1
|
Singh P, Guin D, Pattnaik B, Kukreti R. Mapping the genetic architecture of idiopathic pulmonary fibrosis: Meta-analysis and epidemiological evidence of case-control studies. Gene 2024; 895:147993. [PMID: 37977320 DOI: 10.1016/j.gene.2023.147993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/23/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a rare and devastating fibrotic lung disorder with unknown etiology. Although it is believed that genetic component is an important risk factor for IPF, a comprehensive understanding of its genetic landscape is lacking. Hence, we aimed to highlight the susceptibility genes and pathways implicated in IPF pathogenesis through a two-staged systematic literature search of genetic association studies on IPF, followed by meta-analysis and pathway enrichment analysis. METHODS This study was performed based on PRISMA guidelines (PROSPERO, registration number: CRD42022297970). The first search was performed (using PubMed and Web of Science) retrieving a total of 5642 articles, of which 52 were eligible for inclusion in the first stage. The second search was performed (using PubMed, Web of Science and Scopus) for ten polymorphisms, identified from the first search, with 2 or more studies. Finally, seven polymorphisms, [rs35705950/MUC5B, rs2736100/TERT, rs2609255/FAM13A, rs2076295/DSP, rs12610495/DPP9, rs111521887/TOLLIP and rs1800470/TGF-β1] qualified for meta-analyses. The epidemiological credibility was evaluated using Venice criteria. RESULTS From the systematic review, 222 polymorphisms in 118 genes showed a significant association with IPF susceptibility. Meta-analyses findings revealed significant association of rs35705950/T [OR = 3.92(3.26-4.57)], rs2609255/G [OR = 1.50(1.18-1.82)], rs2076295/G [OR = 1.19(0.82-1.756)], rs12610495/G [OR = 1.28(1.12-1.44)], rs2736100/C [OR = 0.68(0.54-0.82), rs111521887/G [OR = 1.34(1.06-1.61)] and suggestive evidence for rs1800470/T [OR = 1.08(0.82-1.34)] with IPF susceptibility. Four polymorphisms- rs35705950/MUC5B, rs2736100/TERT, rs2076295/DSP and rs111521887/TOLLIP, exhibited substantial epidemiological evidence supporting their association with IPF risk. Gene ontology and pathway enrichment analysis performed on IPF risk-associated genes identified a critical role of genes in mucin production, immune response and inflammation, host defence, cell-cell adhesion and telomere maintenance. CONCLUSIONS Our findings present the most prominent IPF-associated genetic risk variants involved in alveolar epithelial injuries (MUC5B, TERT, FAM13A, DSP, DPP9) and epithelial-mesenchymal transition (TOLLIP, TGF-β1), providing genetic and biological insights into IPF pathogenesis. However, further experimental research and human studies with larger sample sizes, diverse ethnic representation, and rigorous design are warranted.
Collapse
Affiliation(s)
- Pooja Singh
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC, Ghaziabad, Uttar Pradesh, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Debleena Guin
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, New Delhi, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Bijay Pattnaik
- Centre of Excellence for Translational Research in Asthma and Lung Diseases, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Ritushree Kukreti
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC, Ghaziabad, Uttar Pradesh, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India.
| |
Collapse
|
2
|
Hoorn QA, Zayas GA, Rodriguez EE, Jensen LM, Mateescu RG, Hansen PJ. Identification of quantitative trait loci and associated candidate genes for pregnancy success in Angus-Brahman crossbred heifers. J Anim Sci Biotechnol 2023; 14:137. [PMID: 37932831 PMCID: PMC10629031 DOI: 10.1186/s40104-023-00940-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/10/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND In beef cattle, more than 50% of the energy input to produce a unit of beef is consumed by the female that produced the calf. Development of genomic tools to identify females with high genetic merit for reproductive function could increase the profitability and sustainability of beef production. RESULTS Genome-wide association studies (GWAS) were performed using a single-step genomic best linear unbiased prediction approach on pregnancy outcome traits from a population of Angus-Brahman crossbred heifers. Furthermore, a validation GWAS was performed using data from another farm. Heifers were genotyped with the Bovine GGP F250 array that contains 221,077 SNPs. In the discovery population, heifers were bred in winter breeding seasons involving a single round of timed artificial insemination (AI) followed by natural mating for 3 months. Two phenotypes were analyzed: pregnancy outcome to first-service AI (PAI; n = 1,481) and pregnancy status at the end of the breeding season (PEBS; n = 1,725). The heritability was estimated as 0.149 and 0.122 for PAI and PEBS, respectively. In the PAI model, one quantitative trait locus (QTL), located between 52.3 and 52.5 Mb on BTA7, explained about 3% of the genetic variation, in a region containing a cluster of γ-protocadherin genes and SLC25A2. Other QTLs explaining between 0.5% and 1% of the genetic variation were found on BTA12 and 25. In the PEBS model, a large QTL on BTA7 was synonymous with the QTL for PAI, with minor QTLs located on BTA5, 9, 10, 11, 19, and 20. The validation population for pregnancy status at the end of the breeding season were Angus-Brahman crossbred heifers bred by natural mating. In concordance with the discovery population, the large QTL on BTA7 and QTLs on BTA10 and 12 were identified. CONCLUSIONS In summary, QTLs and candidate SNPs identified were associated with pregnancy outcomes in beef heifers, including a large QTL associated with a group of protocadherin genes. Confirmation of these associations with larger populations could lead to the development of genomic predictions of reproductive function in beef cattle. Moreover, additional research is warranted to study the function of candidate genes associated with QTLs.
Collapse
Affiliation(s)
- Quinn A Hoorn
- Department of Animal Sciences, Donald Henry Barron Reproductive and Perinatal Biology Research Program, and the Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Gabriel A Zayas
- Department of Animal Sciences, Donald Henry Barron Reproductive and Perinatal Biology Research Program, and the Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Eduardo E Rodriguez
- Department of Animal Sciences, Donald Henry Barron Reproductive and Perinatal Biology Research Program, and the Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Laura M Jensen
- Present address: School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia
| | - Raluca G Mateescu
- Department of Animal Sciences, Donald Henry Barron Reproductive and Perinatal Biology Research Program, and the Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Peter J Hansen
- Department of Animal Sciences, Donald Henry Barron Reproductive and Perinatal Biology Research Program, and the Genetics Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
3
|
Thibault RT, Amaral OB, Argolo F, Bandrowski AE, Davidson AR, Drude NI. Open Science 2.0: Towards a truly collaborative research ecosystem. PLoS Biol 2023; 21:e3002362. [PMID: 37856538 PMCID: PMC10617723 DOI: 10.1371/journal.pbio.3002362] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/31/2023] [Indexed: 10/21/2023] Open
Abstract
Conversations about open science have reached the mainstream, yet many open science practices such as data sharing remain uncommon. Our efforts towards openness therefore need to increase in scale and aim for a more ambitious target. We need an ecosystem not only where research outputs are openly shared but also in which transparency permeates the research process from the start and lends itself to more rigorous and collaborative research. To support this vision, this Essay provides an overview of a selection of open science initiatives from the past 2 decades, focusing on methods transparency, scholarly communication, team science, and research culture, and speculates about what the future of open science could look like. It then draws on these examples to provide recommendations for how funders, institutions, journals, regulators, and other stakeholders can create an environment that is ripe for improvement.
Collapse
Affiliation(s)
- Robert T. Thibault
- 1 Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, Unites States of America
| | - Olavo B. Amaral
- Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Anita E. Bandrowski
- FAIR Data Informatics Lab, Department of Neuroscience, UCSD, San Diego, California, United States of America
- SciCrunch Inc., San Diego, California, United States of America
| | - Alexandra R, Davidson
- Institute for Evidence-Based Health Care, Bond University, Robina, Australia
- Faculty of Health Science and Medicine, Bond University, Robina, Australia
| | - Natascha I. Drude
- Berlin Institute of Health (BIH) at Charité, BIH QUEST Center for Responsible Research, Berlin, Germany
| |
Collapse
|
4
|
Axfors C, Patel CJ, Ioannidis JPA. Published registry-based pharmacoepidemiologic associations show limited concordance with agnostic medication-wide analyses. J Clin Epidemiol 2023; 160:33-45. [PMID: 37224981 DOI: 10.1016/j.jclinepi.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/13/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023]
Abstract
OBJECTIVES To assess how the results of published national registry-based pharmacoepidemiology studies (where select associations are of interest) compare with an agnostic medication-wide approach (where all possible drug associations are tested). STUDY DESIGN AND SETTING We systematically searched for publications that reported drug associations with any, breast, colon/colorectal, or prostate cancer in the Swedish Prescribed Drug Registry. Results were compared against a previously performed agnostic medication-wide study on the same registry. PROTOCOL https://osf.io/kqj8n. RESULTS Most published studies (25/32) investigated previously reported associations. 421/913 (46%) associations had statistically significant results. 134 of the 162 unique drug-cancer associations could be paired with 70 associations in the agnostic study (corresponding drug categories and cancer types). Published studies reported smaller effect sizes and absolute effect sizes than the agnostic study, and generally used more adjustments. Agnostic analyses were less likely to report statistically significant protective associations (based on a multiplicity-corrected threshold) than their paired associations in published studies (McNemar odds ratio 0.13, P = 0.0022). Among 162 published associations, 36 (22%) showed increased risk signal and 25 (15%) protective signal at P < 0.05, while for agnostic associations, 237 (11%) showed increased risk signal and 108 (5%) protective signal at a multiplicity-corrected threshold. Associations belonging to drug categories targeted by individual published studies vs. nontargeted had smaller average effect sizes; smaller P values; and more frequent risk signals. CONCLUSION Published pharmacoepidemiology studies using a national registry addressed mostly previously proposed associations, were mostly "negative", and showed only modest concordance with their respective agnostic analyses in the same registry.
Collapse
Affiliation(s)
- Cathrine Axfors
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA; Department for Women's and Children's Health, Uppsala University, Uppsala, Sweden.
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - John P A Ioannidis
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA; Departments of Medicine, of Epidemiology and Population Health, of Biomedical Data Science, and of Statistics, Stanford University, Stanford, CA, USA
| |
Collapse
|
5
|
Jurrjens AW, Seldin MM, Giles C, Meikle PJ, Drew BG, Calkin AC. The potential of integrating human and mouse discovery platforms to advance our understanding of cardiometabolic diseases. eLife 2023; 12:e86139. [PMID: 37000167 PMCID: PMC10065800 DOI: 10.7554/elife.86139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/15/2023] [Indexed: 04/01/2023] Open
Abstract
Cardiometabolic diseases encompass a range of interrelated conditions that arise from underlying metabolic perturbations precipitated by genetic, environmental, and lifestyle factors. While obesity, dyslipidaemia, smoking, and insulin resistance are major risk factors for cardiometabolic diseases, individuals still present in the absence of such traditional risk factors, making it difficult to determine those at greatest risk of disease. Thus, it is crucial to elucidate the genetic, environmental, and molecular underpinnings to better understand, diagnose, and treat cardiometabolic diseases. Much of this information can be garnered using systems genetics, which takes population-based approaches to investigate how genetic variance contributes to complex traits. Despite the important advances made by human genome-wide association studies (GWAS) in this space, corroboration of these findings has been hampered by limitations including the inability to control environmental influence, limited access to pertinent metabolic tissues, and often, poor classification of diseases or phenotypes. A complementary approach to human GWAS is the utilisation of model systems such as genetically diverse mouse panels to study natural genetic and phenotypic variation in a controlled environment. Here, we review mouse genetic reference panels and the opportunities they provide for the study of cardiometabolic diseases and related traits. We discuss how the post-GWAS era has prompted a shift in focus from discovery of novel genetic variants to understanding gene function. Finally, we highlight key advantages and challenges of integrating complementary genetic and multi-omics data from human and mouse populations to advance biological discovery.
Collapse
Affiliation(s)
- Aaron W Jurrjens
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, United States
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| |
Collapse
|
6
|
Friedman JM, van Essen P, van Karnebeek CDM. Cerebral palsy and related neuromotor disorders: Overview of genetic and genomic studies. Mol Genet Metab 2022; 137:399-419. [PMID: 34872807 DOI: 10.1016/j.ymgme.2021.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022]
Abstract
Cerebral palsy (CP) is a debilitating condition characterized by abnormal movement or posture, beginning early in development. Early family and twin studies and more recent genomic investigations clearly demonstrate that genetic factors of major effect contribute to the etiology of CP. Most copy number variants and small alterations of nucleotide sequence that cause CP arise as a result of de novo mutations, so studies that estimate heritability on basis of recurrence frequency within families substantially underestimate genetic contributions to the etiology. At least 4% of patients with typical CP have disease-causing CNVs, and at least 14% have disease-causing single nucleotide variants or indels. The rate of pathogenic genomic lesions is probably more than twice as high among patients who have atypical CP, i.e., neuromotor dysfunction with additional neurodevelopmental abnormalities or malformations, or with MRI findings and medical history that are not characteristic of a perinatal insult. Mutations of many different genetic loci can produce a CP-like phenotype. The importance of genetic variants of minor effect and of epigenetic modifications in producing a multifactorial predisposition to CP is less clear. Recognizing the specific cause of CP in an affected individual is essential to providing optimal clinical management. An etiological diagnosis provides families an "enhanced compass" that improves overall well-being, facilitates access to educational and social services, permits accurate genetic counseling, and, for a subset of patients such as those with underlying inherited metabolic disorders, may make precision therapy that targets the pathophysiology available. Trio exome sequencing with assessment of copy number or trio genome sequencing with bioinformatics analysis for single nucleotide variants, indels, and copy number variants is clinically indicated in the initial workup of CP patients, especially those with additional malformations or neurodevelopmental abnormalities.
Collapse
Affiliation(s)
- Jan M Friedman
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Peter van Essen
- Department of Pediatrics, Amalia Children's Hospital, Radboud Centre for Mitochondrial Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clara D M van Karnebeek
- Department of Pediatrics, Amalia Children's Hospital, Radboud Centre for Mitochondrial Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Departments of Human Genetics and Pediatrics, Emma Children's Hospital, Amsterdam University Medical Centres, Amsterdam, the Netherlands; Department of Pediatrics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
7
|
Ishigaki K. Beyond GWAS: from simple associations to functional insights. Semin Immunopathol 2021; 44:3-14. [PMID: 34605948 DOI: 10.1007/s00281-021-00894-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022]
Abstract
Each human, when born, has slightly different DNA sequences, which make each of us unique. The variations in DNA sequences are called genetic variants. The primary aim of genome-wide association study (GWAS) is to detect associations between genetic variants and human phenotypes. Since GWAS focuses on germ-line variants, there is no reverse causation. Therefore, GWAS is one of the few tools that can assess the causality of human diseases. In the past 10 years, many large-scale GWAS have been conducted. Although the primary outputs of GWAS are just a series of statistics, its downstream analyses provided many insights beyond simple associations: the causal mechanisms for autoimmune diseases and shared etiology between diseases. Moreover, GWAS downstream analyses generated scores potentially helpful in predicting clinical outcomes of each patient. This review focuses on GWAS for autoimmune diseases and introduces significant achievements of its downstream analyses. We also provide future directions that potentially overcome current limitations. We restrict our discussion to common autoimmune diseases (e.g., rheumatoid arthritis) since rare Mendelian diseases possess distinct genetic etiologies and are not tested by GWAS.
Collapse
Affiliation(s)
- Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan.
| |
Collapse
|
8
|
Kim SK, Nguyen C, Avins AL, Abrams GD. Identification of Three Loci Associated with Achilles Tendon Injury Risk from a Genome-wide Association Study. Med Sci Sports Exerc 2021; 53:1748-1755. [PMID: 33606446 PMCID: PMC8282631 DOI: 10.1249/mss.0000000000002622] [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: 11/21/2022]
Abstract
PURPOSE This study aimed to screen the entire genome for genetic markers associated with risk for Achilles tendon injury. METHODS A genome-wide association analysis was performed using data from the Kaiser Permanente Research Board and the UK Biobank. Achilles tendon injury cases were identified based on electronic health records from the Kaiser Permanente Research Board databank and the UK Biobank from individuals of European ancestry. Genome-wide association analyses from both cohorts were tested for Achilles tendon injury using a logistic regression model adjusting for sex, height, weight, and race/ethnicity using allele counts for single nucleotide polymorphisms (SNP). Previously identified genes within the literature were also tested for association with Achilles tendon injury. RESULTS There were a total of 12,354 cases of Achilles tendon injury and 483,080 controls within the two combined cohorts, with 67 SNP in three chromosomal loci demonstrating a genome-wide significant association with Achilles tendon injury. The first locus contains a single SNP (rs183364169) near the CDCP1 and TMEM158 genes on chromosome 3. The second locus contains 65 SNP in three independently segregating sets near the MPP7 gene on chromosome 10. The last locus contains a single SNP (rs4454832) near the SOX21 and GPR180 genes on chromosome 13. The current data were used to test 14 candidate genes previously reported to show an association with Achilles tendon injury, but none showed a significant association (all P > 0.05). CONCLUSION Three loci were identified as potential risk factors for Achilles tendon injury and deserve further validation and investigation of molecular mechanisms.
Collapse
Affiliation(s)
- Stuart K. Kim
- Department of Developmental Biology, Stanford University Medical School, Stanford, CA
| | - Condor Nguyen
- Department of Developmental Biology, Stanford University Medical School, Stanford, CA
| | - Andy L. Avins
- Kaiser Permanente Northern California, Division of Research, Oakland, CA
| | - Geoffrey D. Abrams
- Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA
| |
Collapse
|
9
|
Ikle JM, Gloyn AL. 100 YEARS OF INSULIN: A brief history of diabetes genetics: insights for pancreatic beta-cell development and function. J Endocrinol 2021; 250:R23-R35. [PMID: 34196608 PMCID: PMC9037733 DOI: 10.1530/joe-21-0067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/30/2021] [Indexed: 12/30/2022]
Abstract
Since the discovery of insulin 100 years ago, our knowledge and understanding of diabetes have grown exponentially. Specifically, with regards to the genetics underlying diabetes risk, our discoveries have paralleled developments in our understanding of the human genome and our ability to study genomics at scale; these advancements in genetics have both accompanied and led to those in diabetes treatment. This review will explore the timeline and history of gene discovery and how this has coincided with progress in the fields of genomics. Examples of genetic causes of monogenic diabetes are presented and the continuing expansion of allelic series in these genes and the challenges these now cause for diagnostic interpretation along with opportunities for patient stratification are discussed.
Collapse
Affiliation(s)
- Jennifer M Ikle
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Anna L Gloyn
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| |
Collapse
|
10
|
Kim SK, Nguyen C, Avins AL, Abrams GD. Three genes associated with anterior and posterior cruciate ligament injury : a genome-wide association analysis. Bone Jt Open 2021; 2:414-421. [PMID: 34169730 PMCID: PMC8244791 DOI: 10.1302/2633-1462.26.bjo-2021-0040.r1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIMS The aim of this study was to screen the entire genome for genetic markers associated with risk for anterior cruciate ligament (ACL) and posterior cruciate ligament (PCL) injury. METHODS Genome-wide association (GWA) analyses were performed using data from the Kaiser Permanente Research Board (KPRB) and the UK Biobank. ACL and PCL injury cases were identified based on electronic health records from KPRB and the UK Biobank. GWA analyses from both cohorts were tested for ACL and PCL injury using a logistic regression model adjusting for sex, height, weight, age at enrolment, and race/ethnicity using allele counts for single nucleotide polymorphisms (SNPs). The data from the two GWA studies were combined in a meta-analysis. Candidate genes previously reported to show an association with ACL injury in athletes were also tested for association from the meta-analysis data from the KPRB and the UK Biobank GWA studies. RESULTS There was a total of 2,214 cases of ACL and PCL injury and 519,869 controls within the two cohorts, with three loci demonstrating a genome-wide significant association in the meta-analysis: INHBA, AEBP2, and LOC101927869. Of the eight candidate genes previously studied in the literature, six were present in the current dataset, and only COL3A1 (rs1800255) showed a significant association (p = 0.006). CONCLUSION Genetic markers in three novel loci in this study and one previously-studied candidate gene were identified as potential risk factors for ACL and PCL injury and deserve further validation and investigation of molecular mechanisms. Cite this article: Bone Jt Open 2021;2(6):414-421.
Collapse
Affiliation(s)
- Stuart K Kim
- Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Condor Nguyen
- Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Andrew L Avins
- Kaiser Permanente Northern California, Division of Research, Oakland, California, USA
| | - Geoffrey D Abrams
- Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
11
|
Kim SK, Roche MD, Fredericson M, Dragoo JL, Horton BH, Avins AL, Belanger HG, Ioannidis JPA, Abrams GD. A Genome-wide Association Study for Concussion Risk. Med Sci Sports Exerc 2021; 53:704-711. [PMID: 33017352 DOI: 10.1249/mss.0000000000002529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE This study aimed to screen the entire genome for genetic markers associated with risk for concussion. METHODS A genome-wide association analyses was performed using data from the Kaiser Permanente Research Bank and the UK Biobank. Concussion cases were identified based on electronic health records from the Kaiser Permanente Research Bank and the UK Biobank from individuals of European ancestry. Genome-wide association analyses from both cohorts were tested for concussion using a logistic regression model adjusting for sex, height, weight, and race/ethnicity using allele counts for single nucleotide polymorphisms. Previously identified genes within the literature were also tested for association with concussion. RESULTS There were a total of 4064 cases of concussion and 291,472 controls within the databases, with two single nucleotide polymorphisms demonstrating a genome-wide significant association with concussion. The first polymorphism, rs144663795 (P = 9.7 × 10-11; OR = 2.91 per allele copy), is located within the intron of SPATA5. Strong, deleterious mutations in SPATA5 cause intellectual disability, hearing loss, and vision loss. The second polymorphism, rs117985931 (P = 3.97 × 10-9; OR = 3.59 per allele copy), is located within PLXNA4. PLXNA4 plays a key role is axon outgrowth during neural development, and DNA variants in PLXNA4 are associated with risk for Alzheimer's disease. Previous investigations have identified five candidate genes that may be associated with concussion, but none showed a significant association in the current model (P < 0.05). CONCLUSION Two genetic markers were identified as potential risk factors for concussion and deserve further validation and investigation of molecular mechanisms.
Collapse
Affiliation(s)
- Stuart K Kim
- Department of Developmental Biology, Stanford University Medical School, Stanford, CA
| | - Megan D Roche
- Department Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA
| | - Michael Fredericson
- Department Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA
| | - Jason L Dragoo
- UCHealth Steadman Hawkins Clinic Denver-Surgery Center, Englewood, CO
| | - Brandon H Horton
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Andy L Avins
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | | | | | - Geoffrey D Abrams
- Department Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA
| |
Collapse
|
12
|
Libowitz MR, Nurmi EL. The Burden of Antipsychotic-Induced Weight Gain and Metabolic Syndrome in Children. Front Psychiatry 2021; 12:623681. [PMID: 33776816 PMCID: PMC7994286 DOI: 10.3389/fpsyt.2021.623681] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Antipsychotic medications are critical to child and adolescent psychiatry, from the stabilization of psychotic disorders like schizophrenia, bipolar disorder, and psychotic depression to behavioral treatment of autism spectrum disorder, tic disorders, and pediatric aggression. While effective, these medications carry serious risk of adverse events-most commonly, weight gain and cardiometabolic abnormalities. Negative metabolic consequences affect up to 60% of patients and present a major obstacle to long-term treatment. Since antipsychotics are often chronically prescribed beginning in childhood, cardiometabolic risk accumulates. An increased susceptibility to antipsychotic-induced weight gain (AIWG) has been repeatedly documented in children, particularly rapid weight gain. Associated cardiometabolic abnormalities include central obesity, insulin resistance, dyslipidemia, and systemic inflammation. Lifestyle interventions and medications such as metformin have been proposed to reduce risk but remain limited in efficacy. Furthermore, antipsychotic medications touted to be weight-neutral in adults can cause substantial weight gain in children. A better understanding of the biological underpinnings of AIWG could inform targeted and potentially more fruitful treatments; however, little is known about the underlying mechanism. As yet, modest genetic studies have nominated a few risk genes that explain only a small percentage of the risk. Recent investigations have begun to explore novel potential mechanisms of AIWG, including a role for gut microbiota and microbial metabolites. This article reviews the problem of AIWG and AP metabolic side effects in pediatric populations, proposed mechanisms underlying this serious side effect, and strategies to mitigate adverse impact. We suggest future directions for research efforts that may advance the field and lead to improved clinical interventions.
Collapse
Affiliation(s)
| | - Erika L. Nurmi
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Two GWAS-identified variants are associated with lumbar spinal stenosis and Gasdermin-C expression in Chinese population. Sci Rep 2020; 10:21069. [PMID: 33273635 PMCID: PMC7713291 DOI: 10.1038/s41598-020-78249-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
The aim of this study is to investigate the expression levels of genome-wide association studies (GWAS)-identified variants near Gasdermin-C (GSDMC) and its association with lumbar disc degeneration (LDD) in a Chinese population. In accordance with previously reported findings, our study involved the top 4 variants; rs6651255, rs7833174, rs4130415, and rs7816342. A total of 800 participants, 400 LDD patients and 400 controls were involved in the study. The LDD patients were divided into two mutually exclusive subgroups: subgroup 1: lumbar disc herniation; subgroup 2: lumbar spinal stenosis. Genotyping were performed using TaqMan assay, and Enzyme-Linked Immunosorbent Assay (ELISA) used to measure the plasma GSDMC levels, while quantitative reverse-transcription (qRT)-PCR and immunohistochemistry (IHC) were used to evaluate the GSDMC expression levels. Among the studied variants, there were no statistically significant differences in allelic and genotypic frequencies between LDD patients and their controls (all P > 0.05). However, the subgroup analysis revealed a significant association between rs6651255 and rs7833174 in patients with lumbar spinal stenosis (subgroup 2). Furthermore, the max-statistic test revealed that the inheritance models of two variants of lumbar spinal stenosis were represented by the recessive model. The plasma and mRNA expression levels of GSDMC were significantly higher in patients with lumbar spinal stenosis compared with the control group (P < 0.05). Furthermore, the CC genotypes of rs6651255 and rs7833174 were significantly associated with increased plasma expression levels of GSDMC in patients with lumbar spinal stenosis (P < 0.01). Two GWAS-identified variants (rs6651255 and rs7833174) near GSDMC were associated with a predisposition to lumbar spinal stenosis. GSDMC protein and mRNA expression levels may have prognostic qualities as biomarkers for the existence, occurrence or development of lumbar spinal stenosis.
Collapse
|
14
|
Goodman M, Li J, Flanders WD, Mahood D, Anthony LG, Zhang Q, LaKind JS. Epidemiology of PCBs and neurodevelopment: Systematic assessment of multiplicity and completeness of reporting. GLOBAL EPIDEMIOLOGY 2020. [DOI: 10.1016/j.gloepi.2020.100040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
15
|
Association of the soluble CTLA4 with schizophrenia: an observational study. JOURNAL OF BIO-X RESEARCH 2020. [DOI: 10.1097/jbr.0000000000000070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
16
|
Jatho A, Tran BT, Cambia JM, Nanyingi M, Mugisha NM. Cancer Risk Studies and Priority Areas for Cancer Risk Appraisal in Uganda. Ann Glob Health 2020; 86:78. [PMID: 32704483 PMCID: PMC7350938 DOI: 10.5334/aogh.2873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Research into aetiologies and prevention of the commonest cancers and implementation of primary and secondary prevention can reduce cancer risk and improve quality of life. Moreover, monitoring the prevalence of cancer risk factors in a specific population helps guide cancer prevention and early detection efforts and national cancer control programming. Objective This article aims to provide the scope and findings of cancer risk studies conducted in Uganda to guide researchers, health-care professionals, and policymakers. Methods Between November 2019 to January 2020, we searched peer-reviewed published articles in Pubmed, EMBASE and Cochrane Library (Cochrane central register of controlled trials-CENTRAL). We followed the recommendation of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses - the PRISMA. The primary focus was to identify cancer risk and prevention studies conducted in Uganda and published in peer-reviewed journals from January 2000 and January 2020. We used key Boolean search terms with their associated database strings. Results We identified 416 articles, screened 269 non-duplicate articles and obtained 77 full-text articles for review. Out of the 77 studies, we identified one (1%) randomized trial, two (2.5%) retrospective cohort studies and 14 (18%) case-control studies, 46 (60%) cross-sectional studies, five (6.4%) ecological studies, three panel studies (4%) and six (8%) qualitative studies. Cervical cancer was the most studied type of cancer in Uganda (23.4%, n = 18 studies), followed by lymphomas - both Hodgkin and Non-Hodgkin sub-types (20.7%), n = 16 studies) and breast cancer (15.6%, n = 12 studies). In lymphoma studies, Burkitt lymphoma was the most studied type of lymphoma (76%, n = 13 studies). The studies concentrated on specific cancer risk awareness, risk perceptions, attitudes, uptake of screening, uptake of human papillomavirus vaccination, the prevalence of some of the known cancer risk factors and obstacles to accessing screening services. Conclusion The unmet need for comprehensive cancer risk and prevention studies is enormous in Uganda. Future studies need to comprehensively investigate the known and putative cancer risk factors and prioritize the application of the higher-hierarchy evidence-generating epidemiological studies to guide planning of the national cancer control program.
Collapse
Affiliation(s)
- Alfred Jatho
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, KR
- Uganda Cancer Institute, Kampala, UG
| | - Binh Thang Tran
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, KR
- Institute of Research and Development, Duy Tan University, Da Nang, VN
| | - Jansen Marcos Cambia
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, KR
| | | | | |
Collapse
|
17
|
Agler CS, Divaris K. Sources of bias in genomics research of oral and dental traits. COMMUNITY DENTAL HEALTH 2020; 37:102-106. [PMID: 32031351 PMCID: PMC7316399 DOI: 10.1922/cdh_specialissue_divaris05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Evidence regarding the genomic basis of oral/dental traits and diseases is a fundamental pillar of the emerging notion of precision health. During the last decade, technological advances have improved the feasibility and affordability of conducting genome-wide association studies (GWAS) and studying the associations of emanating data with both common and rare oral conditions. Most evidence thus far emanates from GWAS of dental caries and periodontal disease that have tested the associations of several million single nucleotide polymorphisms (SNPs) with typically binary, health vs. disease phenotypes. GWAS offer advantages over the previous candidate-gene studies, mainly owing to their agnostic (i.e., unbiased, or hypothesis-free) nature. Nevertheless, GWAS are prone to virtually all sources of random and systematic error. Here, we review common sources of bias in genomics research with focus on GWAS including: type I and II errors, population stratification and heterogeneity, selection bias, adjustment for heritable covariates, appropriate reference panels for imputation, and gene annotation. We argue that valid and precise phenotype measurement is a key requirement, as GWAS sample sizes and thus statistical power increase. Finally, we stress that the lack of diversity of populations with phenotypes and genotypes is a major limitation for the generalizability and ultimate translation of the emerging genomics evidence-base into oral health promotion for all.
Collapse
Affiliation(s)
- Cary S Agler
- Adams School of Dentistry, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Kimon Divaris
- Adams School of Dentistry, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
18
|
Hobbs BD, Cho MH. Why is Disease Penetration So Variable? Role of Genetic Modifiers of Lung Function in Alpha-1 Antitrypsin Deficiency. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:214-223. [PMID: 32621460 DOI: 10.15326/jcopdf.7.3.2019.0159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Individuals with alpha-1 antitrypsin deficiency (AATD) have marked heterogeneity in lung function, suspected to be related to a combination of both environmental (e.g., cigarette smoking) and genetic factors. Lung function is heritable in the general population and in persons with severe AATD. Several genetic modifiers of lung function in persons with AATD have been described; however, replication is lacking. A genome-wide association study (GWAS) of lung function in persons with AATD has yet to be performed and may inform whether genetic determinants of lung function are overlapping in persons with AATD and in the general population. As GWASs require large sample sizes for adequate power, genetic risk scores offer an alternate approach to assess the overlap of genetic determinants of lung function in the general population in persons with AATD. Where GWASs are limited to common genetic variant discovery, whole genome sequencing (for rare variant discovery) and integrative genomic studies (examining the influence of genetic variants on gene, protein, and metabolite levels) offer potential for an expanded discovery of genetic modifiers of lung function in AATD. In the following review we examine past descriptions of genetic modifiers of lung function in AATD and describe a path forward to further investigate and define the likely genetic modifiers of lung function in AATD.
Collapse
Affiliation(s)
- Brian D Hobbs
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Michael H Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
19
|
Pietschnig J, Siegel M, Eder JSN, Gittler G. Effect Declines Are Systematic, Strong, and Ubiquitous: A Meta-Meta-Analysis of the Decline Effect in Intelligence Research. Front Psychol 2019; 10:2874. [PMID: 31920891 PMCID: PMC6930891 DOI: 10.3389/fpsyg.2019.02874] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
Empirical sciences in general and psychological science in particular are plagued by replicability problems and biased published effect sizes. Although dissemination bias-related phenomena such as publication bias, time-lag bias, or visibility bias are well-known and have been intensively studied, another variant of effect distorting mechanisms, so-called decline effects, have not. Conceptually, decline effects are rooted in low initial (exploratory) study power due to strategic researcher behavior and can be expected to yield overproportional effect declines. Although decline effects have been documented in individual meta-analytic investigations, systematic evidence for decline effects in the psychological literature remains to date unavailable. Therefore, we present in this meta-meta-analysis a systematic investigation of the decline effect in intelligence research. In all, data from 22 meta-analyses comprising 36 meta-analytical and 1,391 primary effect sizes (N = 697,000+) that have been published in the journal Intelligence were included in our analyses. Two different analytic approaches showed consistent evidence for a higher prevalence of cross-temporal effect declines compared to effect increases, yielding a ratio of about 2:1. Moreover, effect declines were considerably stronger when referenced to the initial primary study within a meta-analysis, yielding about twice the magnitude of effect increases. Effect misestimations were more substantial when initial studies had smaller sample sizes and reported larger effects, thus indicating suboptimal initial study power as the main driver of effect misestimations in initial studies. Post hoc study power comparisons of initial versus subsequent studies were consistent with this interpretation, showing substantially lower initial study power of declining, than of increasing effects. Our findings add another facet to the ever accumulating evidence about non-trivial effect misestimations in the scientific literature. We therefore stress the necessity for more rigorous protocols when it comes to designing and conducting primary research as well as reporting findings in exploratory and replication studies. Increasing transparency in scientific processes such as data sharing, (exploratory) study preregistration, but also self- (or independent) replication preceding the publication of exploratory findings may be suitable approaches to strengthen the credibility of empirical research in general and psychological science in particular.
Collapse
Affiliation(s)
- Jakob Pietschnig
- Department of Applied Psychology: Health, Development, Enhancement and Intervention, Faculty of Psychology, University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
20
|
Bierut LJ. Using Genomic Profiling for Understanding and Improving Response to Smoking Cessation Treatment. CURR EPIDEMIOL REP 2019; 6:486-490. [DOI: 10.1007/s40471-019-00220-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
Cerrone M, Remme CA, Tadros R, Bezzina CR, Delmar M. Beyond the One Gene-One Disease Paradigm: Complex Genetics and Pleiotropy in Inheritable Cardiac Disorders. Circulation 2019; 140:595-610. [PMID: 31403841 DOI: 10.1161/circulationaha.118.035954] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inheritable cardiac disorders, which may be associated with cardiomyopathic changes, are often associated with increased risk of sudden death in the young. Early linkage analysis studies in Mendelian forms of these diseases, such as hypertrophic cardiomyopathy and long-QT syndrome, uncovered large-effect genetic variants that contribute to the phenotype. In more recent years, through genotype-phenotype studies and methodological advances in genetics, it has become evident that most inheritable cardiac disorders are not monogenic but, rather, have a complex genetic basis wherein multiple genetic variants contribute (oligogenic or polygenic inheritance). Conversely, studies on genes underlying these disorders uncovered pleiotropic effects, with a single gene affecting multiple and apparently unrelated phenotypes. In this review, we explore these 2 phenomena: on the one hand, the evidence that variants in multiple genes converge to generate one clinical phenotype, and, on the other, the evidence that variants in one gene can lead to apparently unrelated phenotypes. Although multiple conditions are addressed to illustrate these concepts, the experience obtained in the study of long-QT syndrome, Brugada syndrome, and arrhythmogenic cardiomyopathy, and in the study of functions related to SCN5A (the gene coding for the α-subunit of the most abundant sodium channel in the heart) and PKP2 (the gene coding for the desmosomal protein plakophilin-2), as well, is discussed in more detail.
Collapse
Affiliation(s)
- Marina Cerrone
- Leon H. Charney Division of Cardiology (M.C., M.D.), NYU School of Medicine, New York.,Inherited Arrhythmias Clinic and Heart Rhythm Center, Leon H. Charney Division of Cardiology (M.C.), NYU School of Medicine, New York
| | - Carol Ann Remme
- Inherited Arrhythmias Clinic and Heart Rhythm Center, Leon H. Charney Division of Cardiology (M.C.), NYU School of Medicine, New York
| | - Rafik Tadros
- Amsterdam UMC, University of Amsterdam, Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, AMC Heart Center, The Netherlands (C.A.R., C.R.B.)
| | - Connie R Bezzina
- Inherited Arrhythmias Clinic and Heart Rhythm Center, Leon H. Charney Division of Cardiology (M.C.), NYU School of Medicine, New York
| | - Mario Delmar
- Leon H. Charney Division of Cardiology (M.C., M.D.), NYU School of Medicine, New York
| |
Collapse
|
22
|
Replication Study for the Association of GWAS-associated Loci With Adolescent Idiopathic Scoliosis Susceptibility and Curve Progression in a Chinese Population. Spine (Phila Pa 1976) 2019; 44:464-471. [PMID: 30234802 DOI: 10.1097/brs.0000000000002866] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN A genetic association (replication) study. OBJECTIVE The aim of this study was to replicate and further evaluate the association among seven genome-wide association studies (GWAS)-identified single nucleotide polymorphisms (SNPs) in Chinese girls with adolescent idiopathic scoliosis (AIS) with disease onset, curve types, and progression. SUMMARY OF BACKGROUND DATA AIS is the most common pediatric spinal deformity with a strong genetic predisposition. Recent GWAS identified 10 new disease predisposition loci for AIS. METHODS Three hundred nineteen female AIS patients with Cobb angle ≥ 10 and 201 healthy controls were studied for the association with disease onset. Seven GWAS-identified SNPs (rs11190870 in LBX1, rs12946942 in SOX9/KCNJ2, rs13398147 in PAX3/EPH4, rs241215 in AJAP1, rs3904778 in BNC2, rs6570507 in GPR126, and rs678741 in LBX1-AS1) were analyzed. In subgroup analysis, AIS patients were subdivided by curve types and disease progression to examine for genotype association. RESULTS We replicated the association with disease onset in four common SNPs rs11190870, rs3904778, rs6570507, and rs678741. In addition, rs1190870 and rs678741 remained significantly associated in the right thoracic curves only subgroup. However, no significant difference was observed with both clinical curve progression or Cobb angle. CONCLUSION This study replicated the associations of four GWAS-associated SNPs with occurrence of AIS in our Chinese population. However, none of these SNPs was associated with curve severity and progression. The results suggest that curve progression may be determined by environmental (nongenetic) factor, but further study with a larger sample size is required to address this issue. LEVEL OF EVIDENCE 4.
Collapse
|
23
|
Kleinstein SE, Shea PR, Allen AS, Koelle DM, Wald A, Goldstein DB. Genome-wide association study (GWAS) of human host factors influencing viral severity of herpes simplex virus type 2 (HSV-2). Genes Immun 2019; 20:112-120. [PMID: 29535370 PMCID: PMC6113125 DOI: 10.1038/s41435-018-0013-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/24/2017] [Accepted: 12/01/2017] [Indexed: 12/28/2022]
Abstract
Herpes simplex virus type 2 (HSV-2) is an incurable viral infection with severity ranging from asymptomatic to frequent recurrences. The viral shedding rate has been shown as a reproducible HSV-2 severity end point that correlates with lesion rates. We used a genome-wide association study (GWAS) to investigate the role of common human genetic variation in HSV-2 severity. We performed a GWAS on 223 HSV-2-positive participants of European ancestry. Severity was measured by viral shedding rate, as defined by the percent of days PCR+ for HSV-2 DNA over at least 30 days. Analyses were performed under linear regression models, adjusted for age, sex, and ancestry. There were no genome-wide significant (p < 5E-08) associations with HSV-2 viral shedding rate. The top nonsignificant SNP (rs75932292, p = 6.77E-08) associated with HSV-2 viral shedding was intergenic, with the nearest known biologically interesting gene (ABCA1) ~130 kbp downstream. Several other SNPs approaching significance were in or near genes with viral or neurological associations, including four SNPs in KIF1B. The current study is the first comprehensive genome-wide investigation of human genetic variation in virologic severity of established HSV-2 infection. However, no significant associations were observed with HSV-2 virologic severity, leaving the exact role of human variation in HSV-2 severity unclear.
Collapse
Affiliation(s)
- Sarah E Kleinstein
- Institute for Genomic Medicine, Columbia University, New York, NY, 10032, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Patrick R Shea
- Institute for Genomic Medicine, Columbia University, New York, NY, 10032, USA
| | - Andrew S Allen
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27708, USA
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Benaroya Research Institute, Seattle, WA, 98101, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA, 98195, USA
- Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Anna Wald
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Department of Epidemiology, University of Washington, Seattle, WA, 98195, USA
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
24
|
Reiner AP, Johnson AD. Platelet Genomics. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
25
|
Livshits G, Malkin I, Bowyer RC, Verdi S, Bell JT, Menni C, Williams FM, Steves CJ. Multi-OMICS analyses of frailty and chronic widespread musculoskeletal pain suggest involvement of shared neurological pathways. Pain 2018; 159:2565-2572. [PMID: 30086113 PMCID: PMC6250282 DOI: 10.1097/j.pain.0000000000001364] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/25/2018] [Indexed: 01/25/2023]
Abstract
Chronic widespread musculoskeletal pain (CWP) and frailty are prevalent conditions in older people. We have shown previously that interindividual variation in frailty and CWP is genetically determined. We also reported an association of frailty and CWP caused by shared genetic and common environmental factors. The aim of this study was to use omic approaches to identify molecular genetic factors underlying the heritability of frailty and its genetic correlation with CWP. Frailty was quantified through the Rockwood Frailty Index (FI) as a proportion of deficits from 33 binary health deficit questions in 3626 female twins. Common widespread pain was assessed using a screening questionnaire. OMICS analysis included 305 metabolites and whole-genome (>2.5 × 10 SNPs) and epigenome (∼1 × 10 MeDIP-seq regions) assessments performed on fasting blood samples. Using family-based statistical analyses, including path analysis, we examined how FI scores were related to molecular genetic factors and to CWP, taking into account known risk factors such as fat mass and smoking. Frailty Index was significantly correlated with 51 metabolites after correction for multiple testing, with 20 metabolites having P-values between 2.1 × 10 and 4.0 × 10. Three metabolites (uridine, C-glycosyl tryptophan, and N-acetyl glycine) were statistically independent and thought to exert a direct effect on FI. Epiandrosterone sulphate, previously shown to be highly inversely associated with CWP, was found to exert an indirect influence on FI. Bioinformatics analysis of genome-wide association study and EWAS showed that FI and its covariation with CWP was through genomic regions involved in neurological pathways. Neurological pathway involvement accounts for the associated conditions of aging CWP and FI.
Collapse
Affiliation(s)
- Gregory Livshits
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Ida Malkin
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruth C.E. Bowyer
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Serena Verdi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Frances M.K. Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Claire J. Steves
- Clinical Age Research Unit, King's College Hospitals Foundation Trust, London, United Kingdom
| |
Collapse
|
26
|
Marigorta UM, Rodríguez JA, Gibson G, Navarro A. Replicability and Prediction: Lessons and Challenges from GWAS. Trends Genet 2018; 34:504-517. [PMID: 29716745 PMCID: PMC6003860 DOI: 10.1016/j.tig.2018.03.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/12/2018] [Accepted: 03/26/2018] [Indexed: 12/29/2022]
Abstract
Since the publication of the Wellcome Trust Case Control Consortium (WTCCC) landmark study a decade ago, genome-wide association studies (GWAS) have led to the discovery of thousands of risk variants involved in disease etiology. This success story has two angles that are often overlooked. First, GWAS findings are highly replicable. This is an unprecedented phenomenon in complex trait genetics, and indeed in many areas of science, which in past decades have been plagued by false positives. At a time of increasing concerns about the lack of reproducibility, we examine the biological and methodological reasons that account for the replicability of GWAS and identify the challenges ahead. In contrast to the exemplary success of disease gene discovery, at present GWAS findings are not useful for predicting phenotypes. We close with an overview of the prospects for individualized prediction of disease risk and its foreseeable impact in clinical practice.
Collapse
Affiliation(s)
- Urko M Marigorta
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA; These authors contributed equally
| | - Juan Antonio Rodríguez
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain; Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona, Catalonia, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain; These authors contributed equally. https://twitter.com/jrotwitguez
| | - Greg Gibson
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Arcadi Navarro
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain; Institute of Evolutionary Biology (UPF-CSIC), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain; National Institute for Bioinformatics (INB), Barcelona, Catalonia, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), PRBB, Barcelona, Catalonia, Spain.
| |
Collapse
|
27
|
Madlon-Kay S, Montague MJ, Brent LJN, Ellis S, Zhong B, Snyder-Mackler N, Horvath JE, Skene JHP, Platt ML. Weak effects of common genetic variation in oxytocin and vasopressin receptor genes on rhesus macaque social behavior. Am J Primatol 2018; 80:e22873. [PMID: 29931777 DOI: 10.1002/ajp.22873] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/01/2018] [Accepted: 04/02/2018] [Indexed: 02/02/2023]
Abstract
The neuropeptides oxytocin (OT) and arginine vasopressin (AVP) influence pair bonding, attachment, and sociality, as well as anxiety and stress responses in humans and other mammals. The effects of these peptides are mediated by genetic variability in their associated receptors, OXTR and the AVPR gene family. However, the role of these genes in regulating social behaviors in non-human primates is not well understood. To address this question, we examined whether genetic variation in the OT receptor gene OXTR and the AVP receptor genes AVPR1A and AVPR1B influence naturally-occurring social behavior in free-ranging rhesus macaques-gregarious primates that share many features of their biology and social behavior with humans. We assessed rates of social behavior across 3,250 hr of observational behavioral data from 201 free-ranging rhesus macaques on Cayo Santiago island in Puerto Rico, and used genetic sequence data to identify 25 OXTR, AVPR1A, and AVPR1B single-nucleotide variants (SNVs) in the population. We used an animal model to estimate the effects of 12 SNVs (n = 3 OXTR; n = 5 AVPR1A; n = 4 AVPR1B) on rates of grooming, approaches, passive contact, contact aggression, and non-contact aggression, given and received. Though we found evidence for modest heritability of these behaviors, estimates of effect sizes of the selected SNVs were close to zero, indicating that common OXTR and AVPR variation contributed little to social behavior in these animals. Our results are consistent with recent findings in human genetics that the effects of individual common genetic variants on complex phenotypes are generally small.
Collapse
Affiliation(s)
- Seth Madlon-Kay
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael J Montague
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lauren J N Brent
- Centre for Research in Animal Behaviour, University of Exeter, Exeter, Devon
| | - Samuel Ellis
- Centre for Research in Animal Behaviour, University of Exeter, Exeter, Devon
| | - Brian Zhong
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Noah Snyder-Mackler
- Department of Psychology, University of Washington, Seattle, Washington.,Center for Studies in Demography and Ecology, University of Washington, Seattle, Washington.,Washington National Primate Research Center, University of Washington, Seattle, Washington
| | - Julie E Horvath
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, North Carolina.,North Carolina Museum of Natural Sciences, Raleigh, North Carolina.,Department of Evolutionary Anthropology, Duke University, Durham, North Carolina
| | | | - Michael L Platt
- Department of Psychology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Marketing, The Wharton School, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
28
|
Traylor M, Walker JL, Corrigan AA, Hernandez MA, Newhouse SJ, Folarin AA, Patel H, Ross PJ, Sanderson JD, Spicer J, Prescott NJ, Mathew CG, Marinaki AM, Lewis CM. Exome array analysis of adverse reactions to fluoropyrimidine-based therapy for gastrointestinal cancer. PLoS One 2018; 13:e0188911. [PMID: 29715290 PMCID: PMC5929530 DOI: 10.1371/journal.pone.0188911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/15/2017] [Indexed: 12/31/2022] Open
Abstract
Fluoropyrimidines, including 5-fluororacil (5FU) and its pro-drug Capecitabine, are the common treatment for colorectal, breast, neck and head cancers—either as monotherapy or in combination therapy. Adverse reactions (ADRs) to the treatment are common and often result in treatment discontinuation or dose reduction. Factors contributing to ADRs, including genetic variation, are poorly characterized. We performed exome array analysis to identify genetic variants that contribute to adverse reactions. Our final dataset consisted of 504 European ancestry individuals undergoing fluoropyrimidine-based therapy for gastrointestinal cancer. A subset of 254 of these were treated with Capecitabine. All individuals were genotyped on the Illumina HumanExome Array. Firstly, we performed SNP and gene-level analyses of protein-altering variants on the array to identify novel associations the following ADRs, which were grouped into four phenotypes based on symptoms of diarrhea, mucositis, and neutropenia and hand-and-foot syndrome. Secondly, we performed detailed analyses of the HLA region on the same phenotypes after imputing the HLA alleles and amino acids. No protein-altering variants, or sets of protein-altering variants collapsed into genes, were associated with the main outcomes after Bonferroni correction. We found evidence that the HLA region was enriched for associations with Hand-and-Foot syndrome (p = 0.023), but no specific SNPs or HLA alleles were significant after Bonferroni correction. Larger studies will be required to characterize the genetic contribution to ADRs to 5FU. Future studies that focus on the HLA region are likely to be fruitful.
Collapse
Affiliation(s)
- Matthew Traylor
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
- * E-mail:
| | - Jemma L. Walker
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Adele A. Corrigan
- Purine Research Laboratory, GSTS Pathology, Guy’s and St. Thomas’ Hospital NHS Foundation Trust, St. Thomas Hospital, London, United Kingdom
| | - Monica A. Hernandez
- Purine Research Laboratory, GSTS Pathology, Guy’s and St. Thomas’ Hospital NHS Foundation Trust, St. Thomas Hospital, London, United Kingdom
| | - Stephen J. Newhouse
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health at South London and Maudsley NHS Foundation Trust and (Institute of Psychiatry), King’s College London, London, United Kingdom
| | - Amos A. Folarin
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health at South London and Maudsley NHS Foundation Trust and (Institute of Psychiatry), King’s College London, London, United Kingdom
| | - Hamel Patel
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health at South London and Maudsley NHS Foundation Trust and (Institute of Psychiatry), King’s College London, London, United Kingdom
| | - Paul J. Ross
- Department of Gastroenterology, Guy’s and St. Thomas’ NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Jeremy D. Sanderson
- Department of Gastroenterology, Guy’s and St. Thomas’ NHS Foundation Trust and King’s College London, London, United Kingdom
| | - James Spicer
- Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Natalie J. Prescott
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
| | - Christopher G. Mathew
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
| | - Anthony M. Marinaki
- Purine Research Laboratory, GSTS Pathology, Guy’s and St. Thomas’ Hospital NHS Foundation Trust, St. Thomas Hospital, London, United Kingdom
| | - Cathryn M. Lewis
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
29
|
Schielzeth H, Rios Villamil A, Burri R. Success and failure in replication of genotype-phenotype associations: How does replication help in understanding the genetic basis of phenotypic variation in outbred populations? Mol Ecol Resour 2018; 18:739-754. [PMID: 29575806 DOI: 10.1111/1755-0998.12780] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 12/29/2022]
Abstract
Recent developments in sequencing technologies have facilitated genomewide mapping of phenotypic variation in natural populations. Such mapping efforts face a number of challenges potentially leading to low reproducibility. However, reproducible research forms the basis of scientific progress. We here discuss the options for replication and the reasons for potential nonreproducibility. We then review the evidence for reproducible quantitative trait loci (QTL) with a focus on natural animal populations. Existing case studies of replication fall into three categories: (i) traits that have been mapped to major effect loci (including chromosomal inversion and supergenes) by independent research teams; (ii) QTL fine-mapped in discovery populations; and (iii) attempts to replicate QTL across multiple populations. Major effect loci, in particular those associated with inversions, have been successfully replicated in several cases within and across populations. Beyond such major effect variants, replication has been more successful within than across populations, suggesting that QTL discovered in natural populations may often be population-specific. This suggests that biological causes (differences in linkage patterns, allele frequencies or context-dependencies of QTL) contribute to nonreproducibility. Evidence from other fields, notably animal breeding and QTL mapping in humans, suggests that a significant fraction of QTL is indeed reproducible in direction and magnitude at least within populations. However, there is also a large number of QTL that cannot be easily reproduced. We put forward that more studies should explicitly address the causes and context-dependencies of QTL signals, in particular to disentangle linkage differences, allele frequency differences and gene-by-environment interactions as biological causes of nonreproducibility of QTL, especially between populations.
Collapse
Affiliation(s)
- Holger Schielzeth
- Population Ecology Group, Institute of Ecology and Evolution, Friedrich Schiller University, Jena, Germany
| | - Alejandro Rios Villamil
- Population Ecology Group, Institute of Ecology and Evolution, Friedrich Schiller University, Jena, Germany
| | - Reto Burri
- Population Ecology Group, Institute of Ecology and Evolution, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
30
|
Shridhar K, Aggarwal A, Walia GK, Gulati S, Geetha AV, Prabhakaran D, Dhillon PK, Rajaraman P. Single nucleotide polymorphisms as markers of genetic susceptibility for oral potentially malignant disorders risk: Review of evidence to date. Oral Oncol 2018; 61:146-51. [PMID: 27688118 PMCID: PMC5046699 DOI: 10.1016/j.oraloncology.2016.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/28/2016] [Accepted: 08/18/2016] [Indexed: 12/11/2022]
Abstract
We reviewed single nucleotide polymorphisms for oral pre-cancer susceptibility. All of them were pathway based candidate gene association studies. The current level of evidence is very limited. Integrated characterization of germline/somatic alterations in oral cancer & pre-cancer is needed.
Background Oral cancers are preceded by oral potentially malignant disorders (OPMD). Understanding genetic susceptibility for OPMD risk could provide an opportunity for risk assessment of oral cancer through early disease course. We conducted a review of single nucleotide polymorphism (SNP) studies for OPMD risk. Methods We identified all relevant studies examining associations of SNPs with OPMD (leukoplakia, erythroplakia and oral sub-mucous fibrosis) conducted world-wide between January, 2000 and February, 2016 using a combined keyword search on PubMed. Of these, 47 studies that presented results as odds ratios and 95% CI were considered for full review. Results The majority of eligible studies that explored candidate gene associations for OPMD were small (N < 200 cases), limiting their scope to provide strong inference for any SNP identified to date in any population. Commonly studied SNPs were genes of carcinogen metabolism (n = 18 studies), DNA repair (n = 11 studies), cell cycle control (n = 8 studies), extra-cellular matrix alteration (n = 8 studies) and immune-inflammatory (n = 6 studies) pathways. Based on significant associations as reported by two or more studies, suggestive markers included SNPs in GSTM1 (null), CCND1 (G870A), MMP3 (-1171; promotor region), TNFα (-308; rs800629), XPD (codon 751) and Gemin3 (rs197412) as well as in p53 (codon 72) in Indian populations. However, an equal or greater number of studies reported null or mixed associations for SNPs in GSTM1 (null), p53 (codon 72), XPD (codon 751), XRCC (rs25487 C/T), GSTT1 (null) and CYP1A1m1 (MspI site). Conclusion Candidate gene association studies have not yielded consistent data on risk loci for OPMD. High-throughput genotyping approaches for OPMD, with concurrent efforts for oral cancer, could prove useful in identifying robust risk-loci to help understand early disease course susceptibility for oral cancer.
Collapse
Affiliation(s)
- Krithiga Shridhar
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Haryana, India.
| | - Aastha Aggarwal
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Haryana, India.
| | - Gagandeep Kaur Walia
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Haryana, India.
| | - Smriti Gulati
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Haryana, India.
| | - A V Geetha
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Haryana, India.
| | - D Prabhakaran
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Haryana, India; Centre for Chronic Disease Control, Haryana, India; London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | - Preet K Dhillon
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Haryana, India.
| | | |
Collapse
|
31
|
Kafkafi N, Agassi J, Chesler EJ, Crabbe JC, Crusio WE, Eilam D, Gerlai R, Golani I, Gomez-Marin A, Heller R, Iraqi F, Jaljuli I, Karp NA, Morgan H, Nicholson G, Pfaff DW, Richter SH, Stark PB, Stiedl O, Stodden V, Tarantino LM, Tucci V, Valdar W, Williams RW, Würbel H, Benjamini Y. Reproducibility and replicability of rodent phenotyping in preclinical studies. Neurosci Biobehav Rev 2018; 87:218-232. [PMID: 29357292 PMCID: PMC6071910 DOI: 10.1016/j.neubiorev.2018.01.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/13/2017] [Accepted: 01/11/2018] [Indexed: 12/15/2022]
Abstract
The scientific community is increasingly concerned with the proportion of
published “discoveries” that are not replicated in subsequent
studies. The field of rodent behavioral phenotyping was one of the first to
raise this concern, and to relate it to other methodological issues: the complex
interaction between genotype and environment; the definitions of behavioral
constructs; and the use of laboratory mice and rats as model species for
investigating human health and disease mechanisms. In January 2015, researchers
from various disciplines gathered at Tel Aviv University to discuss these
issues. The general consensus was that the issue is prevalent and of concern,
and should be addressed at the statistical, methodological and policy levels,
but is not so severe as to call into question the validity and the usefulness of
model organisms as a whole. Well-organized community efforts, coupled with
improved data and metadata sharing, have a key role in identifying specific
problems and promoting effective solutions. Replicability is closely related to
validity, may affect generalizability and translation of findings, and has
important ethical implications.
Collapse
Affiliation(s)
| | | | | | - John C Crabbe
- Oregon Health & Science University, and VA Portland Health Care System, United States
| | | | | | | | | | | | | | | | | | - Natasha A Karp
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | - William Valdar
- University of North Carolina at Chapel Hill, United States
| | | | | | | |
Collapse
|
32
|
Roos TR, Roos AK, Avins AL, Ahmed MA, Kleimeyer JP, Fredericson M, Ioannidis JPA, Dragoo JL, Kim SK. Genome-wide association study identifies a locus associated with rotator cuff injury. PLoS One 2017; 12:e0189317. [PMID: 29228018 PMCID: PMC5724859 DOI: 10.1371/journal.pone.0189317] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/22/2017] [Indexed: 01/10/2023] Open
Abstract
Rotator cuff tears are common, especially in the fifth and sixth decades of life, but can also occur in the competitive athlete. Genetic differences may contribute to overall injury risk. Identifying genetic loci associated with rotator cuff injury could shed light on the etiology of this injury. We performed a genome-wide association screen using publically available data from the Research Program in Genes, Environment and Health including 8,357 cases of rotator cuff injury and 94,622 controls. We found rs71404070 to show a genome-wide significant association with rotator cuff injury with p = 2.31x10-8 and an odds ratio of 1.25 per allele. This SNP is located next to cadherin8, which encodes a protein involved in cell adhesion. We also attempted to validate previous gene association studies that had reported a total of 18 SNPs showing a significant association with rotator cuff injury. However, none of the 18 SNPs were validated in our dataset. rs71404070 may be informative in explaining why some individuals are more susceptible to rotator cuff injury than others.
Collapse
Affiliation(s)
- Thomas R. Roos
- Department of Developmental Biology, Stanford University Medical Center, Stanford, CA, United States of America
- Department of Health Research and Policy, Division of Epidemiology, Stanford University Medical Center, Stanford, CA, United States of America
| | - Andrew K. Roos
- Department of Developmental Biology, Stanford University Medical Center, Stanford, CA, United States of America
- Department of Health Research and Policy, Division of Epidemiology, Stanford University Medical Center, Stanford, CA, United States of America
| | - Andrew L. Avins
- Kaiser Permanente Northern California, Division of Research, Oakland, CA, United States of America
| | - Marwa A. Ahmed
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, United States of America
| | - John P. Kleimeyer
- Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA, United States of America
| | - Michael Fredericson
- Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA, United States of America
| | - John P. A. Ioannidis
- Department of Medicine, Stanford Prevention Research Center and Dept. of Health Research and Policy, Division of Epidemiology, Stanford University School of Medicine, and Dept. of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, United States of America
| | - Jason L. Dragoo
- Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA, United States of America
| | - Stuart K. Kim
- Department of Developmental Biology, Stanford University Medical Center, Stanford, CA, United States of America
| |
Collapse
|
33
|
Yeung EW, Craggs JG, Gizer IR. Comorbidity of Alcohol Use Disorder and Chronic Pain: Genetic Influences on Brain Reward and Stress Systems. Alcohol Clin Exp Res 2017; 41:1831-1848. [PMID: 29048744 DOI: 10.1111/acer.13491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/25/2017] [Indexed: 01/10/2023]
Abstract
Alcohol use disorder (AUD) is highly comorbid with chronic pain (CP). Evidence has suggested that neuroadaptive processes characterized by reward deficit and stress surfeit are involved in the development of AUD and pain chronification. Neurological data suggest that shared genetic architecture associated with the reward and stress systems may contribute to the comorbidity of AUD and CP. This monograph first delineates the prevailing theories of the development of AUD and pain chronification focusing on the reward and stress systems. It then provides a brief summary of relevant neurological findings followed by an evaluation of evidence documented by molecular genetic studies. Candidate gene association studies have provided some initial support for the genetic overlap between AUD and CP; however, these results must be interpreted with caution until studies with sufficient statistical power are conducted and replications obtained. Genomewide association studies have suggested a number of genes (e.g., TBX19, HTR7, and ADRA1A) that are either directly or indirectly related to the reward and stress systems in the AUD and CP literature. Evidence reviewed in this monograph suggests that shared genetic liability underlying the comorbidity between AUD and CP, if present, is likely to be complex. As the advancement in molecular genetic methods continues, future studies may show broader central nervous system involvement in AUD-CP comorbidity.
Collapse
Affiliation(s)
- Ellen W Yeung
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri.,Institute for Interdisciplinary Salivary Bioscience Research, University of California at Irvine, Irvine, California
| | - Jason G Craggs
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri.,School of Health Professions, University of Missouri, Columbia, Missouri
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
34
|
On the transdiagnostic nature of peripheral biomarkers in major psychiatric disorders: A systematic review. Neurosci Biobehav Rev 2017; 83:97-108. [PMID: 28986182 DOI: 10.1016/j.neubiorev.2017.10.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/31/2017] [Accepted: 10/01/2017] [Indexed: 12/20/2022]
Abstract
The search for biomarkers has been a leading endeavor in biological psychiatry. To analyze its evolution over the years, we performed a systematic review to evaluate (a) the most studied peripheral molecular markers in major psychiatric disorders, (b) the main features of studies proposing them as biomarkers and (c) whether their patterns of variation are similar across disorders. Of the six molecules most commonly studied as plasmatic markers of schizophrenia, major depressive disorder or bipolar disorder, five (BDNF, TNF-alpha, IL-6, C-reactive protein and cortisol) were the same across diagnoses. An analysis of this literature showed that, while 66% of studies compared patients and controls, only 34% were longitudinal, and only 10% presented a measure of diagnostic or prognostic efficacy. Meta-analyses showed variation in the levels of these molecules to be robust across studies, but similar among disorders, suggesting them to reflect transdiagnostic systemic consequences of psychiatric illness. Based on this, we discuss how current publication practices have led to research fragmentation across diagnoses, and suggest approaches to face this issue.
Collapse
|
35
|
Kim SK, Kleimeyer JP, Ahmed MA, Avins AL, Fredericson M, Dragoo JL, Ioannidis JPA. Two genetic loci associated with ankle injury. PLoS One 2017; 12:e0185355. [PMID: 28957384 PMCID: PMC5619760 DOI: 10.1371/journal.pone.0185355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/11/2017] [Indexed: 11/18/2022] Open
Abstract
Ankle injuries, including sprains, strains and other joint derangements and instability, are common, especially for athletes involved in indoor court or jumping sports. Identifying genetic loci associated with these ankle injuries could shed light on their etiologies. A genome-wide association screen was performed using publicly available data from the Research Program in Genes, Environment and Health (RPGEH) including 1,694 cases of ankle injury and 97,646 controls. An indel (chr21:47156779:D) that lies close to a collagen gene, COL18A1, showed an association with ankle injury at genome-wide significance (p = 3.8x10-8; OR = 1.99; 95% CI = 1.75-2.23). A second DNA variant (rs13286037 on chromosome 9) that lies within an intron of the transcription factor gene NFIB showed an association that was nearly genome-wide significant (p = 5.1x10-8; OR = 1.63; 95% CI = 1.46-1.80). The ACTN3 R577X mutation was previously reported to show an association with acute ankle sprains, but did not show an association in this cohort. This study is the first genome-wide screen for ankle injury that yields insights regarding the genetic etiology of ankle injuries and provides DNA markers with the potential to inform athletes about their genetic risk for ankle injury.
Collapse
Affiliation(s)
- Stuart K. Kim
- Dept. Developmental Biology, Stanford University Medical Center, Stanford, CA, United States of America
| | - John P. Kleimeyer
- Dept. Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA, United States of America
| | - Marwa A. Ahmed
- Dept. Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, United States of America
| | - Andrew L. Avins
- Kaiser Permanente Northern California, Division of Research, Oakland, CA, United States of America
| | - Michael Fredericson
- Dept. Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA, United States of America
| | - Jason L. Dragoo
- Dept. Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA, United States of America
| | - John P. A. Ioannidis
- Dept. of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, United States of America
- Dept. of Health Research and Policy, Division of Epidemiology, Stanford University School of Medicine, Stanford, CA, United States of America
- Dept. of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, United States of America
| |
Collapse
|
36
|
Gondalia R, Avery CL, Napier MD, Méndez-Giráldez R, Stewart JD, Sitlani CM, Li Y, Wilhelmsen KC, Duan Q, Roach J, North KE, Reiner AP, Zhang ZM, Tinker LF, Yanosky JD, Liao D, Whitsel EA. Genome-wide Association Study of Susceptibility to Particulate Matter-Associated QT Prolongation. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:067002. [PMID: 28749367 PMCID: PMC5714283 DOI: 10.1289/ehp347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 09/07/2016] [Accepted: 09/19/2016] [Indexed: 05/02/2023]
Abstract
BACKGROUND Ambient particulate matter (PM) air pollution exposure has been associated with increases in QT interval duration (QT). However, innate susceptibility to PM-associated QT prolongation has not been characterized. OBJECTIVE To characterize genetic susceptibility to PM-associated QT prolongation in a multi-racial/ethnic, genome-wide association study (GWAS). METHODS Using repeated electrocardiograms (1986–2004), longitudinal data on PM<10 μm in diameter (PM10), and generalized estimating equations methods adapted for low-prevalence exposure, we estimated approximately 2.5×106 SNP×PM10 interactions among nine Women’s Health Initiative clinical trials and Atherosclerosis Risk in Communities Study subpopulations (n=22,158), then combined subpopulation-specific results in a fixed-effects, inverse variance-weighted meta-analysis. RESULTS A common variant (rs1619661; coded allele: T) significantly modified the QT-PM10 association (p=2.11×10−8). At PM10 concentrations >90th percentile, QT increased 7 ms across the CC and TT genotypes: 397 (95% confidence interval: 396, 399) to 404 (403, 404) ms. However, QT changed minimally across rs1619661 genotypes at lower PM10 concentrations. The rs1619661 variant is on chromosome 10, 132 kilobase (kb) downstream from CXCL12, which encodes a chemokine, stromal cell-derived factor 1, that is expressed in cardiomyocytes and decreases calcium influx across the L-type Ca2+ channel. CONCLUSIONS The findings suggest that biologically plausible genetic factors may alter susceptibility to PM10-associated QT prolongation in populations protected by the U.S. Environmental Protection Agency’s National Ambient Air Quality Standards. Independent replication and functional characterization are necessary to validate our findings. https://doi.org/10.1289/EHP347
Collapse
Affiliation(s)
- Rahul Gondalia
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Christy L Avery
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Melanie D Napier
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Raúl Méndez-Giráldez
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - James D Stewart
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Computer Science, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kirk C Wilhelmsen
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
- The Renaissance Computing Institute, Chapel Hill, North Carolina, USA
| | - Qing Duan
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jeffrey Roach
- Research Computing Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
- Carolina Center for Genome Sciences, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Alexander P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Zhu-Ming Zhang
- Epidemiologic Cardiology Research Center, Dept. of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Lesley F Tinker
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jeff D Yanosky
- Division of Epidemiology, Dept. of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Duanping Liao
- Division of Epidemiology, Dept. of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Eric A Whitsel
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
37
|
Jiang H, Yang Q, Jiang J, Zhan X, Xiao Z. Association between COL11A1 (rs1337185) and ADAMTS5 (rs162509) gene polymorphisms and lumbar spine pathologies in Chinese Han population: an observational study. BMJ Open 2017; 7:e015644. [PMID: 28583914 PMCID: PMC5623369 DOI: 10.1136/bmjopen-2016-015644] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES A previous study identified a significant association between several single nucleotide polymorphisms (SNPs) and lumbar disc degeneration (LDD) in Indians. To validate the association between these SNPs and specific lumbar spine pathologies, we performed a case-control study in Chinese Han population. DESIGN An observational study. SETTING University Hospital in Nanning, China. PARTICIPANTS This study included 428 patients with LDD and 400 normal controls. OUTCOME MEASURES Patients with LDD were classified into four subgroups, including disc herniation only (subgroup 1), discopathies or/and osteochondrosis associated with disc herniation (subgroup 2), spinal stenosis or/and spondylolisthesis (subgroup 3) and degenerative scoliosis (subgroup 4). This study was conducted by examining two aspects: environmental factors and SNP genotyping. The environmental factors were evaluated with a questionnaire survey including questions about body mass index, smoking habits, the physical demands of their job and exposure to vibrations. Rs1337185, rs5275, rs5277, rs7575934, rs3213718 and rs162509 were genotyped using a PCR-based invader assay. RESULTS The physical workload was significantly higher in patients with lumbar spine pathologies than in the normal controls (p=0.035). The genotype and allele frequencies of rs1337185 and rs162509 were significantly different between the patients with LDD and the normal controls. In rs1337185, a significant association was found between the C allele (risk allele) and the presence of disc herniation (OR=1.80; 95% CI 1.21 to 2.68; p=0.003, adjusted p=0.012) and the presence of spinal stenosis and spondylolisthesis (OR=1.92; 95% CI 1.29 to 2.89; p=0.001, adjusted p=0.004). In rs162509, the G allele represented 1.58-fold increased risk to suffer from disc herniation (OR=1.58; 95% CI 1.20 to 2.09; p=0.001, adjusted p=0.004). CONCLUSION The SNPs rs1337185 in COL11A1 and rs162509 in ADAMTS5 are associated with susceptibility to LDD. The C allele of rs1337185 is risky for patients who are affected by lumbar pathologies such as disc herniation, stenosis and spondylolisthesis. The G allele of rs16250 represents a risk factor for the development of disc herniation.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qinghua Yang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jie Jiang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinli Zhan
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zengming Xiao
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
38
|
Kim SK, Roos TR, Roos AK, Kleimeyer JP, Ahmed MA, Goodlin GT, Fredericson M, Ioannidis JPA, Avins AL, Dragoo JL. Genome-wide association screens for Achilles tendon and ACL tears and tendinopathy. PLoS One 2017; 12:e0170422. [PMID: 28358823 PMCID: PMC5373512 DOI: 10.1371/journal.pone.0170422] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/04/2017] [Indexed: 11/18/2022] Open
Abstract
Achilles tendinopathy or rupture and anterior cruciate ligament (ACL) rupture are substantial injuries affecting athletes, associated with delayed recovery or inability to return to competition. To identify genetic markers that might be used to predict risk for these injuries, we performed genome-wide association screens for these injuries using data from the Genetic Epidemiology Research on Adult Health and Aging (GERA) cohort consisting of 102,979 individuals. We did not find any single nucleotide polymorphisms (SNPs) associated with either of these injuries with a p-value that was genome-wide significant (p<5x10-8). We found, however, four and three polymorphisms with p-values that were borderline significant (p<10-6) for Achilles tendon injury and ACL rupture, respectively. We then tested SNPs previously reported to be associated with either Achilles tendon injury or ACL rupture. None showed an association in our cohort with a false discovery rate of less than 5%. We obtained, however, moderate to weak evidence for replication in one case; specifically, rs4919510 in MIR608 had a p-value of 5.1x10-3 for association with Achilles tendon injury, corresponding to a 7% chance of false replication. Finally, we tested 2855 SNPs in 90 candidate genes for musculoskeletal injury, but did not find any that showed a significant association below a false discovery rate of 5%. We provide data containing summary statistics for the entire genome, which will be useful for future genetic studies on these injuries.
Collapse
Affiliation(s)
- Stuart K. Kim
- Department Developmental Biology, Stanford University Medical Center, Stanford CA, United States of America
| | - Thomas R. Roos
- Department Developmental Biology, Stanford University Medical Center, Stanford CA, United States of America
- Department Health Research and Policy, Division of Epidemiology, Stanford University Medical Center, Stanford CA, United States of America
| | - Andrew K. Roos
- Department Developmental Biology, Stanford University Medical Center, Stanford CA, United States of America
- Department Health Research and Policy, Division of Epidemiology, Stanford University Medical Center, Stanford CA, United States of America
| | - John P. Kleimeyer
- Department Orthopaedic Surgery, Stanford University Medical Center, Stanford CA, United States of America
| | - Marwa A. Ahmed
- Department Orthopaedic Surgery, Stanford University Medical Center, Stanford CA, United States of America
| | - Gabrielle T. Goodlin
- College of Medicine, California Northstate University, Elk Grove CA, United States of America
| | - Michael Fredericson
- Department Orthopaedic Surgery, Stanford University Medical Center, Stanford CA, United States of America
| | - John P. A. Ioannidis
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford CA, United States of America
- Department of Health Research and Policy, Division of Epidemiology, Stanford University School of Medicine, Stanford CA, United States of America
- Department of Statistics, Stanford University School of Humanities and Sciences, Stanford CA, United States of America
| | - Andrew L. Avins
- Kaiser Permanente Northern California, Division of Research, Oakland, California, United States of America
| | - Jason L. Dragoo
- Department Orthopaedic Surgery, Stanford University Medical Center, Stanford CA, United States of America
| |
Collapse
|
39
|
Ortega MS, Denicol AC, Cole JB, Null DJ, Taylor JF, Schnabel RD, Hansen PJ. Association of single nucleotide polymorphisms in candidate genes previously related to genetic variation in fertility with phenotypic measurements of reproductive function in Holstein cows. J Dairy Sci 2017; 100:3725-3734. [PMID: 28259397 DOI: 10.3168/jds.2016-12260] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 01/07/2017] [Indexed: 11/19/2022]
Abstract
Many genetic markers related to health or production traits are not evaluated in populations independent of the discovery population or related to phenotype. Here we evaluated 68 single nucleotide polymorphisms (SNP) in candidate genes previously associated with genetic merit for fertility and production traits for association with phenotypic measurements of fertility in a population of Holstein cows that was selected based on predicted transmitting ability (PTA) for daughter pregnancy rate (DPR; high, ≥1, n = 989; low, ≤ -1.0, n = 1,285). Cows with a high PTA for DPR had higher pregnancy rate at first service, fewer services per conception, and fewer days open than cows with a low PTA for DPR. Of the 68 SNP, 11 were associated with pregnancy rate at first service, 16 with services per conception, and 19 with days open. Single nucleotide polymorphisms in 12 genes (BDH2, BSP3, CAST, CD2, CD14, FUT1, FYB, GCNT3, HSD17B7, IBSP, OCLN, and PCCB) had significant associations with 2 fertility traits, and SNP in 4 genes (CSPP1, FCER1G, PMM2, and TBC1D24) had significant associations with each of the 3 traits. Results from this experiment were compared with results from 2 earlier studies in which the SNP were associated with genetic estimates of fertility. One study involved the same animals as used here, and the other study was of an independent population of bulls. A total of 13 SNP associated with 1 or more phenotypic estimates of fertility were directionally associated with genetic estimates of fertility in the same cow population. Moreover, 14 SNP associated with reproductive phenotype were directionally associated with genetic estimates of fertility in the bull population. Nine SNP (located in BCAS, BSP3, CAST, FUT1, HSD17B7, OCLN, PCCB, PMM2, and TBC1D24) had a directional association with fertility in all 3 studies. Examination of the function of the genes with SNP associated with reproduction in more than one study indicates the importance of steroid hormones and immune function as determinants of reproductive function. All but 1 of the 68 evaluated SNP were variable in 11 breeds besides Holstein, indicating the potential effects of these SNP on reproductive function across breeds of cattle.
Collapse
Affiliation(s)
- M Sofia Ortega
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program and Genetics Institute, University of Florida, Gainesville 32611
| | - Anna C Denicol
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program and Genetics Institute, University of Florida, Gainesville 32611
| | - John B Cole
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705
| | - Daniel J Null
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705
| | - Jeremy F Taylor
- Division of Animal Sciences, University of Missouri, Columbia 65211
| | - Robert D Schnabel
- Division of Animal Sciences, University of Missouri, Columbia 65211; Informatics Institute, University of Missouri, Columbia 65211
| | - Peter J Hansen
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program and Genetics Institute, University of Florida, Gainesville 32611.
| |
Collapse
|
40
|
Nagrani R, Mhatre S, Rajaraman P, Chatterjee N, Akbari MR, Boffetta P, Brennan P, Badwe R, Gupta S, Dikshit R. Association of Genome-Wide Association Study (GWAS) Identified SNPs and Risk of Breast Cancer in an Indian Population. Sci Rep 2017; 7:40963. [PMID: 28098224 PMCID: PMC5241870 DOI: 10.1038/srep40963] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/05/2016] [Indexed: 01/24/2023] Open
Abstract
To date, no studies have investigated the association of the GWAS-identified SNPs with BC risk in Indian population. We investigated the association of 30 previously reported and replicated BC susceptibility SNPs in 1,204 cases and 1,212 controls from a hospital based case-control study conducted at the Tata Memorial Hospital, Mumbai. As a measure of total susceptibility burden, the polygenic risk score (PRS) for each individual was defined by the weighted sum of genotypes from 21 independent SNPs with weights derived from previously published estimates of association odds-ratios. Logistic regression models were used to assess risk associated with individual SNPs and overall PRS, and stratified by menopausal and receptor status. A total of 11 SNPs from eight genomic regions (FGFR2, 9q31.2, MAP3K, CCND1, ZM1Z1, RAD51L11, ESR1 and UST) showed statistically significant (p-value ≤ 0.05) evidence of association, either overall or when stratified by menopausal status or hormone receptor status. BC SNPs previously identified in Caucasian population showed evidence of replication in the Indian population mainly with respect to risk of postmenopausal and hormone receptor positive BC.
Collapse
Affiliation(s)
- Rajini Nagrani
- Centre for Cancer Epidemiology, Tata Memorial Centre, Mumbai, India
| | - Sharayu Mhatre
- Centre for Cancer Epidemiology, Tata Memorial Centre, Mumbai, India
| | - Preetha Rajaraman
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, USA
| | - Nilanjan Chatterjee
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, USA
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, USA
- Department of Oncology, School of Medicine, Johns Hopkins University, USA
| | - Mohammad R. Akbari
- Women’s College Research Institute, Women’s College Hospital, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Paolo Boffetta
- Institute For Translational Epidemiology, Mount Sinai Hospital, One Gustave L.Levy Place New York, NY, USA
| | - Paul Brennan
- Genetic Epidemiology Group, International Agency for Research on Cancer, 150 Cours Albert Thomas, 69372 Lyon CEDEX, France
| | - Rajendra Badwe
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Sudeep Gupta
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Rajesh Dikshit
- Centre for Cancer Epidemiology, Tata Memorial Centre, Mumbai, India
| |
Collapse
|
41
|
Dubois J, Adolphs R. Building a Science of Individual Differences from fMRI. Trends Cogn Sci 2016; 20:425-443. [PMID: 27138646 DOI: 10.1016/j.tics.2016.03.014] [Citation(s) in RCA: 404] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/28/2016] [Accepted: 03/31/2016] [Indexed: 11/19/2022]
Abstract
To date, fMRI research has been concerned primarily with evincing generic principles of brain function through averaging data from multiple subjects. Given rapid developments in both hardware and analysis tools, the field is now poised to study fMRI-derived measures in individual subjects, and to relate these to psychological traits or genetic variations. We discuss issues of validity, reliability and statistical assessment that arise when the focus shifts to individual subjects and that are applicable also to other imaging modalities. We emphasize that individual assessment of neural function with fMRI presents specific challenges and necessitates careful consideration of anatomical and vascular between-subject variability as well as sources of within-subject variability.
Collapse
Affiliation(s)
- Julien Dubois
- Division of the Humanities and Social Sciences, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Ralph Adolphs
- Division of the Humanities and Social Sciences, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
42
|
Ortega MS, Denicol AC, Cole JB, Null DJ, Hansen PJ. Use of single nucleotide polymorphisms in candidate genes associated with daughter pregnancy rate for prediction of genetic merit for reproduction in Holstein cows. Anim Genet 2016; 47:288-97. [DOI: 10.1111/age.12420] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2015] [Indexed: 12/20/2022]
Affiliation(s)
- M. S. Ortega
- Department of Animal Sciences; D.H. Barron Reproductive and Perinatal Biology Research Program and Genetics Institute; University of Florida; Gainesville FL USA
| | - A. C. Denicol
- Department of Animal Sciences; D.H. Barron Reproductive and Perinatal Biology Research Program and Genetics Institute; University of Florida; Gainesville FL USA
| | - J. B. Cole
- Animal Genomics and Improvement Laboratory; Agricultural Research Service; United States Department of Agriculture; Beltsville MD USA
| | - D. J. Null
- Animal Genomics and Improvement Laboratory; Agricultural Research Service; United States Department of Agriculture; Beltsville MD USA
| | - P. J. Hansen
- Department of Animal Sciences; D.H. Barron Reproductive and Perinatal Biology Research Program and Genetics Institute; University of Florida; Gainesville FL USA
| |
Collapse
|
43
|
Endler L, Betancourt AJ, Nolte V, Schlötterer C. Reconciling Differences in Pool-GWAS Between Populations: A Case Study of Female Abdominal Pigmentation in Drosophila melanogaster. Genetics 2016; 202:843-55. [PMID: 26715669 PMCID: PMC4788253 DOI: 10.1534/genetics.115.183376] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/21/2015] [Indexed: 12/16/2022] Open
Abstract
The degree of concordance between populations in the genetic architecture of a given trait is an important issue in medical and evolutionary genetics. Here, we address this problem, using a replicated pooled genome-wide association study approach (Pool-GWAS) to compare the genetic basis of variation in abdominal pigmentation in female European and South African Drosophila melanogaster. We find that, in both the European and the South African flies, variants near the tan and bric-à-brac 1 (bab1) genes are most strongly associated with pigmentation. However, the relative contribution of these loci differs: in the European populations, tan outranks bab1, while the converse is true for the South African flies. Using simulations, we show that this result can be explained parsimoniously, without invoking different causal variants between the populations, by a combination of frequency differences between the two populations and dominance for the causal alleles at the bab1 locus. Our results demonstrate the power of cost-effective, replicated Pool-GWAS to shed light on differences in the genetic architecture of a given trait between populations.
Collapse
Affiliation(s)
- Lukas Endler
- Institut für Populationsgenetik, Vetmeduni Vienna, 1210 Vienna, Austria
| | | | - Viola Nolte
- Institut für Populationsgenetik, Vetmeduni Vienna, 1210 Vienna, Austria
| | | |
Collapse
|
44
|
Munn-Chernoff MA, Baker JH. A Primer on the Genetics of Comorbid Eating Disorders and Substance Use Disorders. EUROPEAN EATING DISORDERS REVIEW 2015; 24:91-100. [PMID: 26663753 DOI: 10.1002/erv.2424] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 11/12/2015] [Accepted: 11/13/2015] [Indexed: 01/05/2023]
Abstract
Eating disorders (EDs) and substance use disorders (SUDs) frequently co-occur; however, the reasons for this are unclear. We review the current literature on genetic risk for EDs and SUDs, as well as preliminary findings exploring whether these classes of disorders have overlapping genetic risk. Overall, genetic factors contribute to individual differences in liability to multiple EDs and SUDs. Although initial family studies concluded that no shared familial (which includes genetic) risk between EDs and SUDs exists, twin studies suggest a moderate proportion of shared variance is attributable to overlapping genetic factors, particularly for those EDs characterized by binge eating and/or inappropriate compensatory behaviours. No adoption or molecular genetic studies have examined shared genetic risk between these classes of disorders. Research investigating binge eating and inappropriate compensatory behaviours using emerging statistical genetic methods, as well as examining gene-environment interplay, will provide important clues into the aetiology of comorbid EDs and SUDs.
Collapse
Affiliation(s)
| | - Jessica H Baker
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
45
|
Brandys MK, de Kovel CGF, Kas MJ, van Elburg AA, Adan RAH. Overview of genetic research in anorexia nervosa: The past, the present and the future. Int J Eat Disord 2015; 48:814-25. [PMID: 26171770 DOI: 10.1002/eat.22400] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/05/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND Even though the evidence supporting the presence of a heritable component in the aetiology of anorexia nervosa (AN) is strong, the underlying genetic mechanisms remain poorly understood. The recent publication of a genome-wide association study (GWAS) of AN (Boraska, Mol Psychiatry, 2014) was an important step in genetic research in AN. OBJECTIVE To briefly sum up strengths and weaknesses of candidate-gene and genome-wide approaches, to discuss the genome-wide association studies of AN and to make predictions about the genetic architecture of AN by comparing it to that of schizophrenia (since the diseases share some similarities and genetic research in schizophrenia is more advanced). METHOD Descriptive literature review. RESULTS Despite remarkable efforts, the gene-association studies in AN did not advance our knowledge as much as had been hoped, although some results still await replication. DISCUSSION Continuous effort of participants, clinicians and researchers remains necessary to ensure that genetic research in AN follows a similarly successful path as in schizophrenia. Identification of genetic susceptibility loci provides a basis for follow-up studies.
Collapse
Affiliation(s)
- Marek K Brandys
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Utrecht Research Group for Eating Disorders, Utrecht, The Netherlands
| | - Carolien G F de Kovel
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martien J Kas
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Utrecht Research Group for Eating Disorders, Utrecht, The Netherlands
| | - Annemarie A van Elburg
- Utrecht Research Group for Eating Disorders, Utrecht, The Netherlands.,Department Clinical and Health Psychology, Fac. of Social Sciences, University of Utrecht, Utrecht, The Netherlands.,Rintveld, Center for Eating Disorders, Altrecht Mental Health Institute, Zeist, The Netherlands
| | - Roger A H Adan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Utrecht Research Group for Eating Disorders, Utrecht, The Netherlands.,Rintveld, Center for Eating Disorders, Altrecht Mental Health Institute, Zeist, The Netherlands
| |
Collapse
|
46
|
Association of the glucokinase gene promoter polymorphism -30G > A (rs1799884) with gestational diabetes mellitus susceptibility: a case-control study and meta-analysis. Arch Gynecol Obstet 2015; 292:291-8. [PMID: 25633883 DOI: 10.1007/s00404-015-3635-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 01/22/2015] [Indexed: 12/23/2022]
Abstract
PURPOSE Several studies have examined the association between glucokinase (GCK)-30G > A polymorphism and gestational diabetes mellitus (GDM). However, the results are still controversial. We performed the case-control study to investigate whether GCK-30G > A polymorphism correlates with the susceptibility of GDM in Chinese populations, and then conducted a meta-analysis by combining the previous studies. METHODS We recruited 948 GDM patients and 975 controls from May 2011 to August 2013. All the subjects were genotyped using the PCR-based invader assay. The differences of allelic frequencies and genotype distributions between GDM patients and controls were investigated in case-control study. A systematic search of all relevant studies was conducted. The observational studies that were related to an association between the glucokinase (GCK)-30G > A polymorphism and GDM were identified. The association between the glucokinase (GCK)-30G > A polymorphism and GDM susceptibility was assessed using genetic models. RESULTS The case-control study showed that GCK-30G > A polymorphism was associated with the susceptibility of GDM in a Chinese population. Furthermore, other six previously reported studies were included to perform meta-analysis. The meta-analysis showed that GCK-30G > A polymorphism was associated with GDM in Caucasian and Asian. CONCLUSIONS This study suggested that GCK-30G > A polymorphism may be associated with the susceptibility of GDM in a Chinese population. The further meta-analysis provides additional evidence supporting the above result that the risk allele of the GCK-30G > A polymorphism may increase GDM risk.
Collapse
|
47
|
Gorlov IP, Moore JH, Peng B, Jin JL, Gorlova OY, Amos CI. SNP characteristics predict replication success in association studies. Hum Genet 2014; 133:1477-86. [PMID: 25273843 PMCID: PMC4384517 DOI: 10.1007/s00439-014-1493-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/25/2014] [Indexed: 02/03/2023]
Abstract
Successful independent replication is the most direct approach for distinguishing real genotype-disease associations from false discoveries in genome-wide association studies (GWAS). Selecting SNPs for replication has been primarily based on P values from the discovery stage, although additional characteristics of SNPs may be used to improve replication success. We used disease-associated SNPs from more than 2,000 published GWASs to identify predictors of SNP reproducibility. SNP reproducibility was defined as a proportion of successful replications among all replication attempts. The study reporting association for the first time was considered to be discovery and all consequent studies targeting the same phenotype replications. We found that -Log(P), where P is a P value from the discovery study, is the strongest predictor of the SNP reproducibility. Other significant predictors include type of the SNP (e.g., missense vs intronic SNPs) and minor allele frequency. Features of the genes linked to the disease-associated SNP also predict SNP reproducibility. Based on empirically defined rules, we developed a reproducibility score (RS) to predict SNP reproducibility independently of -Log(P). We used data from two lung cancer GWAS studies as well as recently reported disease-associated SNPs to validate RS. Minus Log(P) outperforms RS when the very top SNPs are selected, while RS works better with relaxed selection criteria. In conclusion, we propose an empirical model to predict SNP reproducibility, which can be used to select SNPs for validation and prioritization.
Collapse
Affiliation(s)
- Ivan P Gorlov
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, 74 College Street Vail 7th Floor, HB 7260 Vail, Hanover, NH, 03755, USA,
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Intracranial aneurysms, also called cerebral aneurysms, are dilatations in the arteries that supply blood to the brain. Rupture of an intracranial aneurysm leads to a subarachnoid hemorrhage, which is fatal in about 50% of the cases. Intracranial aneurysms can be repaired surgically or endovascularly, or by combining these two treatment modalities. They are relatively common with an estimated prevalence of unruptured aneurysms of 2%-6% in the adult population, and are considered a complex disease with both genetic and environmental risk factors. Known risk factors include smoking, hypertension, increasing age, and positive family history for intracranial aneurysms. Identifying the molecular mechanisms underlying the pathogenesis of intracranial aneurysms is complex. Genome-wide approaches such as DNA linkage and genetic association studies, as well as microarray-based mRNA expression studies, provide unbiased approaches to identify genetic risk factors and dissecting the molecular pathobiology of intracranial aneurysms. The ultimate goal of these studies is to use the information in clinical practice to predict an individual's risk for developing an aneurysm or monitor its growth or rupture risk. Another important goal is to design new therapies based on the information on mechanisms of disease processes to prevent the development or halt the progression of intracranial aneurysms.
Collapse
Affiliation(s)
- Gerard Tromp
- The Sigfried and Janet Weis Center for Research, Geisinger Health System , Danville, Pennsylvania , USA
| | | | | | | |
Collapse
|
49
|
Guo Q, Lv SZ, Wu SW, Tian X, Li ZY. Association between single nucleotide polymorphism of IL15RA gene with susceptibility to ossification of the posterior longitudinal ligament of the spine. J Orthop Surg Res 2014; 9:103. [PMID: 25387549 PMCID: PMC4232694 DOI: 10.1186/s13018-014-0103-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 10/14/2014] [Indexed: 11/10/2022] Open
Abstract
Background Previous studies reported the association between single nucleotide polymorphism (SNP) of IL15 receptor alpha (IL15RA) gene with susceptibility to ossification of the posterior longitudinal ligament of the spine (OPLL). However, the results were still in controversy. Therefore, the purpose of the present study was to investigate the association between SNPs of IL15RA gene with susceptibility to OPLL in a Chinese Han population. Methods A total of 235 OPLL patients and 250 age-matched healthy controls were recruited. All the subjects were genotyped using the PCR (polymerase chain reaction)-based invader assay. A case–control study was performed to define the contribution of rs2228059 and rs2296139 to predisposition of OPLL. We also performed subgroup analysis according to the different gender. Results A significant association of rs2228059 with OPLL was observed in the Chinese Han population (p <0.001, OR = 1.63, 95% CI = 1.26–2.11). The subgroup analysis showed that there was a significant association between the allele frequency of rs2228059 and the susceptibility of OPLL in males (p = 0.002, OR = 1.72, 95% CI = 1.23–2.42). However, there was no significant association between SNP of rs2296139 and susceptibility to OPLL. Conclusions The present study demonstrates that the SNP of rs2228059 in IL15RA gene is associated with susceptibility to OPLL in a Chinese Han population, especially in males.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Spinal Surgery, Tianjin Baodi Hospital, No 8 Guangchuan Street, Baodi District, Tianjin, 301800, China.
| | - Shou-Zheng Lv
- Department of Spinal Surgery, Tianjin Baodi Hospital, No 8 Guangchuan Street, Baodi District, Tianjin, 301800, China.
| | - Shu-Wen Wu
- Department of Spinal Surgery, Tianjin Baodi Hospital, No 8 Guangchuan Street, Baodi District, Tianjin, 301800, China.
| | - Xu Tian
- Department of Orthopaedics Institute, Tianjin Hospital, 406 Jiefang Nan Street, Hexi District, Tianjin, 300211, China.
| | - Zhi-Yong Li
- Department of Spinal Surgery, Tianjin Baodi Hospital, No 8 Guangchuan Street, Baodi District, Tianjin, 301800, China.
| |
Collapse
|
50
|
Herdegen S, Holmes G, Cyriac A, Calin-Jageman IE, Calin-Jageman RJ. Characterization of the rapid transcriptional response to long-term sensitization training in Aplysia californica. Neurobiol Learn Mem 2014; 116:27-35. [PMID: 25117657 DOI: 10.1016/j.nlm.2014.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/17/2014] [Accepted: 07/24/2014] [Indexed: 11/24/2022]
Abstract
We used a custom-designed microarray and quantitative PCR to characterize the rapid transcriptional response to long-term sensitization training in the marine mollusk Aplysia californica. Aplysia were exposed to repeated noxious shocks to one side of the body, a procedure known to induce a long-lasting, transcription-dependent increase in reflex responsiveness that is restricted to the side of training. One hour after training, pleural ganglia from the trained and untrained sides of the body were harvested; these ganglia contain the sensory nociceptors which help mediate the expression of long-term sensitization memory. Microarray analysis from 8 biological replicates suggests that long-term sensitization training rapidly regulates at least 81 transcripts. We used qPCR to test a subset of these transcripts and found that 83% were confirmed in the same samples, and 86% of these were again confirmed in an independent sample. Thus, our new microarray design shows strong convergent and predictive validity for analyzing the transcriptional correlates of memory in Aplysia. Fully validated transcripts include some previously identified as regulated in this paradigm (ApC/EBP and ApEgr) but also include novel findings. Specifically, we show that long-term sensitization training rapidly up-regulates the expression of transcripts which may encode Aplysia homologs of a C/EBPγ transcription factor, a glycine transporter (GlyT2), and a vacuolar-protein-sorting-associated protein (VPS36).
Collapse
Affiliation(s)
- Samantha Herdegen
- Neuroscience Program, Dominican University, River Forest, IL, United States
| | - Geraldine Holmes
- Neuroscience Program, Dominican University, River Forest, IL, United States
| | - Ashly Cyriac
- Neuroscience Program, Dominican University, River Forest, IL, United States
| | | | | |
Collapse
|