, Fatumo S, Gill D. Genetic heterogeneity in cardiovascular disease across ancestries: Insights for mechanisms and therapeutic intervention. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e8. [PMID: 38550935 PMCID: PMC10953756 DOI: 10.1017/pcm.2022.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 11/03/2024]
Abstract
Cardiovascular diseases (CVDs) are complex in their aetiology, arising due to a combination of genetics, lifestyle and environmental factors. By nature of this complexity, different CVDs vary in their molecular mechanisms, clinical presentation and progression. Although extensive efforts are being made to develop novel therapeutics for CVDs, genetic heterogeneity is often overlooked in the development process. By considering molecular mechanisms at an individual and ancestral level, a richer understanding of the influence of environmental and lifestyle factors can be gained and more refined therapeutic interventions can be developed. It is therefore expedient to understand the molecular and clinical heterogeneity in CVDs that exists across different populations. In this review, we highlight how the mechanisms underlying CVDs vary across diverse population ancestry groups due to genetic heterogeneity. We then discuss how such genetic heterogeneity is being leveraged to inform therapeutic interventions and personalised medicine, highlighting examples across the CVD spectrum. Finally, we present an overview of how polygenic risk scores and Mendelian randomisation can foster more robust insight into disease mechanisms and therapeutic intervention in diverse populations. Fulfilment of the vision of precision medicine requires more exhaustive leveraging of the genetic variability across diverse ancestry populations to improve our understanding of disease onset, progression and response to therapeutic intervention.
Collapse
Affiliation(s)
- Opeyemi Soremekun
- The African Computational Genomics (TACG) Research Group, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Marie-Joe Dib
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- British Heart Foundation Centre of Excellence, Imperial College London, London, UK
| | - Skanda Rajasundaram
- Centre for Evidence-Based Medicine, University of Oxford, Oxford, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Segun Fatumo
- The African Computational Genomics (TACG) Research Group, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Non-Communicable Disease Epidemiology (NCDE), London School of Hygiene and Tropical Medicine, London, UK
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- British Heart Foundation Centre of Excellence, Imperial College London, London, UK
| |
Collapse
|
54
|
Lee IH, Kong SW. ADGR: Admixture-Informed Differential Gene Regulation. Genes (Basel) 2023; 14:147. [PMID: 36672888 PMCID: PMC9859415 DOI: 10.3390/genes14010147] [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: 11/17/2022] [Revised: 12/15/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
The regulatory elements in proximal and distal regions of genes are involved in the regulation of gene expression. Risk alleles in intronic and intergenic regions may alter gene expression by modifying the binding affinity and stability of diverse DNA-binding proteins implicated in gene expression regulation. By focusing on the local ancestral structure of coding and regulatory regions using the paired whole-genome sequence and tissue-wide transcriptome datasets from the Genotype-Tissue Expression project, we investigated the impact of genetic variants, in aggregate, on tissue-specific gene expression regulation. Local ancestral origins of the coding region, immediate and distant upstream regions, and distal regulatory region were determined using RFMix with the reference panel from the 1000 Genomes Project. For each tissue, inter-individual variation of gene expression levels explained by concordant or discordant local ancestry between coding and regulatory regions was estimated. Compared to European, African descent showed more frequent change in local ancestral structure, with shorter haplotype blocks. The expression level of the Adenosine Deaminase Like (ADAL) gene was significantly associated with admixed ancestral structure in the regulatory region across multiple tissue types. Further validations are required to understand the impact of the local ancestral structure of regulatory regions on gene expression regulation in humans and other species.
Collapse
Affiliation(s)
- In-Hee Lee
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA 02215, USA
| | - Sek Won Kong
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA 02215, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
55
|
Khalil D, Giurgescu C, Misra DP, Templin T, Jenuwine E, Drury SS. Psychosocial Factors and Telomere Length Among Parents and Infants of Immigrant Arab American Families. Biol Res Nurs 2023; 25:137-149. [PMID: 36036284 PMCID: PMC10331091 DOI: 10.1177/10998004221124145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Immigrant Arab American families face multiple stressors related to migration and resettlement. Telomere length (TL) is an established biomarker of aging and psychosocial stress. No published studies have concurrently examined the association between maternal and paternal psychosocial factors and infants' TL. The purpose of this study was to: (1) compare mother, father, and infant TLs; (2) explore the association of maternal and paternal psychosocial factors (acculturative stress and depressive symptoms) with maternal and paternal TL; and (3) explore the association of maternal and paternal psychosocial factors with infants' TL among Arab American immigrants. Method: Using a cross-sectional exploratory design, a sample of 52 immigrant Arab American mother-father-infant triads were recruited from community centers. Data were collected in a single home visit when the infant was 6-24 months old. Each parent completed the study questionnaires addressing their psychosocial factors (acculturative stress, and depressive symptoms), then parents and infants provided buccal cell for TL measurement. Results: Maternal TL was positively correlated to infants' TL (r = .31, p = .04) and significantly shorter (p < .001). Paternal TL was not correlated with infant TL but was significantly shorter than infant's TL (p < .001). Maternal depression was significantly correlated with mothers' TL (r = .4, p = .007). Higher levels of maternal depressive symptoms were significantly associated with shorter infant TL when controlling for background characteristics. Conclusions: Our pilot study is the first study to examine maternal and paternal psychosocial factors related to migration and infants' TL. More research is needed to advance our understanding of the effects of immigration on the intergenerational transfer of stress and trauma.
Collapse
Affiliation(s)
- Dalia Khalil
- College of Nursing, Wayne State University, Detroit, MI, USA
| | - Carmen Giurgescu
- College of Nursing, University of Central Florida, Orlando, FL, USA
| | - Dawn P. Misra
- College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Thomas Templin
- College of Nursing, Wayne State University, Detroit, MI, USA
| | | | - Stacy S. Drury
- The Department of Psychiatry and Behavioral Sciences, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
56
|
Giudicelli GC, De Souza CMB, Veronese FV, Pereira LV, Hünemeier T, Vianna FSL. Precision medicine implementation challenges for APOL1 testing in chronic kidney disease in admixed populations. Front Genet 2022; 13:1016341. [PMID: 36588788 PMCID: PMC9797503 DOI: 10.3389/fgene.2022.1016341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Chronic Kidney Disease (CKD) is a public health problem that presents genetic and environmental risk factors. Two alleles in the Apolipoprotein L1 (APOL1) gene were associated with chronic kidney disease; these alleles are common in individuals of African ancestry but rare in European descendants. Genomic studies on Afro-Americans have indicated a higher prevalence and severity of chronic kidney disease in people of African ancestry when compared to other ethnic groups. However, estimates in low- and middle-income countries are still limited. Precision medicine approaches could improve clinical outcomes in carriers of risk alleles in the Apolipoprotein L1 gene through early diagnosis and specific therapies. Nevertheless, to enhance the definition of studies on these variants, it would be necessary to include individuals with different ancestry profiles in the sample, such as Latinos, African Americans, and Indigenous peoples. There is evidence that measuring genetic ancestry improves clinical care for admixed people. For chronic kidney disease, this knowledge could help establish public health strategies for monitoring patients and understanding the impact of the Apolipoprotein L1 genetic variants in admixed populations. Therefore, researchers need to develop resources, methodologies, and incentives for vulnerable and disadvantaged communities, to develop and implement precision medicine strategies and contribute to consolidating diversity in science and precision medicine in clinical practice.
Collapse
Affiliation(s)
- Giovanna Câmara Giudicelli
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Instituto Nacional de Ciência e Tecnologia de Genética Médica Populacional, Porto Alegre, RS, Brazil
| | - Celia Mariana Barbosa De Souza
- Departamento de Nefrologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Francisco Veríssimo Veronese
- Departamento de Nefrologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lygia V. Pereira
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Tábita Hünemeier
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
- Institut de Biologia Evolutiva, CSIC/Universitat Pompeu Fabra, Barcelona, Spain
| | - Fernanda Sales Luiz Vianna
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Instituto Nacional de Ciência e Tecnologia de Genética Médica Populacional, Porto Alegre, RS, Brazil
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Programa de Medicina Personalizada Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
57
|
Matshabane OP, Whitted CG, Koehly LM. Addressing diversity and inclusion challenges in global neuro-psychiatric and behavioral genomics research. Front Genet 2022; 13:1021649. [PMID: 36583023 PMCID: PMC9792473 DOI: 10.3389/fgene.2022.1021649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Advancements in neuro-psychiatric and behavioral genomics offer significant opportunities for better understanding the human brain, behavior and associated disorders. Such advancements may help us prevent, manage and/or cure complex conditions. The serious challenge confronted by these disciplines however is diversity. Both fields lack diversity in terms of genomic reference datasets needed for discovery research, engagement of diverse communities in translational research and in terms of diverse and multidisciplinary scientific teams. This is a challenge because diversity is needed on all levels in order to increase representation and inclusion of all populations across the globe as we move research activities forward. The lack of diversity can translate to an inability to use scientific innovations from these fields for the benefit of all people everywhere and signifies a missed opportunity to address pervasive global health inequities. In this commentary we identify three persistent barriers to reaching diversity targets while focusing on discovery and translational science. Additionally, we propose four suggestions on how to advance efforts and rapidly move towards achieving diversity and inclusion in neuro-psychiatric and behavioral genomics. Without systematically addressing the diversity gap within these fields, the benefits of the science may not be relevant and accessible to all people.
Collapse
Affiliation(s)
| | | | - Laura M. Koehly
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
58
|
Schneeweiss MR, Dale B, Ejaz R. Diagnosis and clinical presentation of two individuals with a rare TCF20 pathogenic variant. BMJ Case Rep 2022; 15:e248995. [PMID: 36593604 PMCID: PMC9730367 DOI: 10.1136/bcr-2022-248995] [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] [Indexed: 12/13/2022] Open
Abstract
TCF20-associated neurodevelopmental disorder (TAND) is a rare and phenotypically variable genetic condition. Common features include intellectual disability, neurobehavioural concerns, postnatal tall stature and hypotonia.Two unrelated early adolescent males were referred to genetics for assessment of developmental delay. The first male of Caucasian descent had a history of autism spectrum disorder (ASD), mitral valve prolapse and subtle craniofacial dysmorphisms. The second male of Somali descent had a history of intellectual disability, thick corpus callosum and ASD. Whole-exome sequencing revealed a pathogenic variant in TCF20 in both individuals. Further testing revealed that the former individual's mother was mosaic for the TCF20 pathogenic variant.We report two individuals with TCF20 pathogenic variants presenting with unique findings, including thick corpus callosum, family history of mosaicism and cardiac anomalies. These examples expand the TAND phenotype, describe associated dysmorphism in a minority group and highlight the importance of rare disease research.
Collapse
Affiliation(s)
| | - Breanne Dale
- Genetics and Metabolics Clinic, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Resham Ejaz
- Division of Genetics, Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster Children's Hospital, Hamilton, Ontario, Canada
| |
Collapse
|
59
|
Stallings SC, Cunningham-Erves J, Frazier C, Ichimura JS, Hurd TC, Jurinsky J, Acquaye A, Dalton JS, Wilkins CH. Development and Validation of the Perceptions of Research Trustworthiness Scale to Measure Trust Among Minoritized Racial and Ethnic Groups in Biomedical Research in the US. JAMA Netw Open 2022; 5:e2248812. [PMID: 36580334 PMCID: PMC9856656 DOI: 10.1001/jamanetworkopen.2022.48812] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
IMPORTANCE Historically, trust in biomedical research has been lower among minoritized racial and ethnic groups who are underrepresented in and excluded from research, with the same groups experiencing worse health outcomes. Unfortunately, instruments that measure trust may not capture components of trust relevant to minoritized racial and ethnic groups. OBJECTIVE To develop and validate a scale to measure trust in biomedical research among minoritized racial and ethnic groups. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional, community-based survey study compared trust and distrust in biomedical research among Black, Latino, and White subgroups in the US using the Perceptions of Research Trustworthiness (PoRT) scale. The scale was developed between March 22, 2016, and September 19, 2018, as part of this study, and its structure, reliability, and validity were examined during pilot (n = 381) and validation (n = 532) phases between February 4, 2019, and July 27, 2021. Convenience samples of adult participants (aged ≥18 years) were recruited locally (Nashville, Tennessee, and San Antonio, Texas) and nationally through the ResearchMatch and Cint online platforms. MAIN OUTCOMES AND MEASURES Overall and individual item Trust and Distrust subscale scores were compared. Overall Trust and Distrust scores were compared by race and ethnicity using a Kruskal-Wallis H test and individual item scores were compared using independent samples t test. RESULTS Of the 532 participants in the scale validation study, 144 (27.1%) were Black, 90 (16.9%) were Latino, and 282 (53.0%) were White. Participants had a median age of 43 years (range, 18-90 years), 352 (66.2%) were women, and 198 (37.2%) had educational attainment levels less than a college degree. Factor analysis of the 18-item PoRT scale revealed a 2-factor structure with two 9-item PoRT subscales (Trust and Distrust), which demonstrated high internal consistency (Cronbach α = 0.72 and 0.87, respectively). Mean (SD) Trust subscale scores were lower among Black (34.33 [2.02]) and Latino (34.55 [1.97]) participants compared with White participants (36.32 [1.81]; P < .001). Mean (SD) Distrust subscale scores were higher among Black (21.0 [2.15]) and Latino (20.53 [2.21]) participants compared with White participants (18.4 [2.03]; P < .001). Individual item results showed that Black and Latino participants were less trusting and more distrusting than White individuals on items related to risks, harms, secrecy, confidentiality, and privacy. CONCLUSIONS AND RELEVANCE These findings suggest that the PoRT scale incorporates trust and trustworthiness concepts relevant among Black and Latino individuals and may allow more precise assessment of trust in research among these groups.
Collapse
Affiliation(s)
- Sarah C. Stallings
- Division of Geriatric Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Meharry-Vanderbilt Alliance, Nashville, Tennessee
| | | | - Carleigh Frazier
- Public Health Department, University of California, Merced, Visalia
| | | | - Thelma C. Hurd
- Department of Surgery, University of Texas Health Science Center, San Antonio
- Department of Human and Organizational Development, Peabody College of Education and Human Development, Vanderbilt University, Nashville, Tennessee
| | - Jordan Jurinsky
- Department of Human and Organizational Development, Peabody College of Education and Human Development, Vanderbilt University, Nashville, Tennessee
| | - Amber Acquaye
- Department of Human and Organizational Development, Peabody College of Education and Human Development, Vanderbilt University, Nashville, Tennessee
| | | | - Consuelo H. Wilkins
- Division of Geriatric Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Meharry-Vanderbilt Alliance, Nashville, Tennessee
| |
Collapse
|
60
|
Blostein FA, Fisher J, Dou J, Schneper L, Ware EB, Notterman DA, Mitchell C, Bakulski KM. Polymethylation scores for prenatal maternal smoke exposure persist until age 15 and are detected in saliva in the Fragile Families and Child Wellbeing cohort. Epigenetics 2022; 17:2223-2240. [PMID: 35980258 PMCID: PMC9665138 DOI: 10.1080/15592294.2022.2112815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/05/2022] [Indexed: 01/18/2023] Open
Abstract
Prenatal maternal smoking is associated with low birthweight, neurological disorders, and asthma in exposed children. DNA methylation signatures can function as biomarkers of prenatal smoke exposure. However, the robustness of DNA methylation signatures across child ages, genetic ancestry groups, or tissues is not clear. Using coefficients from a meta-analysis of prenatal smoke exposure and DNA methylation in newborn cord blood, we created polymethylation scores of saliva DNA methylation from children at ages 9 and 15 in the Fragile Families and Child Wellbeing study. In the full sample at age 9 (n = 753), prenatal smoke exposure was associated with a 0.51 (95%CI: 0.35, 0.66) standard deviation higher polymethylation score. The direction and magnitude of the association was consistent in European and African genetic ancestry samples. In the full sample at age 15 (n = 747), prenatal smoke exposure was associated with a 0.48 (95%CI: 0.32, 0.63) standard deviation higher polymethylation score, and the association was attenuated among the European and Admixed-Latin genetic ancestry samples. The polymethylation score classified prenatal smoke exposure accurately (AUC age 9 = 0.77, age 15 = 0.76). Including the polymethylation score increased the AUC of base model covariates by 5 (95% CI: (2.1, 7.2)) percentage points, while including a single candidate site in the AHRR gene did not (P-value = 0.19). Polymethylation scores for prenatal smoking were portable across genetic ancestries and more accurate than an individual DNA methylation site. Polymethylation scores from saliva samples could serve as robust and practical biomarkers of prenatal smoke exposure.
Collapse
Affiliation(s)
- Freida A. Blostein
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Jonah Fisher
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - John Dou
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Lisa Schneper
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Erin B. Ware
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Daniel A. Notterman
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Colter Mitchell
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Kelly M. Bakulski
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| |
Collapse
|
61
|
Atkins R, Kelly TA, Johnson S, Deatrick JA, Wunnenberg M, Joseph PV, Pudasainee-Kapri S, Gage G, Pontes NMH, Cresse N, Williams W. Beliefs and willingness towards participating in genetic testing for depression in low-income and racial/ethnic minority mothers at-risk. Arch Psychiatr Nurs 2022; 41:153-165. [PMID: 36428043 PMCID: PMC11036348 DOI: 10.1016/j.apnu.2022.07.002] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 06/03/2022] [Accepted: 07/03/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE To identify mothers' salient normative, behavioral and control beliefs and willingness towards participating in genetic salivary testing for depression. DESIGN A qualitative, descriptive design was employed. 41 multi-ethnic mothers completed surveys that underwent directed content analysis according to The Theory of Planned Behavior. Percentages and frequency counts were used to categorize responses and calculate willingness. FINDINGS Salient beliefs included: Behavioral: Finding a cure/treatment for depression (29.3 %), Normative: Family would approve (46.3 %), and Church associates would disapprove (19.5 %). CONTROL Lacking information/explanations (34.1 %) as barriers, convenient locations (24.4 %) as facilitators. Most mothers indicated a willingness to participate (90.2 %). CONCLUSIONS Interventions should target families, emphasize benefits, explain purposes and procedures, and use community based participatory methods.
Collapse
Affiliation(s)
- Rahshida Atkins
- The College of New Jersey, School of Nursing and Health Sciences, 2000 Pennington Road, Room 113, Ewing, NJ 08102, USA.
| | - Terri-Ann Kelly
- Rutgers The State University of New Jersey School of Nursing-Camden, 530 Federal Street, Camden, NJ 08102, USA.
| | - Shanda Johnson
- New Jersey City University, 2039 John F. Kennedy Blvd, Jersey City, NJ 07305, USA.
| | - Janet A Deatrick
- The University of Pennsylvania School of Nursing, 418 Curie Boulevard, Philadelphia, PA, USA.
| | - Mary Wunnenberg
- Rutgers The State University of New Jersey School of Nursing-Camden, 530 Federal Street, Camden, NJ 08102, USA.
| | - Paule V Joseph
- Section of Sensory Science and Metabolism (SenSMet), National Institute on Alcohol Abuse and Alcoholism & National Institute of Nursing Research, National Institutes of Health, Bethesda, MD.
| | - Sangita Pudasainee-Kapri
- Rutgers The State University of New Jersey School of Nursing-Camden, 530 Federal Street, Camden, NJ 08102, USA.
| | - Gale Gage
- Essex County College, Division of Nursing & Health Sciences, 303 University Avenue, Newark, NJ 07102, USA
| | - Nancy M H Pontes
- Rutgers The State University of New Jersey School of Nursing-Camden, 530 Federal Street, Camden, NJ 08102, USA.
| | - Nancy Cresse
- Rutgers The State University of New Jersey School of Nursing-Camden, 530 Federal Street, Camden, NJ 08102, USA.
| | - Wanda Williams
- Rutgers The State University of New Jersey School of Nursing-Camden, 530 Federal Street, Camden, NJ 08102, USA.
| |
Collapse
|
62
|
Whole-genome analysis of monozygotic Brazilian twins discordant for type 1 narcolepsy: a case report. BMC Neurol 2022; 22:439. [DOI: 10.1186/s12883-022-02921-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/16/2022] [Indexed: 11/19/2022] Open
Abstract
Abstract
Background
Narcolepsy type 1 (NT1) is a rare and chronic neurological disease characterized by sudden sleep attacks, overwhelming daytime drowsiness, and cataplexy. When associated with a sudden loss of muscle tone (cataplexy) narcolepsy is classified as type 1, while the absence of cataplexy indicates type 2. Genetic, degenerative, and immunological hypotheses to explain the pathophysiology of NT1 are still a matter of debate. To contribute to the understanding of NT1 genetic basis, here we describe, for the first time, a whole genome analysis of a monozygotic twin pair discordant for NT1.
Case presentation
We present the case of a pair of 17-year-old male, monozygotic twins discordant for NT1. The affected twin had Epworth Sleepiness Scale (ESS) of 20 (can range from 0 to 24), cataplexy, hypnagogic hallucinations, polysomnography without abnormalities, multiple sleep latency tests (MSLT) positive for narcolepsy, a mean sleep latency of 3 min, sleep-onset REM periods SOREMPs of 5, presence of allele HLA-DQB1*06:02, and Hypocretin-1 level of zero pg/mL (normal values are > 200 pg/mL). The other twin had no narcolepsy symptoms (ESS of 4), normal polysomnography, MSLT without abnormalities, presence of allele HLA-DQB1*06:02, and Hypocretin-1 level of 396,74 pg/mL. To describe the genetic background for the NT1 discordant manifestations in this case, we present the whole-genome analysis of this monozygotic twin pair. The whole-genome comparison revealed that both twins have identical NT1 pathogenic mutations in known genes, such as HLA-DQB1*06:02:01, HLA-DRB1*11:01:02/*15:03:01. The affected twin has the expected clinical manifestation while the unaffected twin has an unexpected phenotype. The unaffected twin has significantly more frameshift mutations as compared to the affected twin (108 versus 75) and mutations that affect stop codons (61 versus 5 in stop gain, 26 versus 2 in start lost).
Conclusions
The differences observed in frameshift and stop codon mutations in the unaffected twin are consistent with loss-of-function effects and protective alleles, that are almost always associated with loss-of-function rare alleles. Also, overrepresentation analysis of genes containing variants with potential clinical relevance in the unaffected twin shows that most mutations are in genes related to immune regulation function, Golgi apparatus, MHC, and olfactory receptor. These observations support the hypothesis that NT1 has an immunological basis although protective mutations in non-HLA alleles might interfere with the expression of the NT1 phenotype and consequently, with the clinical manifestation of the disease.
Collapse
|
63
|
McGuinness S, Hughes L, Moss‐Morris R, Hunter M, Norton S, Moon Z. Adherence to adjuvant endocrine therapy among White British and ethnic minority breast cancer survivors in the United Kingdom. Eur J Cancer Care (Engl) 2022; 31:e13722. [PMID: 36255032 PMCID: PMC9787781 DOI: 10.1111/ecc.13722] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Around half of women do not take adjuvant endocrine therapy (AET) as prescribed. Research suggests that adherence rates vary across ethnic groups. This study compared AET adherences rates in White British women and women from minority ethnic groups in the United Kingdom. METHODS This is an observational study with 2001 breast cancer survivors recruited from outpatient clinics. Eligible women were diagnosed with primary breast cancer and prescribed AET within the last 3 years. Adherence was measured using the Medication Adherence Rating Scale. Eligible women were asked to complete a questionnaire pack that collected sociodemographic data such as age, relationship status and ethnicity. Independent samples t tests and χ2 tests were used to compare White British women and women from minority ethnic groups on self-reported adherence to AET. RESULTS Of White British women, 27.8% were classed as non-adherent, compared to 44.4% of women from minority ethnic groups. A logistic regression controlling for relevant demographics indicated that women from minority ethnic groups had a significantly higher risk of non-adherence than women who were White British (odds ratio = 1.50, p = 0.03) CONCLUSION: Rates of non-adherence to AET are higher in women from minority ethnic groups, which may contribute towards racial disparities in breast cancer outcomes. Research with larger and more diverse samples is needed to explore this further and to investigate the psychosocial factors driving differences in adherence.
Collapse
Affiliation(s)
- Serena McGuinness
- Faculty of Nursing Midwifery and Palliative CareKing's College LondonLondonUK
| | - Lyndsay Hughes
- Psychology Department, Institute of Psychiatry, Psychology and NeuroscienceKing's College London, Guy's HospitalLondonUK
| | - Rona Moss‐Morris
- Psychology Department, Institute of Psychiatry, Psychology and NeuroscienceKing's College London, Guy's HospitalLondonUK
| | - Myra Hunter
- Psychology Department, Institute of Psychiatry, Psychology and NeuroscienceKing's College London, Guy's HospitalLondonUK
| | - Sam Norton
- Psychology Department, Institute of Psychiatry, Psychology and NeuroscienceKing's College London, Guy's HospitalLondonUK,Centre for Rheumatic DiseasesKing's College London, Weston Education CentreLondonUK
| | - Zoe Moon
- Centre for Behavioural Medicine, School of PharmacyUniversity College LondonLondonUK
| |
Collapse
|
64
|
Horgan D, Mia R, Erhabor T, Hamdi Y, Dandara C, Lal JA, Fokom Domgue J, Ewumi O, Nyawira T, Meyer S, Kondji D, Francisco NM, Ikeda S, Chuah C, De Guzman R, Paul A, Reddy Nallamalla K, Park WY, Tripathi V, Tripathi R, Johns A, Singh MP, Phipps ME, Dube F, Whittaker K, Mukherji D, Rasheed HMA, Kozaric M, Pinto JA, Doral Stefani S, Augustovski F, Aponte Rueda ME, Fujita Alarcon R, Barrera-Saldana HA. Fighting Cancer around the World: A Framework for Action. Healthcare (Basel) 2022; 10:2125. [PMID: 36360466 PMCID: PMC9690702 DOI: 10.3390/healthcare10112125] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/11/2022] [Accepted: 10/21/2022] [Indexed: 09/05/2023] Open
Abstract
Tackling cancer is a major challenge right on the global level. Europe is only the tip of an iceberg of cancer around the world. Prosperous developed countries share the same problems besetting Europe-and the countries and regions with fewer resources and less propitious conditions are in many cases struggling often heroically against a growing tide of disease. This paper offers a view on these geographically wider, but essentially similar, challenges, and on the prospects for and barriers to better results in this ceaseless battle. A series of panels have been organized by the European Alliance for Personalised Medicine (EAPM) to identify different aspects of cancer care around the globe. There is significant diversity in key issues such as NGS, RWE, molecular diagnostics, and reimbursement in different regions. In all, it leads to disparities in access and diagnostics, patients' engagement, and efforts for a better understanding of cancer.
Collapse
Affiliation(s)
- Denis Horgan
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium;
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India; (J.A.L.); (V.T.)
| | - Rizwana Mia
- Grants, Innovation & Product Development, South African Medical Research Council, Francie Van Zijl Drive, Parow Valley, Cape Town 7505, South Africa;
| | - Tosan Erhabor
- Medical Laboratory Science Council of Nigeria (MLSCN), Durumi, Abuja 900110, Nigeria;
| | - Yosr Hamdi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia;
- Laboratory of Human and Experimental Pathology, Institut Pasteur de Tunis, Tunis 1002, Tunisia
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Observatory, Cape Town 7925, South Africa;
| | - Jonathan A. Lal
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India; (J.A.L.); (V.T.)
- Institute for Public Health Genomics, Department of Genetics and Cell Biology, GROW School of Oncology and Developmental Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Joel Fokom Domgue
- Departments of Epidemiology, and Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Centre, Houston, TX 77030, USA;
- Department of Obstetrics and Gynecology, Faculty of Medicine and Biomedical Sciences, University of Yaounde, Yaounde VF7W+4M9, Cameroon
| | - Oladimeji Ewumi
- Freelance Health Care, Life Sciences, Medical Artificial Intelligence Content Writer, Lagos 100253, Nigeria;
| | - Teresia Nyawira
- National Commission for Science, Technology and Innovation in Kenya (NACOSTI), Nairobi 00100, Kenya;
| | | | - Dominique Kondji
- Health & Development Communication, Building Capacities for Better Health in Africa, Yaounde P.O. Box 2032, Cameroon;
| | - Ngiambudulu M. Francisco
- Grupo de Investigação Microbiana e Imunológica, Instituto Nacional de Investigação em Saúde (National Institute for Health Research), Luanda 3635, Angola;
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan;
| | - Chai Chuah
- Singularity University, P.O. Box 165, Gold Coast, QLD 4227, Australia;
| | - Roselle De Guzman
- Oncology and Pain Management Section, Manila Central University–Filemon D. Tanchoco Medical Foundation Hospital, Caloocan 1400, Philippines;
| | - Anupriya Paul
- Department of Mathematics and Statistics, Faculty of Science, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India;
| | | | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Centre, Sungkyunkwan University, Seoul 06351, Korea;
| | - Vijay Tripathi
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India; (J.A.L.); (V.T.)
| | - Ravikant Tripathi
- Ministry of Labor, Health Department Government of India, New Delhi 110001, India;
| | - Amber Johns
- Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW 2010, Australia;
| | - Mohan P. Singh
- Centre of Biotechnology, University of Allahabad, Allahabad 211002, India;
| | - Maude E. Phipps
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya 47500, Selangor, Malaysia;
| | - France Dube
- Astra Zeneca, 1800 Concord Pike, Wilmington, DE 19803, USA;
| | | | - Deborah Mukherji
- Global Health Institute, American University of Beirut, Beirut VFXP+7QF, Lebanon;
- Department of Hematology/Oncology, American University of Beirut Medical Centre, Beirut P.O. Box 11-0236, Lebanon
| | | | - Marta Kozaric
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium;
| | - Joseph A. Pinto
- Centre for Basic and Translational Research, Auna Ideas, Lima 15036, Peru;
| | | | - Federico Augustovski
- Health Technology Assessment and Health Economics, Department of the Institute for Clinical Effectiveness and Health Policy (IECS-CONICET), Buenos Aires C1056ABH, Argentina;
| | | | - Ricardo Fujita Alarcon
- Centro de Genética y Biología Molecular, Universidad de San Martín de Porres, Lima 15024, Peru;
| | - Hugo A. Barrera-Saldana
- Innbiogem SC/Vitagenesis SA at National Laboratory for Services of Research, Development, and Innovation for the Pharma and Biotech Industries (LANSEIDI) of CONACyT Vitaxentrum Group, Monterrey 64630, Mexico;
- Schools of Medicine and Biology, Autonomous University of Nuevo Leon, Monterrey 66451, Mexico
| |
Collapse
|
65
|
Lee SSJ, Fullerton SM, McMahon CE, Bentz M, Saperstein A, Jeske M, Vasquez E, Foti N, Saco L, Shim JK. Targeting Representation: Interpreting Calls for Diversity in Precision Medicine Research. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2022; 95:317-326. [PMID: 36187415 PMCID: PMC9511949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Scientists have identified a "diversity gap" in genetic samples and health data, which have been drawn predominantly from individuals of European ancestry, as posing an existential threat to the promise of precision medicine. Inadequate inclusion as articulated by scientists, policymakers, and ethicists has prompted large-scale initiatives aimed at recruiting populations historically underrepresented in biomedical research. Despite explicit calls to increase diversity, the meaning of diversity - which dimensions matter for what outcomes and why - remain strikingly imprecise. Drawing on our document review and qualitative data from observations and interviews of funders and research teams involved in five precision medicine research (PMR) projects, we note that calls for increasing diversity often focus on "representation" as the goal of recruitment. The language of representation is used flexibly to refer to two objectives: achieving sufficient genetic variation across populations and including historically disenfranchised groups in research. We argue that these dual understandings of representation are more than rhetorical slippage, but rather allow for the contemporary collection of samples and data from marginalized populations to stand in as correcting historical exclusion of social groups towards addressing health inequity. We trace the unresolved historical debates over how and to what extent researchers should procure diversity in PMR and how they contributed to ongoing uncertainty about what axes of diversity matter and why. We argue that ambiguity in the meaning of representation at the outset of a study contributes to a lack of clear conceptualization of diversity downstream throughout subsequent phases of the study.
Collapse
Affiliation(s)
- Sandra Soo-Jin Lee
- Division of Ethics, Department of Medical Humanities
and Ethics, Columbia University, New York, NY, USA,To whom all correspondence should be addressed:
Sandra Soo-Jin Lee, Department of Medical Humanities and Ethics, Columbia
University, New York, NY; ; ORCID: https://www.orcid.org/0000-0002-2312-9814
| | - Stephanie M. Fullerton
- Department of Bioethics & Humanities, School of
Medicine, University of Washington, Seattle, WA, USA
| | - Caitlin E. McMahon
- Division of Ethics, Department of Medical Humanities
and Ethics, Columbia University, New York, NY, USA
| | - Michael Bentz
- Division of Ethics, Department of Medical Humanities
and Ethics, Columbia University, New York, NY, USA
| | | | - Melanie Jeske
- Department of Social and Behavioral Sciences,
University of California, San Francisco, CA, USA
| | - Emily Vasquez
- Department of Sociology, University of
Illinois-Chicago, Chicago, IL, USA
| | - Nicole Foti
- Department of Social and Behavioral Sciences,
University of California, San Francisco, CA, USA
| | - Larissa Saco
- Department of Sociology, University of California,
Davis, Davis, CA, USA
| | - Janet K. Shim
- Department of Social and Behavioral Sciences,
University of California, San Francisco, CA, USA
| |
Collapse
|
66
|
Abdelzaher H, Tawfik SM, Nour A, Abdelkader S, Elbalkiny ST, Abdelkader M, Abbas WA, Abdelnaser A. Climate change, human health, and the exposome: Utilizing OMIC technologies to navigate an era of uncertainty. Front Public Health 2022; 10:973000. [PMID: 36211706 PMCID: PMC9533016 DOI: 10.3389/fpubh.2022.973000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/17/2022] [Indexed: 01/25/2023] Open
Abstract
Climate change is an anthropogenic phenomenon that is alarming scientists and non-scientists alike. The emission of greenhouse gases is causing the temperature of the earth to rise and this increase is accompanied by a multitude of climate change-induced environmental exposures with potential health impacts. Tracking human exposure has been a major research interest of scientists worldwide. This has led to the development of exposome studies that examine internal and external individual exposures over their lifetime and correlate them to health. The monitoring of health has also benefited from significant technological advances in the field of "omics" technologies that analyze physiological changes on the nucleic acid, protein, and metabolism levels, among others. In this review, we discuss various climate change-induced environmental exposures and their potential health implications. We also highlight the potential integration of the technological advancements in the fields of exposome tracking, climate monitoring, and omics technologies shedding light on important questions that need to be answered.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, The American University in Cairo, New Cairo, Egypt
| |
Collapse
|
67
|
No person left behind: Mapping the health policy landscape for genomics research in the Caribbean. LANCET REGIONAL HEALTH. AMERICAS 2022; 15:100367. [PMID: 36778076 PMCID: PMC9904062 DOI: 10.1016/j.lana.2022.100367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The Caribbean has long been an under-represented geographical region in the field of genomics research. Such under-representation may result in Caribbean people being underserved by precision medicine and other public health benefits of genomics. A collaboration among regional and international researchers aims to address this issue through the H3ECaribbean project (Human Heredity, Environment, and Health in the Caribbean), which builds on the lessons and success of H3Africa. The Caribbean project aims to target issues of social justice by encouraging the inclusion of diverse Caribbean communities in genomics research. This paper explores a framework for the ethical and socially acceptable conduct of genomics research in the Caribbean, taking account of the cultural peculiarities of the region. This is done in part by exploring research ethics issues identified in indigenous communities in North America, Small Island Developing States, and similar endeavours from the African continent. The framework provides guidance for interacting with local community leaders, as well as detailing steps for obtaining informed consent of all participants. Specifically, the authors outline the methods to ensure effective interaction and enforce full transparency with study participants to combat historical neglect when working with under-represented communities in the Caribbean.
Collapse
|
68
|
Young JL, Halley MC, Anguiano B, Fernandez L, Bernstein JA, Wheeler MT, Tabor HK. Beyond race: Recruitment of diverse participants in clinical genomics research for rare disease. Front Genet 2022; 13:949422. [PMID: 36072659 PMCID: PMC9441547 DOI: 10.3389/fgene.2022.949422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: Despite recent attention to increasing diversity in clinical genomics research, researchers still struggle to recruit participants from varied sociodemographic backgrounds. We examined the experiences of parents from diverse backgrounds with enrolling their children in clinical genomics research on rare diseases. We explored the barriers and facilitators parents encountered and possible impacts of sociodemographic factors on their access to research.Methods: We utilized semi-structured interviews with parents of children participating in the Undiagnosed Diseases Network. Interview data were analyzed using comparative content analysis.Results: We interviewed 13 Hispanic, 11 non-Hispanic White, four Asian, and two biracial parents. Participants discussed different pathways to clinical genomics research for rare disease as well as how sociodemographic factors shaped families’ access. Themes focused on variation in: 1) reliance on providers to access research; 2) cultural norms around health communication; 3) the role of social capital in streamlining access; and 4) the importance of language-concordant research engagement.Conclusion: Our findings suggest that variables beyond race/ethnicity may influence access in clinical genomics research. Future efforts to diversify research participation should consider utilizing varied recruitment strategies to reach participants with diverse sociodemographic characteristics.
Collapse
Affiliation(s)
- Jennifer L. Young
- Stanford Center for Biomedical Ethics, Stanford University School of Medicine, Stanford, CA, United States
- Center for Genetic Medicine, Northwestern Feinberg School of Medicine, Chicago, IL, United States
- *Correspondence: Jennifer L. Young,
| | - Meghan C. Halley
- Stanford Center for Biomedical Ethics, Stanford University School of Medicine, Stanford, CA, United States
| | - Beatriz Anguiano
- Human Genetics and Genetic Counseling, Stanford University School of Medicine, Stanford, CA, United States
| | - Liliana Fernandez
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, United States
| | - Jonathan A. Bernstein
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Matthew T. Wheeler
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Holly K. Tabor
- Stanford Center for Biomedical Ethics, Stanford University School of Medicine, Stanford, CA, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | | |
Collapse
|
69
|
Khan AT, Gogarten SM, McHugh CP, Stilp AM, Sofer T, Bowers ML, Wong Q, Cupples LA, Hidalgo B, Johnson AD, McDonald MLN, McGarvey ST, Taylor MR, Fullerton SM, Conomos MP, Nelson SC. Recommendations on the use and reporting of race, ethnicity, and ancestry in genetic research: Experiences from the NHLBI TOPMed program. CELL GENOMICS 2022; 2:100155. [PMID: 36119389 PMCID: PMC9481067 DOI: 10.1016/j.xgen.2022.100155] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
How race, ethnicity, and ancestry are used in genomic research has wide-ranging implications for how research is translated into clinical care and incorporated into public understanding. Correlation between race and genetic ancestry contributes to unresolved complexity for the scientific community, as illustrated by heterogeneous definitions and applications of these variables. Here, we offer commentary and recommendations on the use of race, ethnicity, and ancestry across the arc of genetic research, including data harmonization, analysis, and reporting. While informed by our experiences as researchers affiliated with the NHLBI Trans-Omics for Precision Medicine (TOPMed) program, these recommendations are applicable to basic and translational genomic research in diverse populations with genome-wide data. Moving forward, considerable collaborative effort will be required to ensure that race, ethnicity, and ancestry are described and used appropriately to generate scientific knowledge that yields broad and equitable benefit.
Collapse
Affiliation(s)
- Alyna T. Khan
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Institute for Public Health Genetics, University of Washington, Seattle, WA, USA
| | | | - Caitlin P. McHugh
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Adrienne M. Stilp
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Tamar Sofer
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
| | - Michael L. Bowers
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Quenna Wong
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Bertha Hidalgo
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew D. Johnson
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Merry-Lynn N. McDonald
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephen T. McGarvey
- Department of Epidemiology and International Health Institute, Brown University School of Public Health, Providence, RI, USA
- Department of Anthropology, Brown University, Providence, RI, USA
| | - Matthew R.G. Taylor
- Department of Medicine, Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Sarah C. Nelson
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Institute for Public Health Genetics, University of Washington, Seattle, WA, USA
| |
Collapse
|
70
|
Gould D, Walker R, Makari-Judson G, Seven M. Experiences of individuals with a variant of uncertain significance on genetic testing for hereditary cancer risks: a mixed method systematic review. J Community Genet 2022; 13:371-379. [PMID: 35819584 DOI: 10.1007/s12687-022-00600-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/04/2022] [Indexed: 10/17/2022] Open
Abstract
The expansion of Multi-Gene Panel Testing (MGPT) has led to increased detection of variants of uncertain significance (VUS) among individuals with personal or family history of cancer. However, having a VUS result can impact on emotional and psychological wellbeing and cause challenges for non-geneticist healthcare providers. The purpose of this mixed methods systematic review was to examine what is currently known about the experiences of individuals with a VUS on genetic testing for inherited cancer susceptibility. The initial search was conducted in June 2020 using PUBMED, CINAHL, Web of Science, and PsychInfo according to the Joanna Briggs methodology for systematic reviews. A total of 18 studies met the inclusion criteria. Studies included in this review identified a range of emotional reactions to a VUS result, a general lack of understanding of a VUS result and its implications, frustration with a lack of healthcare provider knowledge, and a need for clear communication with healthcare providers. This review identified critical gaps in current knowledge to guide genetic counseling praxis, specifically in the knowledge of communication patterns and methods of improving communication with healthcare providers and family members and preferred risk management strategies. This will help to improve the counseling process and the management of care during and after genetic testing.
Collapse
Affiliation(s)
| | - Rachel Walker
- University of Massachusetts Amherst, Amherst, MA, USA
| | | | - Memnun Seven
- University of Massachusetts Amherst, Amherst, MA, USA
| |
Collapse
|
71
|
Torres-Gonzalez E, Makova KD. Exploring the Effects of Mitonuclear Interactions on Mitochondrial DNA Gene Expression in Humans. Front Genet 2022; 13:797129. [PMID: 35846132 PMCID: PMC9277102 DOI: 10.3389/fgene.2022.797129] [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: 10/19/2021] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Most mitochondrial protein complexes include both nuclear and mitochondrial gene products, which coevolved to work together. This coevolution can be disrupted due to disparity in genetic ancestry between the nuclear and mitochondrial genomes in recently admixed populations. Such mitonuclear DNA discordance might result in phenotypic effects. Several nuclear-encoded proteins regulate expression of mitochondrial DNA (mtDNA) genes. We hypothesized that mitonuclear DNA discordance affects expression of genes encoded by mtDNA. To test this, we utilized the data from the GTEx project, which contains expression levels for ∼100 African Americans and >600 European Americans. The varying proportion of African and European ancestry in recently admixed African Americans provides a range of mitonuclear discordance values, which can be correlated with mtDNA gene expression levels (adjusted for age and ischemic time). In contrast, European Americans did not undergo recent admixture. We demonstrated that, for most mtDNA protein-coding genes, expression levels in energetically-demanding tissues were lower in African Americans than in European Americans. Furthermore, gene expression levels were lower in individuals with higher mitonuclear discordance, independent of population. Moreover, we found a negative correlation between mtDNA gene expression and mitonuclear discordance. In African Americans, the average value of African ancestry was higher for nuclear-encoded mitochondrial than non-mitochondrial genes, facilitating a match in ancestry with the mtDNA and more optimal interactions. These results represent an example of a phenotypic effect of mitonuclear discordance on human admixed populations, and have potential biomedical applications.
Collapse
Affiliation(s)
| | - Kateryna D. Makova
- Department of Biology, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
72
|
Siqueira ADSE, Fontes ANB, Figueiredo GSF, Guimarães HIP, Treptow JP, Costa MNDM, Souza PA, Rocha RT. Ciência, Tecnologia e Inovações em Oncologia. REVISTA BRASILEIRA DE CANCEROLOGIA 2022. [DOI: 10.32635/2176-9745.rbc.2022v68n2.2809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A medicina genômica e a saúde de precisão deram seu primeiro grande passo no início dos anos 2000. O Projeto Genoma Humano (HGP, do inglês Human Genome Project), iniciado em 1990 e finalizado em 2003, não apenas impactou de forma significativa a compreensão sobre a arquitetura do genoma humano e a correlação deste com diferentes doenças, como também gerou uma revolução tecnológica multidisciplinar. Vinte anos depois, ainda são consideradas inovações em todo mundo, uma vez que sua implementação pelos sistemas de saúde não é simples, requerendo uma gama de ações e iniciativas complexas. Os rápidos avanços nos métodos de diagnóstico molecular, terapias avançadas e medicina de precisão enfatizam a necessidade da tradução desses conhecimentos para otimizar a aplicabilidade nos serviços de saúde da população. O Genomas Brasil apresenta uma prova de conceito que fornecerá dados importantes para a demonstração do valor da saúde de precisão para o SUS, subsidiando a futura tomada de decisão em relação à implementação, à projeção e ao escalonamento para o sistema de saúde público. O programa é fundamental para a transição adequada para um sistema de saúde baseado em saúde de precisão, etapa crítica para a adequação de estratégias, metodologias e protocolos e para o gerenciamento de riscos e potenciais obstáculos a serem enfrentados. Além disso, pretende iniciar uma revolução no SUS por meio do uso de estratégias de fronteira do conhecimento para fornecer o mais preciso e moderno cuidado à saúde à população brasileira, trazendo grandes transformações para a força de trabalho em saúde, para o avanço do conhecimento científico e para a indústria de saúde nacional.
Collapse
|
73
|
Marotta A. When AI Is Wrong: Addressing Liability Challenges in Women’s Healthcare. JOURNAL OF COMPUTER INFORMATION SYSTEMS 2022. [DOI: 10.1080/08874417.2022.2089773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
74
|
Silva P, Dahlke DV, Smith ML, Charles W, Gomez J, Ory MG, Ramos KS. An Idealized Clinicogenomic Registry to Engage Underrepresented Populations Using Innovative Technology. J Pers Med 2022; 12:713. [PMID: 35629136 PMCID: PMC9144063 DOI: 10.3390/jpm12050713] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/18/2022] [Accepted: 04/26/2022] [Indexed: 11/26/2022] Open
Abstract
Current best practices in tumor registries provide a glimpse into a limited time frame over the natural history of disease, usually a narrow window around diagnosis and biopsy. This creates challenges meeting public health and healthcare reimbursement policies that increasingly require robust documentation of long-term clinical trajectories, quality of life, and health economics outcomes. These challenges are amplified for underrepresented minority (URM) and other disadvantaged populations, who tend to view the institution of clinical research with skepticism. Participation gaps leave such populations underrepresented in clinical research and, importantly, in policy decisions about treatment choices and reimbursement, thus further augmenting health, social, and economic disparities. Cloud computing, mobile computing, digital ledgers, tokenization, and artificial intelligence technologies are powerful tools that promise to enhance longitudinal patient engagement across the natural history of disease. These tools also promise to enhance engagement by giving participants agency over their data and addressing a major impediment to research participation. This will only occur if these tools are available for use with all patients. Distributed ledger technologies (specifically blockchain) converge these tools and offer a significant element of trust that can be used to engage URM populations more substantively in clinical research. This is a crucial step toward linking composite cohorts for training and optimization of the artificial intelligence tools for enhancing public health in the future. The parameters of an idealized clinical genomic registry are presented.
Collapse
Affiliation(s)
- Patrick Silva
- Health Science Center, Texas A&M University, 8441 Riverside Pkwy, Bryan, TX 77807, USA; (J.G.); (K.S.R.)
| | - Deborah Vollmer Dahlke
- School of Public Health, Texas A&M Health Science Center, 212 Adriance Lab Rd., College Station, TX 77843, USA; (D.V.D.); (M.L.S.); (M.G.O.)
| | - Matthew Lee Smith
- School of Public Health, Texas A&M Health Science Center, 212 Adriance Lab Rd., College Station, TX 77843, USA; (D.V.D.); (M.L.S.); (M.G.O.)
| | - Wendy Charles
- BurstIQ, 9635 Maroon Circle, #310, Englewood, CO 80112, USA;
| | - Jorge Gomez
- Health Science Center, Texas A&M University, 8441 Riverside Pkwy, Bryan, TX 77807, USA; (J.G.); (K.S.R.)
| | - Marcia G. Ory
- School of Public Health, Texas A&M Health Science Center, 212 Adriance Lab Rd., College Station, TX 77843, USA; (D.V.D.); (M.L.S.); (M.G.O.)
| | - Kenneth S. Ramos
- Health Science Center, Texas A&M University, 8441 Riverside Pkwy, Bryan, TX 77807, USA; (J.G.); (K.S.R.)
| |
Collapse
|
75
|
Auwerx C, Sadler MC, Reymond A, Kutalik Z. From pharmacogenetics to pharmaco-omics: Milestones and future directions. HGG ADVANCES 2022; 3:100100. [PMID: 35373152 PMCID: PMC8971318 DOI: 10.1016/j.xhgg.2022.100100] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The origins of pharmacogenetics date back to the 1950s, when it was established that inter-individual differences in drug response are partially determined by genetic factors. Since then, pharmacogenetics has grown into its own field, motivated by the translation of identified gene-drug interactions into therapeutic applications. Despite numerous challenges ahead, our understanding of the human pharmacogenetic landscape has greatly improved thanks to the integration of tools originating from disciplines as diverse as biochemistry, molecular biology, statistics, and computer sciences. In this review, we discuss past, present, and future developments of pharmacogenetics methodology, focusing on three milestones: how early research established the genetic basis of drug responses, how technological progress made it possible to assess the full extent of pharmacological variants, and how multi-dimensional omics datasets can improve the identification, functional validation, and mechanistic understanding of the interplay between genes and drugs. We outline novel strategies to repurpose and integrate molecular and clinical data originating from biobanks to gain insights analogous to those obtained from randomized controlled trials. Emphasizing the importance of increased diversity, we envision future directions for the field that should pave the way to the clinical implementation of pharmacogenetics.
Collapse
Affiliation(s)
- Chiara Auwerx
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Marie C. Sadler
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Lausanne, Switzerland
| |
Collapse
|
76
|
Moreau CS, Darby AM, Demery AJC, Hernández LMA, Meaders CL. A framework for educating and empowering students by teaching about history and consequences of bias in STEM. Pathog Dis 2022; 80:6564730. [PMID: 35389476 PMCID: PMC9053302 DOI: 10.1093/femspd/ftac006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 12/02/2022] Open
Abstract
Racism and bias are pervasive in society—and science, technology, engineering, and mathematics (STEM) fields are not immune to these issues. It is imperative that we educate ourselves and our students about the history and consequences of this bias in STEM, investigate the research showing bias toward marginalized groups, understand how to interpret misuses of science in perpetuating bias, and identify advances and solutions to overcome racism and bias throughout our professional and personal lives. Here, we present one model for teaching a universal course for participants of all professional stages to address these issues and initiate solutions. As very few institutions require students to enroll in courses on racism and bias in STEM or even offer such courses, our curriculum could be used as a blueprint for implementation across institutions. Ultimately, institutions and academic disciplines can incorporate this important material with more region and/or discipline specific studies of bias.
Collapse
Affiliation(s)
- Corrie S Moreau
- Cornell University, Department of Entomology, Ithaca, NY, USA.,Cornell University, Department of Ecology and Evolutionary Biology, Ithaca, NY, USA
| | - Andrea M Darby
- Cornell University, Department of Entomology, Ithaca, NY, USA
| | | | | | - Clara L Meaders
- University of California San Diego, Division of Biological Sciences, Section of Cell and Developmental Biology, San Diego, CA, USA
| |
Collapse
|
77
|
Osman A, Jonasson J. Cross-ethnic analysis of common gene variants in hemostasis show lopsided representation of global populations in genetic databases. BMC Med Genomics 2022; 15:69. [PMID: 35337356 PMCID: PMC8957123 DOI: 10.1186/s12920-022-01220-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/21/2022] [Indexed: 11/12/2022] Open
Abstract
A majority of studies reporting human genetic variants were performed in populations of European ancestry whereas other global populations, and particularly many ethnolinguistic groups in other continents, are heavily underrepresented in these studies. To investigate the extent of this disproportionate representation of global populations concerning variants of significance to thrombosis and hemostasis, 845 single nucleotide polymorphisms (SNPs) in and around 34 genes associated with thrombosis and hemostasis and included in the commercial Axiom Precision Medicine Research Array (PMRA) were evaluated, using gene frequencies in 3 African (Somali and Luhya in East Africa, and Yoruba in West Africa) and 14 non-African (admixed American, East Asian, European, South Asian, and sub-groups) populations. Among the populations studied, Europeans were observed to be the best represented population by the hemostatic SNPs included in the PMRA. The European population also presented the largest number of common pharmacogenetic and pathogenic hemostatic variants reported in the ClinVar database. The number of such variants decreased the farther the genetic distance a population was from Europeans, with Yoruba and East Asians presenting the least number of clinically significant hemostatic SNPs in ClinVar while also being the two genetically most distinct populations from Europeans among the populations compared. Current study shows the lopsided representation of global populations as regards to hemostatic genetic variants listed in different commercial SNP arrays, such as the PMRA, and reported in genetic databases while also underlining the importance of inclusion of non-European ethnolinguistic populations in genomics studies designed to discover variants of significance to bleeding and thrombotic disorders.
Collapse
Affiliation(s)
- Abdimajid Osman
- Department of Clinical Chemistry, University Hospital in Linköping, Ing. 64, Plan 11, 581 85, Linköping, Sweden. .,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Jon Jonasson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Clinical Genetics, University Hospital in Linköping, Linköping, Sweden
| |
Collapse
|
78
|
Song L, Liu H, Brinkman FSL, Gill E, Griffiths EJ, Hsiao WWL, Savić-Kallesøe S, Moreira S, Van Domselaar G, Zawati MH, Joly Y. Addressing Privacy Concerns in Sharing Viral Sequences and Minimum Contextual Data in a Public Repository During the COVID-19 Pandemic. Front Genet 2022; 12:716541. [PMID: 35401651 PMCID: PMC8988250 DOI: 10.3389/fgene.2021.716541] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/23/2021] [Indexed: 11/28/2022] Open
Abstract
COVID-19 was declared to be a pandemic in March 2020 by the World Health Organization. Timely sharing of viral genomic sequencing data accompanied by a minimal set of contextual data is essential for informing regional, national, and international public health responses. Such contextual data is also necessary for developing, and improving clinical therapies and vaccines, and enhancing the scientific community’s understanding of the SARS-CoV-2 virus. The Canadian COVID-19 Genomics Network (CanCOGeN) was launched in April 2020 to coordinate and upscale existing genomics-based COVID-19 research and surveillance efforts. CanCOGeN is performing large-scale sequencing of both the genomes of SARS-CoV-2 virus samples (VirusSeq) and affected Canadians (HostSeq). This paper addresses the privacy concerns associated with sharing the viral sequence data with a pre-defined set of contextual data describing the sample source and case attribute of the sequence data in the Canadian context. Currently, the viral genome sequences are shared by provincial public health laboratories and their healthcare and academic partners, with the Canadian National Microbiology Laboratory and with publicly accessible databases. However, data sharing delays and the provision of incomplete contextual data often occur because publicly releasing such data triggers privacy and data governance concerns. The CanCOGeN Ethics and Governance Expert Working Group thus has investigated several privacy issues cited by CanCOGeN data providers/stewards. This paper addresses these privacy concerns and offers insights primarily in the Canadian context, although similar privacy considerations also exist in other jurisdictions. We maintain that sharing viral sequencing data and its limited associated contextual data in the public domain generally does not pose insurmountable privacy challenges. However, privacy risks associated with reidentification should be actively monitored due to advancements in reidentification methods and the evolving pandemic landscape. We also argue that during a global health emergency such as COVID-19, privacy should not be used as a blanket measure to prevent such genomic data sharing due to the significant benefits it provides towards public health responses and ongoing research activities.
Collapse
Affiliation(s)
- Lingqiao Song
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Hanshi Liu
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- *Correspondence: Hanshi Liu,
| | | | - Erin Gill
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | | | - William W. L Hsiao
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | | | - Sandrine Moreira
- Institut National de Santé Publique du Québec, Québec, QC, Canada
| | | | - Ma’n H. Zawati
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Yann Joly
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
79
|
Akam EY, Nuako AA, Daniel AK, Stanford FC. Racial Disparities and Cardiometabolic Risk: New Horizons of Intervention and Prevention. Curr Diab Rep 2022; 22:129-136. [PMID: 35175453 PMCID: PMC9908372 DOI: 10.1007/s11892-022-01451-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/03/2022] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW Cardiometabolic diseases are a leading cause of morbidity and mortality in the USA and disproportionately impact racial and ethnic minorities. Multiple factors contribute to this disparity including genetic and socioeconomic factors, the latter of which contributes to disparities both through systemic barriers such as healthcare access and by directly impacting metabolism through epigenetics and environment-related alterations in the gut microbiome. This review will discuss advances in medicine that can be used to identify, prognosticate, and treat cardiometabolic diseases, and how these may be used to address existing disparities. RECENT FINDINGS There is growing research aimed at identifying novel cardiometabolic disease targets and expanding the use of existing pharmacotherapies based on comorbidities. Advances in metabolomics and genomics can give insight into an individual's unique biochemical profile, providing the means for earlier identification of disease and specific treatment targets. Moreover, developments in telehealth and related medical device technologies can expand access to underserved minority populations and improve control of chronic conditions such as diabetes and hypertension. Precision medicine may be integral to bridging the racial gap in cardiometabolic disease outcomes. Developments in genomics, metabolomics, wearable medical devices, and telehealth can result in personalized treatments for patients that account for the socioeconomic and genetic factors that contribute to poor health outcomes in minorities. As research in this field rapidly progresses, special efforts must be made to ensure inclusion of racial and ethnic minority populations in clinical research and equal access to all treatment modalities.
Collapse
Affiliation(s)
- Eftitan Y Akam
- Massachusetts General Hospital Department of Medicine, 55 Fruit Street, Boston, MA, 02114, USA.
- Massachusetts General Hospital Department of Pediatrics, 55 Fruit Street, Boston, MA, 02114, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| | - Akua A Nuako
- Massachusetts General Hospital Department of Medicine, 55 Fruit Street, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Afkera K Daniel
- Massachusetts General Hospital Department of Medicine, 55 Fruit Street, Boston, MA, 02114, USA
- Massachusetts General Hospital Department of Pediatrics, 55 Fruit Street, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Fatima Cody Stanford
- Harvard Medical School, Boston, MA, 02115, USA
- Massachusetts General Hospital, MGH Weight Center, Department of Medicine-Division of Endocrinology-Neuroendocrine, Department of Pediatrics-Division of Endocrinology, Nutrition Obesity Research Center at Harvard (NORCH), 50 Staniford Street, Boston, MA, 02114, USA
| |
Collapse
|
80
|
Hirko KA, Rocque G, Reasor E, Taye A, Daly A, Cutress RI, Copson ER, Lee DW, Lee KH, Im SA, Park YH. The impact of race and ethnicity in breast cancer-disparities and implications for precision oncology. BMC Med 2022; 20:72. [PMID: 35151316 PMCID: PMC8841090 DOI: 10.1186/s12916-022-02260-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide and is one of the leading causes of cancer death. The incidence, pathological features, and clinical outcomes in breast cancer differ by geographical distribution and across racial and ethnic populations. Importantly, racial and ethnic diversity in breast cancer clinical trials is lacking, with both Blacks and Hispanics underrepresented. In this forum article, breast cancer researchers from across the globe discuss the factors contributing to racial and ethnic breast cancer disparities and highlight specific implications of precision oncology approaches for equitable provision of breast cancer care to improve outcomes and address disparities.
Collapse
Affiliation(s)
- Kelly A Hirko
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| | - Gabrielle Rocque
- Department of Internal Medicine, Division of Hematology Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erica Reasor
- Department of Internal Medicine, Division of Hematology Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ammanuel Taye
- Department of Internal Medicine, Division of Hematology Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alex Daly
- Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton and University Hospital Southampton, Southampton, SO16 6YD, UK
| | - Ramsey I Cutress
- Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton and University Hospital Southampton, Southampton, SO16 6YD, UK
| | - Ellen R Copson
- Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton and University Hospital Southampton, Southampton, SO16 6YD, UK
| | - Dae-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro Gangnam-gu, Seoul, 06351, Korea
| |
Collapse
|
81
|
Mbarek H, Devadoss Gandhi G, Selvaraj S, Al-Muftah W, Badji R, Al-Sarraj Y, Saad C, Darwish D, Alvi M, Fadl T, Yasin H, Alkuwari F, Razali R, Aamer W, Abbaszadeh F, Ahmed I, Mokrab Y, Suhre K, Albagha O, Fakhro K, Badii R, Ismail SI, Althani A. Qatar Genome: Insights on Genomics from the Middle East. Hum Mutat 2022; 43:499-510. [PMID: 35112413 DOI: 10.1002/humu.24336] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/16/2021] [Accepted: 01/29/2022] [Indexed: 11/09/2022]
Abstract
Despite recent biomedical breakthroughs and large genomic studies growing momentum, the Middle Eastern population, home to over 400 million people, is under-represented in the human genome variation databases. Here we describe insights from phase 1 of the Qatar Genome Program with whole genome sequenced 6,047 individuals from Qatar. We identified more than 88 million variants of which 24 million are novel and 23 million are singletons. Consistent with the high consanguinity and founder effects in the region, we found that several rare deleterious variants were more common in the Qatari population while others seem to provide protection against diseases and have shaped the genetic architecture of adaptive phenotypes. These results highlight the value of our data as a resource to advance genetic studies in the Arab and neighbouring Middle Eastern populations and will significantly boost the current efforts to improve our understanding of global patterns of human variations, human history and genetic contributions to health and diseases in diverse populations. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hamdi Mbarek
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Geethanjali Devadoss Gandhi
- Department of Biomedical Sciences, College of Health Sciences, Qatar University.,College of Health & Life Sciences, Hamad Bin Khalifa University, Education City, Doha, Qatar
| | - Senthil Selvaraj
- Department of Biomedical Sciences, College of Health Sciences, Qatar University
| | - Wadha Al-Muftah
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Radja Badji
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Yasser Al-Sarraj
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar.,Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Chadi Saad
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Dima Darwish
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Muhammad Alvi
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Tasnim Fadl
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Heba Yasin
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Fatima Alkuwari
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Rozaimi Razali
- Department of Biomedical Sciences, College of Health Sciences, Qatar University
| | - Waleed Aamer
- Human Genetics Department, Sidra Medicine, Doha, Qatar
| | | | - Ikhlak Ahmed
- Sidra Medicine, Biomedical Informatics - Research Branch, Doha, Qatar
| | - Younes Mokrab
- Human Genetics Department, Sidra Medicine, Doha, Qatar
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Omar Albagha
- College of Health & Life Sciences, Hamad Bin Khalifa University, Education City, Doha, Qatar.,Center of Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Khalid Fakhro
- Department of Biomedical Sciences, College of Health Sciences, Qatar University
| | - Ramin Badii
- Molecular Genetics Laboratory, Hamad Medical Corporation, Doha, Qatar
| | | | - Asma Althani
- Qatar Genome Program, Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
82
|
Atutornu J, Milne R, Costa A, Patch C, Middleton A. Towards equitable and trustworthy genomics research. EBioMedicine 2022; 76:103879. [PMID: 35158310 PMCID: PMC8850759 DOI: 10.1016/j.ebiom.2022.103879] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/04/2022] [Accepted: 01/28/2022] [Indexed: 12/11/2022] Open
Abstract
The representation of traditionally scientifically underserved groups in genomic research continues to be low despite concerns about equity and social justice and the scientific and clinical need. Among the factors that account for this are a lack of trust in the research community and limited diversity in this community. The success of the multiple initiatives that aim to improve representation relies on the willingness of underrepresented populations to make data and samples available for research and clinical use. In this narrative review, we propose that this requires building trust, and set out four approaches to demonstrating trustworthiness, including increasing diversity in the research workforce, and meaningful engagement with underrepresented communities in a culturally and linguistically appropriate manner. Capacity building globally will ensure that actual and perceived exploitation and ‘helicopter’ research could be eliminated.
Collapse
Affiliation(s)
- Jerome Atutornu
- Engagement and Society, Wellcome Connecting Science, Wellcome Genome Campus, Cambridge CB10 1SA, UK; School of Health and Sports Sciences, University of Suffolk, Ipswich, IP4 1QJ
| | - Richard Milne
- Engagement and Society, Wellcome Connecting Science, Wellcome Genome Campus, Cambridge CB10 1SA, UK; Kavli Centre for Ethics, Science and the Public, Faculty of Education, University of Cambridge, CB2 8PQ
| | - Alesia Costa
- Engagement and Society, Wellcome Connecting Science, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Christine Patch
- Engagement and Society, Wellcome Connecting Science, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Anna Middleton
- Engagement and Society, Wellcome Connecting Science, Wellcome Genome Campus, Cambridge CB10 1SA, UK; Kavli Centre for Ethics, Science and the Public, Faculty of Education, University of Cambridge, CB2 8PQ.
| |
Collapse
|
83
|
Fatumo S, Chikowore T, Choudhury A, Ayub M, Martin AR, Kuchenbaecker K. A roadmap to increase diversity in genomic studies. Nat Med 2022; 28:243-250. [PMID: 35145307 PMCID: PMC7614889 DOI: 10.1038/s41591-021-01672-4] [Citation(s) in RCA: 222] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022]
Abstract
Two decades ago, the sequence of the first human genome was published. Since then, advances in genome technologies have resulted in whole-genome sequencing and microarray-based genotyping of millions of human genomes. However, genetic and genomic studies are predominantly based on populations of European ancestry. As a result, the potential benefits of genomic research-including better understanding of disease etiology, early detection and diagnosis, rational drug design and improved clinical care-may elude the many underrepresented populations. Here, we describe factors that have contributed to the imbalance in representation of different populations and, leveraging our experiences in setting up genomic studies in diverse global populations, we propose a roadmap to enhancing inclusion and ensuring equal health benefits of genomics advances. Our Perspective highlights the importance of sincere, concerted global efforts toward genomic equity to ensure the benefits of genomic medicine are accessible to all.
Collapse
Affiliation(s)
- Segun Fatumo
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda.
- The Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| | - Tinashe Chikowore
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Muhammad Ayub
- Division of Psychiatry, University College London, London, UK
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Karoline Kuchenbaecker
- Division of Psychiatry, University College London, London, UK
- UCL Genetics Institute, University College London, London, UK
| |
Collapse
|
84
|
Fernandez PW. Prostate Cancer Genomics Research Disparities in Africa: Advancing Knowledge in Resource Constrained Settings. Cancer Control 2022; 29:10732748221095952. [PMID: 35475404 PMCID: PMC9087236 DOI: 10.1177/10732748221095952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/02/2022] [Accepted: 03/24/2022] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer disproportionately affects men of African descent and it is estimated that Africa will bear the highest disease burden in the next decade. Underlying genomic factors may contribute to prostate cancer disparities; however, it is unclear whether Africa has prioritised genomics research toward addressing these disparities. A Pubmed review was performed of publications spanning a 15-year period, with specific focus on prostate cancer genomics research that included samples from Africa and investigators in Africa. Data are presented on research publications from Africa relative to similar publications from different geographical regions, and more specifically, the extent of disparities and the contributions to prostate cancer knowledge as a result of genomics research that included African samples and African institutions. Limited publication output may reflect the infrastructure and funding challenges in Africa. Widespread cooperation should be fostered by sharing capacity and leveraging existing expertise to address the growing cancer burden facing the continent.
Collapse
|
85
|
Ethical, Legal, and Social Implications of Fetal Gene Therapy. Clin Obstet Gynecol 2021; 64:933-940. [PMID: 34668890 DOI: 10.1097/grf.0000000000000653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
As fetal gene therapies move from experimental animal models to human in utero phase I clinical trials, there is a need to consider the ethical, legal, and social implications. While fetal gene therapies are attracting more regulatory oversight than previous fetal interventions such as fetal surgery, old sociological questions should be applied to this new context. As health care pathways around fetal therapy are shaped by the ways in which a pregnant person and the fetus are constituted, and as risks and benefits are evaluated, we cannot afford to lose sight of long-term consequences, especially those pertaining to social inclusion.
Collapse
|
86
|
Gonzalez S, Strizich G, Isasi CR, Hua S, Comas B, Sofer T, Thyagarajan B, Perreira KM, Talavera GA, Daviglus ML, Nelson SC, Giachello AL, Schneiderman N, Kaplan RC. Consent for Use of Genetic Data among US Hispanics/Latinos: Results from the Hispanic Community Health Study/ Study of Latinos. Ethn Dis 2021; 31:547-558. [PMID: 34720558 DOI: 10.18865/ed.31.4.547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Inclusion of historically underrepresented populations in biomedical research is critical for large precision medicine research initiatives. Among 13,721 Hispanic Community Health Study/Study of Latinos (HCHS/SOL) enrollees, we used multivariable-adjusted prevalence ratios to describe characteristics associated with participants' willingness to consent to different levels of biospecimen and genetic data analysis and sharing. At baseline (2008-2011), HCHS/SOL participants almost universally consented to the use of biospecimens and genetic data by study investigators and their collaborators (97.6%; 95%CI: 97.1, 98.0). Fewer consented to biospecimen and genetic data sharing with investigators not affiliated with the HCHS/SOL research team (81%, 95%CI: 80, 82) or any data sharing with commercial/for-profit entities (75%, 95%CI: 74, 76). Those refusing to share their data beyond the study investigators group were more often females, Spanish language-speakers and non-US born individuals. As expected, participants who were retained and reconsented at the six-year follow up visit tended to embrace broader data sharing, although this varied by group. Over time, Puerto Ricans and Dominicans were more likely to convert to broader data sharing than individuals of a Mexican background. Our analysis suggests that acculturation and immigration status of specific Hispanic/Latino communities may influence decisions about participation in genomic research projects and biobanks.
Collapse
Affiliation(s)
- Sara Gonzalez
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Garrett Strizich
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Carmen R Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Simin Hua
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Betsy Comas
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Tamar Sofer
- Department of Biostatistics, Harvard Medical School. Brigham and Women's Hospital, Boston, MA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Krista M Perreira
- Department of Social Medicine, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Gregory A Talavera
- South Bay Latino Research Center, Department of Psychology, San Diego State University, San Diego, CA
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL
| | - Sarah C Nelson
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Aida L Giachello
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Neil Schneiderman
- Department of Psychology and Behavioral Medicine Research Center, University of Miami, Miami, FL
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
87
|
Bentley AR, Chen G, Doumatey AP, Shriner D, Meeks KAC, Gouveia MH, Ekoru K, Zhou J, Adeyemo A, Rotimi CN. GWAS in Africans identifies novel lipids loci and demonstrates heterogenous association within Africa. Hum Mol Genet 2021; 30:2205-2214. [PMID: 34196372 PMCID: PMC8561421 DOI: 10.1093/hmg/ddab174] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 01/11/2023] Open
Abstract
Serum lipids are biomarkers of cardiometabolic disease risk, and understanding genomic factors contributing to their distribution is of interest. Studies of lipids in Africans are rare, though it is expected that such studies could identify novel loci. We conducted a GWAS of 4317 Africans enrolled from Nigeria, Ghana and Kenya. We evaluated linear mixed models of high-density lipoprotein cholesterol (HDLC), low-density lipoprotein cholesterol (LDLC), total cholesterol (CHOL), triglycerides (TG) and TG/HDLC. Replication was attempted in 9542 African Americans (AA). In our main analysis, we identified 28 novel associations in Africans. Of the 18 of these that could be tested in AA, three associations replicated (GPNMB-TG, ENPP1-TG and SMARCA4-LDLC). Five additional novel loci were discovered upon meta-analysis with AA (rs138282551-TG, PGBD5-HDLC, CD80-TG/HDLC, SLC44A1-CHOL and TLL2-CHOL). Analyses considering only those with predominantly West African ancestry (Nigeria, Ghana and AA) yielded new insights: ORC5-LDLC and chr20:60973327-CHOL. Among our novel findings are some loci with known connections to lipids pathways. For instance, rs147706369 (TLL2) alters a regulatory motif for sterol regulatory element-binding proteins, a family of transcription factors that control the expression of a range of enzymes involved in cholesterol, fatty acid and TG synthesis, and rs115749422 (SMARCA4), an independent association near the known LDLR locus that is rare or absent in populations without African ancestry. These findings demonstrate the utility of conducting genomic analyses in Africans for discovering novel loci and provide some preliminary evidence for caution against treating 'African ancestry' as a monolithic category.
Collapse
Affiliation(s)
- Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Karlijn A C Meeks
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Mateus H Gouveia
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Kenneth Ekoru
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
88
|
Tobin MD, Izquierdo AG. Improving ethnic diversity in respiratory genomics research. Eur Respir J 2021; 58:58/4/2101615. [PMID: 34649971 DOI: 10.1183/13993003.01615-2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Martin D Tobin
- Dept of Health Sciences, University of Leicester, Leicester, UK .,Leicester NIHR Biomedical Research Centre, Leicester, UK
| | | |
Collapse
|
89
|
Mahjani B, Bey K, Boberg J, Burton C. Genetics of obsessive-compulsive disorder. Psychol Med 2021; 51:2247-2259. [PMID: 34030745 PMCID: PMC8477226 DOI: 10.1017/s0033291721001744] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Obsessive-compulsive disorder (OCD) is a psychiatric disorder with multiple symptom dimensions (e.g. contamination, symmetry). OCD clusters in families and decades of twin studies clearly demonstrate an important role for genetics in the etiology of the disorder. METHODS In this review, we summarize the genetic epidemiology and molecular genetic studies of OCD and obsessive-compulsive symptoms. RESULTS OCD is a heritable, polygenic disorder with contributions from both common and rare variants, including de novo deleterious variations. Multiple studies have provided reliable support for a large additive genetic contribution to liability to OCD, with discrete OCD symptom dimensions having both shared and unique genetic risks. Genome-wide association studies have not produced significant results yet, likely because of small sample sizes, but larger meta-analyses are forthcoming. Both twin and genome-wide studies show that OCD shares genetic risk with its comorbid conditions (e.g. Tourette syndrome and anorexia nervosa). CONCLUSIONS Despite significant efforts to uncover the genetic basis of OCD, the mechanistic understanding of how genetic and environmental risk factors interact and converge at the molecular level to result in OCD's heterogeneous phenotype is still mostly unknown. Future investigations should increase ancestral genetic diversity, explore age and/or sex differences in genetic risk for OCD and expand the study of pharmacogenetics, gene expression, gene × environment interactions and epigenetic mechanisms for OCD.
Collapse
Affiliation(s)
- Behrang Mahjani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Tics, Obsessive-Compulsive Disorder (OCD) and Related Disorders, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Katharina Bey
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Julia Boberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Christie Burton
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
90
|
Fite BZ, Hinostroza V, States L, Hicks-Nelson A, Baratto L, Kallianos K, Codari M, Yu B, Jha P, Shams M, Stoyanova T, Chapelin FF, Liu A, Rashidi A, Soto F, Quintana Y, Davidzon GA, Marycz K, Gibbs IC, Chonde DB, Patel CB, Daldrup-Link HE. Increasing Diversity in Radiology and Molecular Imaging: Current Challenges. Mol Imaging Biol 2021; 23:625-638. [PMID: 33903986 PMCID: PMC8074707 DOI: 10.1007/s11307-021-01610-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/05/2021] [Accepted: 04/19/2021] [Indexed: 11/28/2022]
Abstract
This paper summarizes the 2020 Diversity in Radiology and Molecular Imaging: What We Need to Know Conference, a three-day virtual conference held September 9-11, 2020. The World Molecular Imaging Society (WMIS) and Stanford University jointly organized this event to provide a forum for WMIS members and affiliates worldwide to openly discuss issues pertaining to diversity in science, technology, engineering, and mathematics (STEM). The participants discussed three main conference themes, "racial diversity in STEM," "women in STEM," and "global health," which were discussed through seven plenary lectures, twelve scientific presentations, and nine roundtable discussions, respectively. Breakout sessions were designed to flip the classroom and seek input from attendees on important topics such as increasing the representation of underrepresented minority (URM) members and women in STEM, generating pipeline programs in the fields of molecular imaging, supporting existing URM and women members in their career pursuits, developing mechanisms to effectively address microaggressions, providing leadership opportunities for URM and women STEM members, improving global health research, and developing strategies to advance culturally competent healthcare.
Collapse
Affiliation(s)
- Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | | | - Lisa States
- Department of Molecular Imaging, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | - Lucia Baratto
- Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Kimberly Kallianos
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Marina Codari
- Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Brenda Yu
- Department of Biophysics, Stanford University, Palo Alto, CA, 94305, USA
| | - Priyanka Jha
- Department of Radiology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Mana Shams
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Tanya Stoyanova
- Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Fanny F Chapelin
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY, 40506, USA
| | - Anna Liu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Ali Rashidi
- Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Fernando Soto
- Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Yuri Quintana
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | | | - Krzysztof Marycz
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Wrocław, Poland
| | - Iris C Gibbs
- Department of Radiation Oncology, Stanford University, Palo Alto, CA, 94305, USA
| | - Daniel B Chonde
- Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Chirag B Patel
- Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Heike Elisabeth Daldrup-Link
- Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA.
- Department of Pediatrics, Stanford University, Palo Alto, CA, 94305, USA.
| |
Collapse
|
91
|
Nagaraj SH, Toombs M. The Gene-Drug Duality: Exploring the Pharmacogenomics of Indigenous Populations. Front Genet 2021; 12:687116. [PMID: 34616423 PMCID: PMC8488351 DOI: 10.3389/fgene.2021.687116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022] Open
Abstract
While pharmacogenomic studies have facilitated the rapid expansion of personalized medicine, the benefits of these findings have not been evenly distributed. Genomic datasets pertaining to Indigenous populations are sorely lacking, leaving members of these communities at a higher risk of adverse drug reactions (ADRs), and associated negative outcomes. Australia has one of the largest Indigenous populations in the world. Pharmacogenomic studies of these diverse Indigenous Australian populations have been hampered by a paucity of data. In this article, we discuss the history of pharmacogenomics and highlight the inequalities that must be addressed to ensure equal access to pharmacogenomic-based healthcare. We also review efforts to conduct the pharmacogenomic profiling of chronic diseases among Australian Indigenous populations and survey the impact of the lack of drug safety-related information on potential ADRs among individuals in these communities.
Collapse
Affiliation(s)
- Shivashankar H. Nagaraj
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Maree Toombs
- School of Public Health, Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| |
Collapse
|
92
|
Bell J, Prictor M, Davenport L, O'Brien L, Wake M. Digital Mega-Studies as a New Research Paradigm: Governing the Health Research of the Future. J Empir Res Hum Res Ethics 2021; 16:344-355. [PMID: 34498950 DOI: 10.1177/15562646211041492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
'Digital Mega-Studies' are entirely or extensively digitised, longitudinal, population-scale initiatives, collecting, storing, and making available individual-level research data of different types and from multiple sources, shaped by technological developments and unforeseeable risks over time. The Australian 'Gen V' project exemplifies this new research paradigm. In 2019, we undertook a multidisciplinary, multi-stakeholder process to map Digital Mega-Studies' key characteristics, legal and governance challenges and likely solutions. We conducted large and small group processes within a one-day symposium and directed online synthesis and group prioritisation over subsequent weeks. We present our methods (including elicitation, affinity mapping and prioritisation processes) and findings, proposing six priority governance principles across three areas-data, participation, trust-to support future high-quality, large-scale digital research in health.
Collapse
Affiliation(s)
- Jessica Bell
- Melbourne Law School, 90147University of Melbourne, Carlton, Victoria, Australia.,School of Law, 2707University of Warwick, Coventry, West Midlands, UK
| | - Megan Prictor
- Melbourne Law School, 90147University of Melbourne, Carlton, Victoria, Australia
| | - Lauren Davenport
- 34361Murdoch Children's Research Institute, Parkville, Australia
| | - Lynda O'Brien
- 34361Murdoch Children's Research Institute, Parkville, Australia
| | - Melissa Wake
- 34361Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Liggins Institute, 1415University of Auckland, Auckland, New Zealand
| |
Collapse
|
93
|
Wiley AS. Pearl lecture: Biological normalcy: A new framework for biocultural analysis of human population variation. Am J Hum Biol 2021; 33:e23563. [PMID: 33458923 DOI: 10.1002/ajhb.23563] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 12/31/2022] Open
Abstract
Biological normalcy is a new analytical framework for understanding the bi-directional relationships between the biology of populations and cultural norms. Populations are characterized by statistical distributions-that is, measures of central tendency and variance-for biological traits, and these co-exist in societies with ideas about what constitutes "normal" human bodies, that is, normative views about what bodies "should" be like. While statistical norms may carry no explicit evaluative weight, the question is how they are related to judgments about what is "normal" or "abnormal." In a 1947 paper, Margaret Mead recognized their potential relationship: "normal…may refer to the statistically usual in the culture-usually without any recognition that this is culturally relative-so that the statistically usual is identified with the basically human…." Despite her observations over 70 years ago, little has been done on this topic, yet such work promises new insights into the relationship between culture and biology, here described at the population level, rather than as individual genetic characteristics. Using examples of sex/gender, race/ethnicity, age, and my work on human variation in the ability to drink milk, I outline the ways in which statistical norms may: influence individuals' perceptions of what is "normal" (Mead's "basically human"); lead to normative judgments about what human biology "should" be ("ethno-biocentrism") that are reinforced by biases in discourse about human variation; and potentially feedback to mold the biological characteristics of a population.
Collapse
Affiliation(s)
- Andrea S Wiley
- Department of Anthropology, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
94
|
Herd P, Mills MC, Dowd JB. Reconstructing Sociogenomics Research: Dismantling Biological Race and Genetic Essentialism Narratives. JOURNAL OF HEALTH AND SOCIAL BEHAVIOR 2021; 62:419-435. [PMID: 34100668 DOI: 10.1177/00221465211018682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
We detail the implications of sociogenomics for social determinants research. We focus on education and race because of how early twentieth-century scientific eugenic thinking facilitated a range of racist and eugenic policies, most of which helped justify and pattern racial and educational morbidity and mortality disparities that remain today, and are central to sociological research. Consequently, we detail the implications of sociogenomics research by unpacking key controversies and opportunities in sociogenomics as they pertain to the understanding of racial and educational inequalities. We clarify why race is not a valid biological or genetic construct, the ways that environments powerfully shape genetic influence, and risks linked to this field of research. We argue that sociologists can usefully engage in genetics research, a domain dominated by psychologists and behaviorists who, given their focus on individuals, have mostly not examined the role of history and social structure in shaping genetic influence.
Collapse
|
95
|
Bree KK, Hensley PJ, Pettaway CA. Germline Mutations in African American Men With Prostate Cancer: Incidence, Implications and Diagnostic Disparities. Urology 2021; 163:148-155. [PMID: 34453957 DOI: 10.1016/j.urology.2021.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/03/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022]
Abstract
Recent data suggests that African American men (AAM) with prostate cancer (PCa) exhibit genetic alterations in highly penetrant germline genes, as well as low penetrant single nucleotide polymorphisms. The importance of germline variants of uncertain significance (VUS) remain poorly elucidated and given the elevated rates of VUS in AAM compared to Caucasians with PCa, further studies are needed to facilitate potential reclassification of VUS. Ongoing efforts to include AAM in genomics research is of paramount importance in order to ensure applicability of discoveries across diverse populations and potentially reduce PCa disparities as we embark on the era of precision medicine.
Collapse
Affiliation(s)
- Kelly K Bree
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Patrick J Hensley
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Curtis A Pettaway
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
96
|
Slunecka JL, van der Zee MD, Beck JJ, Johnson BN, Finnicum CT, Pool R, Hottenga JJ, de Geus EJC, Ehli EA. Implementation and implications for polygenic risk scores in healthcare. Hum Genomics 2021; 15:46. [PMID: 34284826 PMCID: PMC8290135 DOI: 10.1186/s40246-021-00339-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022] Open
Abstract
Increasing amounts of genetic data have led to the development of polygenic risk scores (PRSs) for a variety of diseases. These scores, built from the summary statistics of genome-wide association studies (GWASs), are able to stratify individuals based on their genetic risk of developing various common diseases and could potentially be used to optimize the use of screening and preventative treatments and improve personalized care for patients. Many challenges are yet to be overcome, including PRS validation, healthcare professional and patient education, and healthcare systems integration. Ethical challenges are also present in how this information is used and the current lack of diverse populations with PRSs available. In this review, we discuss the topics above and cover the nature of PRSs, visualization schemes, and how PRSs can be improved. With these tools on the horizon for multiple diseases, scientists, clinicians, health systems, regulatory bodies, and the public should discuss the uses, benefits, and potential risks of PRSs.
Collapse
Affiliation(s)
- John L Slunecka
- Avera Institute for Human Genetics, Avera McKennan & University Health Center, Sioux Falls, SD, USA.
| | - Matthijs D van der Zee
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jeffrey J Beck
- Avera Institute for Human Genetics, Avera McKennan & University Health Center, Sioux Falls, SD, USA
| | - Brandon N Johnson
- Avera Institute for Human Genetics, Avera McKennan & University Health Center, Sioux Falls, SD, USA
| | - Casey T Finnicum
- Avera Institute for Human Genetics, Avera McKennan & University Health Center, Sioux Falls, SD, USA
| | - René Pool
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Erik A Ehli
- Avera Institute for Human Genetics, Avera McKennan & University Health Center, Sioux Falls, SD, USA
| |
Collapse
|
97
|
Olawoye O, Chuka-Okosa C, Akpa O, Realini T, Hauser M, Ashaye A. Eyes of Africa: The Genetics of Blindness: Study Design and Methodology. BMC Ophthalmol 2021; 21:272. [PMID: 34243759 PMCID: PMC8267233 DOI: 10.1186/s12886-021-02029-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/25/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This report describes the design and methodology of the "Eyes of Africa: The Genetics of Blindness," a collaborative study funded through the Human Heredity and Health in Africa (H3Africa) program of the National Institute of Health. METHODS This is a case control study that is collecting a large well phenotyped data set among glaucoma patients and controls for a genome wide association study. (GWAS). Multiplex families segregating Mendelian forms of early-onset glaucoma will also be collected for exome sequencing. DISCUSSION A total of 4500 cases/controls have been recruited into the study at the end of the 3rd funded year of the study. All these participants have been appropriately phenotyped and blood samples have been received from these participants. Recent GWAS of POAG in African individuals demonstrated genome-wide significant association with the APBB2 locus which is an association that is unique to individuals of African ancestry. This study will add to the existing knowledge and understanding of POAG in the African population.
Collapse
Affiliation(s)
- Olusola Olawoye
- Department of Ophthalmology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Chimdi Chuka-Okosa
- Department of Ophthalmology, College of Medicine, University of Nigeria, Enugu Campus, Enugu, Nigeria
| | - Onoja Akpa
- Department of Epidemiology and Medical Statistics, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Tony Realini
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, USA
| | - Michael Hauser
- Department of Medicine, Duke University, NC Durham, USA
- Department of Ophthalmology, Duke University, NC Durham, USA
| | - Adeyinka Ashaye
- Department of Ophthalmology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
98
|
Kalfakakou D, Fostira F, Papathanasiou A, Apostolou P, Dellatola V, Gavra IE, Vlachos IS, Scouras ZG, Drosopoulou E, Yannoukakos D, Konstantopoulou I. CanVaS: Documenting the genetic variation spectrum of Greek cancer patients. Hum Mutat 2021; 42:1081-1093. [PMID: 34174131 DOI: 10.1002/humu.24249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/13/2021] [Accepted: 06/14/2021] [Indexed: 11/08/2022]
Abstract
National genetic variation registries vastly increase the level of detail for the relevant population, while directly affecting patient management. Herein, we report CanVaS, a Cancer Variation reSource aiming to document the genetic variation of cancer patients in Greece. CanVaS comprises germline genetic data from 7,363 Greek individuals with a personal and/or family history of malignancy. The data set incorporates approximately 24,000 functionally annotated rare variants in 97 established or suspected cancer susceptibility genes. For each variant, allele frequency for the Greek population, interpretation for clinical significance, anonymized family and segregation information, as well as phenotypic traits of the carriers, are included. Moreover, information on the geographic distribution of the variants across the country is provided, enabling the study of Greek population isolates. Direct comparisons between Greek (sub)populations with relevant genetic resources are supported, allowing fine-grain localized adjustment of guidelines and clinical decision-making. Most importantly, anonymized data are available for download, while the Leiden Open Variation Database schema is adopted, enabling integration/interconnection with central resources. CanVaS could become a stepping-stone for a countrywide effort to characterize the cancer genetic variation landscape, concurrently supporting national and international cancer research. The database can be accessed at: http://ithaka.rrp.demokritos.gr/CanVaS.
Collapse
Affiliation(s)
- Despoina Kalfakakou
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Molecular Diagnostics Laboratory, Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Florentia Fostira
- Molecular Diagnostics Laboratory, Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Athanasios Papathanasiou
- Molecular Diagnostics Laboratory, Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Paraskevi Apostolou
- Molecular Diagnostics Laboratory, Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Vasiliki Dellatola
- Molecular Diagnostics Laboratory, Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Ioanna E Gavra
- Molecular Diagnostics Laboratory, Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Ioannis S Vlachos
- Department of Pathology, Cancer Research Institute, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Zacharias G Scouras
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Drosopoulou
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Drakoulis Yannoukakos
- Molecular Diagnostics Laboratory, Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Irene Konstantopoulou
- Molecular Diagnostics Laboratory, Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, National Center for Scientific Research "Demokritos", Athens, Greece
| |
Collapse
|
99
|
Gosztyla ML, Kwong L, Murray NA, Williams CE, Behnke N, Curry P, Corbett KD, DSouza KN, Gala de Pablo J, Gicobi J, Javidnia M, Lotay N, Prescott SM, Quinn JP, Rivera ZMG, Smith MA, Tang KTY, Venkat A, Yamoah MA. Responses to 10 common criticisms of anti-racism action in STEMM. PLoS Comput Biol 2021; 17:e1009141. [PMID: 34264941 PMCID: PMC8282043 DOI: 10.1371/journal.pcbi.1009141] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Maya L. Gosztyla
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Lydia Kwong
- Bioethics and Science Policy Program, Duke University, Durham, North Carolina, United States of America
| | - Naomi A. Murray
- Ecology, Evolution, and Biodiversity Program, University of California Davis, Davis, California, United States of America
| | - Claire E. Williams
- Department of Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Nicholas Behnke
- Department of Food, Agricultural, and Biological Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Porsia Curry
- Porsia Curry, Black Resource Center, University of California San Diego, La Jolla, California, United States of America
| | - Kevin D. Corbett
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Karen N. DSouza
- Mayo Clinic Graduate School of Biomedical Science, Rochester, Minnesota, United States of America
| | | | - Joanina Gicobi
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Monica Javidnia
- Department of Neurology, University of Rochester, Rochester, New York, United States of America
| | - Navina Lotay
- Department of Chemistry, University of Toronto, Toronto, Canada
| | - Sidney Madison Prescott
- Executive Women’s MBA Cohort, Women’s College, Brenau University, Gainesville, Georgia, United States of America
- Department of Graduate Studies, Master of Science in Legal Studies Program, Cornell Law School, Ithaca, New York, United States of America
| | - James P. Quinn
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Zeena M. G. Rivera
- Neurosciences Interdepartmental Program, University of California Los Angeles, Los Angeles, California, United States of America
| | - Markia A. Smith
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Karen T. Y. Tang
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Canada
| | - Aarya Venkat
- Department of Biochemistry, University of Georgia, Athens, Georgia, United States of America
| | - Megan A. Yamoah
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| |
Collapse
|
100
|
Friedrich VK, Rubel MA, Schurr TG. Mitochondrial genetic variation in human bioenergetics, adaptation, and adult disease. Am J Hum Biol 2021; 34:e23629. [PMID: 34146380 DOI: 10.1002/ajhb.23629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Mitochondria are critical for the survival of eukaryotic organisms due to their ability to produce cellular energy, which drives virtually all aspects of host biology. However, the effects of mitochondrial DNA (mtDNA) variation in relation to disease etiology and adaptation within contemporary global human populations remains incompletely understood. METHODS To develop a more holistic understanding of the role of mtDNA diversity in human adaptation, health, and disease, we investigated mitochondrial biology and bioenergetics. More specifically, we synthesized details from studies of mitochondrial function and variation in the context of haplogroup background, climatic adaptation, and oxidative disease. RESULTS The majority of studies show that mtDNA variation arose during modern human dispersal around the world. Some of these variants appear to have been positively selected for their adaptiveness in colder climates, with these sequence changes having implications for tissue-specific function and thermogenic capacity. In addition, many variants modulating energy production are also associated with damaging metabolic byproducts and mitochondrial dysfunction, which, in turn, are implicated in the onset and severity of several different adult mitochondrial diseases. Thus, mtDNA variation that governs bioenergetics, metabolism, and thermoregulation may potentially have adverse consequences for human health, depending on the genetic background and context in which it occurs. CONCLUSIONS Our review suggests that the mitochondrial research field would benefit from independently replicating mtDNA haplogroup-phenotype associations across global populations, incorporating potentially confounding environmental, demographic, and disease covariates into studies of mtDNA variation, and extending association-based studies to include analyses of complete mitogenomes and assays of mitochondrial function.
Collapse
Affiliation(s)
- Volney K Friedrich
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meagan A Rubel
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Translational Imaging and Precision Medicine, University of California - San Diego, La Jolla, California, USA
| | - Theodore G Schurr
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|