401
|
Hu M, Tomlinson B. Gender-dependent associations of uric acid levels with a polymorphism inSLC2A9in Han Chinese patients. Scand J Rheumatol 2012; 41:161-3. [DOI: 10.3109/03009742.2011.637952] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
402
|
Hui JY, Choi JWJ, Mount DB, Zhu Y, Zhang Y, Choi HK. The independent association between parathyroid hormone levels and hyperuricemia: a national population study. Arthritis Res Ther 2012; 14:R56. [PMID: 22405053 PMCID: PMC3446422 DOI: 10.1186/ar3769] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 02/08/2012] [Accepted: 03/10/2012] [Indexed: 11/12/2022] Open
Abstract
Introduction Increased frequencies of hyperuricemia and gout have been associated with primary hyperparathyroidism, and recent clinical trials of parathyroid hormone (PTH) have reported hyperuricemic adverse events. We evaluated the potential population impact of PTH on serum uric acid (SUA) levels by using a nationally representative sample of United States adults. Methods By using data from 8,316 participants aged 18 years and older in the National Health and Nutrition Examination Survey 2003 to 2006, we examined the relation between serum PTH and SUA levels with weighted linear regression. Additionally, we examined the relation with hyperuricemia by using weighted logistic regression. Results SUA levels increased with increasing serum PTH concentration. After adjusting for age, sex, dietary factors, glomerular filtration rate (GFR), and other potentially related biomarkers (calcium, phosphorus, alkaline-phosphatase, 25-hydroxyvitamin D), the SUA level differences from the bottom (referent) to top quintiles of serum PTH levels were 0, 8, 13, 14, and 19 μM (95% CI, 12 to 26; P for trend, < 0.001). These estimates were larger among renally impaired individuals (multivariate SUA difference between the extreme quintiles of PTH, 26 versus 15 μM among those with GFR ≥ 60 versus < 60 ml/min per 1.73 m2, respectively) (P for interaction = 0.004). The odds of hyperuricemia by various definitions increased with increasing PTH levels as well (multivariate P values for trend, < 0.05). Conclusions These nationally representative data indicate that serum PTH levels are independently associated with serum uric acid levels and the frequency of hyperuricemia at the population level.
Collapse
Affiliation(s)
- Janet Y Hui
- The Clinical Epidemiology Unit, Boston University School of Medicine, 650 Albany Street, Suite 200, Boston, MA 02118, USA
| | | | | | | | | | | |
Collapse
|
403
|
Qayyum R, Snively BM, Ziv E, Nalls MA, Liu Y, Tang W, Yanek LR, Lange L, Evans MK, Ganesh S, Austin MA, Lettre G, Becker DM, Zonderman AB, Singleton AB, Harris TB, Mohler ER, Logsdon BA, Kooperberg C, Folsom AR, Wilson JG, Becker LC, Reiner AP. A meta-analysis and genome-wide association study of platelet count and mean platelet volume in african americans. PLoS Genet 2012; 8:e1002491. [PMID: 22423221 PMCID: PMC3299192 DOI: 10.1371/journal.pgen.1002491] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 12/05/2011] [Indexed: 12/24/2022] Open
Abstract
Several genetic variants associated with platelet count and mean platelet volume (MPV) were recently reported in people of European ancestry. In this meta-analysis of 7 genome-wide association studies (GWAS) enrolling African Americans, our aim was to identify novel genetic variants associated with platelet count and MPV. For all cohorts, GWAS analysis was performed using additive models after adjusting for age, sex, and population stratification. For both platelet phenotypes, meta-analyses were conducted using inverse-variance weighted fixed-effect models. Platelet aggregation assays in whole blood were performed in the participants of the GeneSTAR cohort. Genetic variants in ten independent regions were associated with platelet count (N = 16,388) with p<5×10(-8) of which 5 have not been associated with platelet count in previous GWAS. The novel genetic variants associated with platelet count were in the following regions (the most significant SNP, closest gene, and p-value): 6p22 (rs12526480, LRRC16A, p = 9.1×10(-9)), 7q11 (rs13236689, CD36, p = 2.8×10(-9)), 10q21 (rs7896518, JMJD1C, p = 2.3×10(-12)), 11q13 (rs477895, BAD, p = 4.9×10(-8)), and 20q13 (rs151361, SLMO2, p = 9.4×10(-9)). Three of these loci (10q21, 11q13, and 20q13) were replicated in European Americans (N = 14,909) and one (11q13) in Hispanic Americans (N = 3,462). For MPV (N = 4,531), genetic variants in 3 regions were significant at p<5×10(-8), two of which were also associated with platelet count. Previously reported regions that were also significant in this study were 6p21, 6q23, 7q22, 12q24, and 19p13 for platelet count and 7q22, 17q11, and 19p13 for MPV. The most significant SNP in 1 region was also associated with ADP-induced maximal platelet aggregation in whole blood (12q24). Thus through a meta-analysis of GWAS enrolling African Americans, we have identified 5 novel regions associated with platelet count of which 3 were replicated in other ethnic groups. In addition, we also found one region associated with platelet aggregation that may play a potential role in atherothrombosis.
Collapse
Affiliation(s)
- Rehan Qayyum
- GeneSTAR Research Program, Division of General
Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United
States of America
| | - Beverly M. Snively
- Department of Biostatistical Sciences, Wake
Forest School of Medicine, Winston-Salem, North Carolina, United States of
America
| | - Elad Ziv
- Department of Medicine, University of
California San Francisco, San Francisco, California, United States of
America
| | - Michael A. Nalls
- Laboratory of Neurogenetics, National
Institute on Aging, National Institutes of Health, Bethesda, Maryland, United
States of America
| | - Yongmei Liu
- Department of Epidemiology and Prevention,
Division of Public Health Sciences, Wake Forest University School of Medicine,
Winston-Salem, North Carolina, United States of America
| | - Weihong Tang
- Division of Epidemiology and Community Health,
University of Minnesota School of Public Health, Minneapolis, Minnesota, United
States of America
| | - Lisa R. Yanek
- GeneSTAR Research Program, Division of General
Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United
States of America
| | - Leslie Lange
- Department of Genetics, School of Medicine,
The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina,
United States of America
| | - Michele K. Evans
- Health Disparities Research Section, Clinical
Research Branch, National Institute on Aging, National Institutes of Health,
Baltimore, Maryland, United States of America
| | - Santhi Ganesh
- Division of Cardiology, University of Michigan
Health System, Ann Arbor, Michigan, United States of America
| | - Melissa A. Austin
- Department of Epidemiology, University of
Washington, Seattle, Washington, United States of America
- Division of Public Health Sciences, Fred
Hutchinson Cancer Research Center, Seattle, Washington, United States of
America
| | | | - Diane M. Becker
- GeneSTAR Research Program, Division of General
Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United
States of America
| | - Alan B. Zonderman
- Laboratory of Personality and Cognition,
National Institute on Aging, National Institutes of Health, Baltimore, Maryland,
United States of America
| | - Andrew B. Singleton
- Laboratory of Neurogenetics, National
Institute on Aging, National Institutes of Health, Bethesda, Maryland, United
States of America
| | - Tamara B. Harris
- Laboratory for Epidemiology, Demography, and
Biometry, National Institute on Aging, National Institutes of Health, Baltimore,
Maryland, United States of America
| | - Emile R. Mohler
- Department of Medicine, University of
Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of
America
| | - Benjamin A. Logsdon
- Program in Biostatistics and Biomathematics,
Division of Public Health Sciences, Fred Hutchinson Cancer Research Center,
Seattle, Washington, United States of America
| | - Charles Kooperberg
- Program in Biostatistics and Biomathematics,
Division of Public Health Sciences, Fred Hutchinson Cancer Research Center,
Seattle, Washington, United States of America
| | - Aaron R. Folsom
- Division of Epidemiology and Community Health,
University of Minnesota School of Public Health, Minneapolis, Minnesota, United
States of America
| | - James G. Wilson
- Department of Medicine, University of
Mississippi Medical Center, Jackson, Mississippi, United States of
America
| | - Lewis C. Becker
- GeneSTAR Research Program, Division of General
Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United
States of America
| | - Alexander P. Reiner
- Department of Epidemiology, University of
Washington, Seattle, Washington, United States of America
| |
Collapse
|
404
|
Association between gout and polymorphisms in GCKR in male Han Chinese. Hum Genet 2012; 131:1261-5. [DOI: 10.1007/s00439-012-1151-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 02/15/2012] [Indexed: 01/17/2023]
|
405
|
Liu LY, Schaub MA, Sirota M, Butte AJ. Sex differences in disease risk from reported genome-wide association study findings. Hum Genet 2012; 131:353-64. [PMID: 21858542 PMCID: PMC3260375 DOI: 10.1007/s00439-011-1081-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/08/2011] [Indexed: 01/01/2023]
Abstract
Men and women differ in susceptibility to many diseases and in responses to treatment. Recent advances in genome-wide association studies (GWAS) provide a wealth of data for associating genetic profiles with disease risk; however, in general, these data have not been systematically probed for sex differences in gene-disease associations. Incorporating sex into the analysis of GWAS results can elucidate new relationships between single nucleotide polymorphisms (SNPs) and human disease. In this study, we performed a sex-differentiated analysis on significant SNPs from GWAS data of the seven common diseases studied by the Wellcome Trust Case Control Consortium. We employed and compared three methods: logistic regression, Woolf's test of heterogeneity, and a novel statistical metric that we developed called permutation method to assess sex effects (PMASE). After correction for false discovery, PMASE finds SNPs that are significantly associated with disease in only one sex. These sexually dimorphic SNP-disease associations occur in Coronary Artery Disease and Crohn's Disease. GWAS analyses that fail to consider sex-specific effects may miss discovering sexual dimorphism in SNP-disease associations that give new insights into differences in disease mechanism between men and women.
Collapse
Affiliation(s)
- Linda Y. Liu
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 1265 Welch Road, MS-5415 Room X-163, Stanford, CA 94305-5415, USA
- Lucile Packard Children’s Hospital, 725 Welch Road, Palo Alto, CA 94304, USA
| | - Marc A. Schaub
- Computer Science Department, Stanford University, 353 Serra Mall, Stanford, CA 94305, USA,
| | - Marina Sirota
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 1265 Welch Road, MS-5415 Room X-163, Stanford, CA 94305-5415, USA
- Lucile Packard Children’s Hospital, 725 Welch Road, Palo Alto, CA 94304, USA
| | - Atul J. Butte
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 1265 Welch Road, MS-5415 Room X-163, Stanford, CA 94305-5415, USA,
- Lucile Packard Children’s Hospital, 725 Welch Road, Palo Alto, CA 94304, USA
| |
Collapse
|
406
|
Abstract
Gout is an inflammatory arthritis caused by monosodium urate (MSU) crystal deposits in and around the joint. The formation of urinary calculi can also occur in gout, but are less common than arthritis. Gout usually presents with recurrent episodes of joint inflammation, which over time lead to tophus formation and joint destruction. In the last decade, significant advances have been made regarding not only the epidemiology and genetics of gout and hyperuricemia but also the mechanisms of inflammation and treatment of gout. In addition, knowledge concerning the key role of interleukin 1 (IL-1) has provided new therapeutic perspectives. However, the current management of gout is often suboptimal, with many Patienten either not receiving adequate treatment or being unable to tolerate existing treatments. New therapeutic agents provide interesting new options for Patienten with difficult-to-treat gouty arthritis.The English full-text version of this is available at SpringerLink (under "Supplemental").
Collapse
|
407
|
Li C, Chu N, Wang B, Wang J, Luan J, Han L, Meng D, Wang Y, Suo P, Cheng L, Ma X, Miao Z, Liu S. Polymorphisms in the presumptive promoter region of the SLC2A9 gene are associated with gout in a Chinese male population. PLoS One 2012; 7:e24561. [PMID: 22393348 PMCID: PMC3290627 DOI: 10.1371/journal.pone.0024561] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 08/15/2011] [Indexed: 01/05/2023] Open
Abstract
Background Glucose transporter 9 (GLUT9) is a high-capacity/low-affinity urate transporter. To date, several recent genome-wide association studies (GWAS) and follow-up studies have identified genetic variants of SLC2A9 associated with urate concentrations and susceptibility to gout. We therefore investigated associations between gout and polymorphisms and haplotypes in the presumptive promoter region of GLUT9 in Chinese males. Methodology/Principal Findings The approximately 2000 bp presumptive promoter region upstream of the start site of exon 1 of GLUT9 was sequenced and subjected to genetic analysis. A genotype-phenotype correlation was performed and polymorphisms-induced changes in transcription factor binding sites were predicted. Of 21 SNPs identified in GLUT9, five had not been previously reported. Two of the SNPs (rs13124007 and rs6850166) were associated with susceptibility to gout (p = 0.009 and p = 0.042, respectively). The C allele of rs13124007 appeared to be the risk allele for predisposition to gout (p = 0.006, OR 1.709 [95% CI 1.162–2.514]). For rs6850166, an increased risk of gout was associated with the A allele (p = 0.029, OR 1.645 [95% CI 1.050–2.577]). After Bonferroni correction, there was statistically difference in rs13124007 allele frequencies between gout cases and controls (P = 0.042). Haplotype analyses showed that haplotype GG was a protective haplotype (p = 0.0053) and haplotype CA was associated with increased risk of gout (p = 0.0326). Genotype-phenotype analysis among gout patients revealed an association of rs13124007 with serum triglycerides levels (P = 0.001). The C to G substitution in polymorphism rs13124007 resulted in a loss of a binding site for transcription factor interferon regulatory factor 1 (IRF-1). Conclusions/Significance Polymorphisms rs13124007 and rs6850166 are associated with susceptibility to gout in Chinese males.
Collapse
Affiliation(s)
- Changgui Li
- The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
- * E-mail: (CL); (SL)
| | - Nan Chu
- The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Binbin Wang
- Graduate school, Peking Union Medical College, Beijing, China
- National Research Institute for Family Planning, Beijing, China
| | - Jing Wang
- Graduate school, Peking Union Medical College, Beijing, China
- National Research Institute for Family Planning, Beijing, China
| | - Jian Luan
- Qingdao Municipal Hospital, Qingdao, China
| | - Lin Han
- The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Dongmei Meng
- The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Yunlong Wang
- The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Peisu Suo
- Graduate school, Peking Union Medical College, Beijing, China
- National Research Institute for Family Planning, Beijing, China
| | - Longfei Cheng
- Graduate school, Peking Union Medical College, Beijing, China
- National Research Institute for Family Planning, Beijing, China
| | - Xu Ma
- Graduate school, Peking Union Medical College, Beijing, China
- National Research Institute for Family Planning, Beijing, China
- World Health Organization Collaborating Centre for Research in Human Reproduction, Beijing, China
| | - Zhimin Miao
- The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Shiguo Liu
- The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
- * E-mail: (CL); (SL)
| |
Collapse
|
408
|
Extra-renal elimination of uric acid via intestinal efflux transporter BCRP/ABCG2. PLoS One 2012; 7:e30456. [PMID: 22348008 PMCID: PMC3277506 DOI: 10.1371/journal.pone.0030456] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 12/20/2011] [Indexed: 02/07/2023] Open
Abstract
Urinary excretion accounts for two-thirds of total elimination of uric acid and the remainder is excreted in feces. However, the mechanism of extra-renal elimination is poorly understood. In the present study, we aimed to clarify the mechanism and the extent of elimination of uric acid through liver and intestine using oxonate-treated rats and Caco-2 cells as a model of human intestinal epithelium. In oxonate-treated rats, significant amounts of externally administered and endogenous uric acid were recovered in the intestinal lumen, while biliary excretion was minimal. Accordingly, direct intestinal secretion was thought to be a substantial contributor to extra-renal elimination of uric acid. Since human efflux transporter BCRP/ABCG2 accepts uric acid as a substrate and genetic polymorphism causing a decrease of BCRP activity is known to be associated with hyperuricemia and gout, the contribution of rBcrp to intestinal secretion was examined. rBcrp was confirmed to transport uric acid in a membrane vesicle study, and intestinal regional differences of expression of rBcrp mRNA were well correlated with uric acid secretory activity into the intestinal lumen. Bcrp1 knockout mice exhibited significantly decreased intestinal secretion and an increased plasma concentration of uric acid. Furthermore, a Bcrp inhibitor, elacridar, caused a decrease of intestinal secretion of uric acid. In Caco-2 cells, uric acid showed a polarized flux from the basolateral to apical side, and this flux was almost abolished in the presence of elacridar. These results demonstrate that BCRP contributes at least in part to the intestinal excretion of uric acid as extra-renal elimination pathway in humans and rats.
Collapse
|
409
|
Kettunen J, Tukiainen T, Sarin AP, Ortega-Alonso A, Tikkanen E, Lyytikäinen LP, Kangas AJ, Soininen P, Würtz P, Silander K, Dick DM, Rose RJ, Savolainen MJ, Viikari J, Kähönen M, Lehtimäki T, Pietiläinen KH, Inouye M, McCarthy MI, Jula A, Eriksson J, Raitakari OT, Salomaa V, Kaprio J, Järvelin MR, Peltonen L, Perola M, Freimer NB, Ala-Korpela M, Palotie A, Ripatti S. Genome-wide association study identifies multiple loci influencing human serum metabolite levels. Nat Genet 2012; 44:269-76. [PMID: 22286219 PMCID: PMC3605033 DOI: 10.1038/ng.1073] [Citation(s) in RCA: 434] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 12/13/2011] [Indexed: 12/12/2022]
Abstract
Nuclear magnetic resonance assays allow for measurement of a wide range of metabolic phenotypes. We report here the results of a GWAS on 8,330 Finnish individuals genotyped and imputed at 7.7 million SNPs for a range of 216 serum metabolic phenotypes assessed by NMR of serum samples. We identified significant associations (P < 2.31 × 10(-10)) at 31 loci, including 11 for which there have not been previous reports of associations to a metabolic trait or disorder. Analyses of Finnish twin pairs suggested that the metabolic measures reported here show higher heritability than comparable conventional metabolic phenotypes. In accordance with our expectations, SNPs at the 31 loci associated with individual metabolites account for a greater proportion of the genetic component of trait variance (up to 40%) than is typically observed for conventional serum metabolic phenotypes. The identification of such associations may provide substantial insight into cardiometabolic disorders.
Collapse
Affiliation(s)
- Johannes Kettunen
- Institute for Molecular Medicine Finland, University of Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
410
|
Saison C, Helias V, Ballif BA, Peyrard T, Puy H, Miyazaki T, Perrot S, Vayssier-Taussat M, Waldner M, Le Pennec PY, Cartron JP, Arnaud L. Null alleles of ABCG2 encoding the breast cancer resistance protein define the new blood group system Junior. Nat Genet 2012; 44:174-7. [PMID: 22246505 DOI: 10.1038/ng.1070] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 12/09/2011] [Indexed: 01/16/2023]
Abstract
The breast cancer resistance protein, also known as ABCG2, is one of the most highly studied ATP-binding cassette (ABC) transporters because of its ability to confer multidrug resistance. The lack of information on the physiological role of ABCG2 in humans severely limits cancer chemotherapeutic approaches targeting this transporter. We report here that ABCG2 comprises the molecular basis of a new blood group system (Junior, Jr) and that individuals of the Jr(a-) blood type have inherited two null alleles of ABCG2. We identified five frameshift and three nonsense mutations in ABCG2. We also show that the prevalence of the Jr(a-) blood type in the Japanese and European Gypsy populations is related to the p.Gln126* and p.Arg236* protein alterations, respectively. The identification of ABCG2(-/-) (Jr(a-)) individuals who appear phenotypically normal is an essential step toward targeting ABCG2 in cancer and also in understanding the physiological and pharmacological roles of this promiscuous transporter in humans.
Collapse
Affiliation(s)
- Carole Saison
- National Institute of Blood Transfusion (INTS), Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
411
|
Karns R, Zhang G, Sun G, Rao Indugula S, Cheng H, Havas-Augustin D, Novokmet N, Rudan D, Durakovic Z, Missoni S, Chakraborty R, Rudan P, Deka R. Genome-wide association of serum uric acid concentration: replication of sequence variants in an island population of the Adriatic coast of Croatia. Ann Hum Genet 2012; 76:121-7. [PMID: 22229870 DOI: 10.1111/j.1469-1809.2011.00698.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A genome-wide association study of serum uric acid (SUA) laevels was performed in a relatively isolated population of European descent from an island of the Adriatic coast of Croatia. The study sample included 532 unrelated and 768 related individuals from 235 pedigrees. Inflation due to relatedness was controlled by using genomic control. Genetic association was assessed with 2,241,249 single nucleotide polymorphisms (SNPs) in 1300 samples after adjusting for age and gender. Our study replicated four previously reported SUA loci (SLC2A9, ABCG2, RREB1, and SLC22A12). The strongest association was found with a SNP in SLC2A9 (rs13129697, P=2.33×10(-19)), which exhibited significant gender-specific effects, 35.76 μmol/L (P=2.11×10(-19)) in females and 19.58 μmol/L (P=5.40×10(-5)) in males. Within this region of high linkage disequilibrium, we also detected a strong association with a nonsynonymous SNP, rs16890979 (P=2.24×10(-17)), a putative causal variant for SUA variation. In addition, we identified several novel loci suggestive of association with uric acid levels (SEMA5A, TMEM18, SLC28A2, and ODZ2), although the P-values (P<5×10(-6)) did not reach the threshold of genome-wide significance. Together, these findings provide further confirmation of previously reported uric-acid-related genetic variants and highlight suggestive new loci for additional investigation.
Collapse
Affiliation(s)
- Rebekah Karns
- Center for Genome Information, Department of Environmental Health, University of Cincinnati, and Cincinnati Children's Hospital, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
412
|
Moayyeri A, Hammond CJ, Valdes AM, Spector TD. Cohort Profile: TwinsUK and healthy ageing twin study. Int J Epidemiol 2012; 42:76-85. [PMID: 22253318 DOI: 10.1093/ije/dyr207] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The UK's largest registry of adult twins, or TwinsUK Registry, started in 1992 and encompasses about 12000 volunteer twins from all over the United Kingdom. More than 70% of the registered twins have filled at least one detailed health questionnaire and about half of them undergone a baseline comprehensive assessment and two follow-up clinical evaluations. The most recent follow-up visit, known as Healthy Ageing Twin Study (HATS), involved 3125 female twins aged >40 years with at least one previous clinical assessment to enable inspection of longitudinal changes in ageing traits and their genetic and environmental components. The study benefits from several state-of-the-art OMICs studies including genome-wide association, next-generation genome and transcriptome sequencing, and epigenetic and metabolomic profiles. This makes our cohort as one of the most deeply phenotyped and genotyped in the world. Several collaborative projects in the field of epidemiology of complex disorders are ongoing in our cohort and interested researchers are encouraged to get in contact for future collaborations.
Collapse
Affiliation(s)
- Alireza Moayyeri
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, London, UK
| | | | | | | |
Collapse
|
413
|
Sull JW, Park EJ, Lee M, Jee SH. Effects of SLC2A9 variants on uric acid levels in a Korean population. Rheumatol Int 2012; 33:19-23. [DOI: 10.1007/s00296-011-2303-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 12/10/2011] [Indexed: 12/16/2022]
|
414
|
Sakurai H. Transporter-centric view of urate metabolism: From genome-wide association study to pathophysiology. JOURNAL OF PHYSICAL FITNESS AND SPORTS MEDICINE 2012. [DOI: 10.7600/jpfsm.1.413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
415
|
Adijanto J, Philp NJ. The SLC16A family of monocarboxylate transporters (MCTs)--physiology and function in cellular metabolism, pH homeostasis, and fluid transport. CURRENT TOPICS IN MEMBRANES 2012. [PMID: 23177990 DOI: 10.1016/b978-0-12-394316-3.00009-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The SLC16A family of monocarboxylate transporters (MCTs) is composed of 14 members. MCT1 through MCT4 (MCTs 1-4) are H(+)-coupled monocarboxylate transporters, MCT8 and MCT10 transport thyroid hormone and aromatic amino acids, while the substrate specificity and function of other MCTs have yet to be determined. The focus of this review is on MCTs 1-4 because their role in lactate transport is intrinsically linked to cellular metabolism in various biological systems, including skeletal muscle, brain, retina, and testis. Although MCTs 1-4 all transport lactate, they differ in their transport kinetics and vary in tissue and subcellular distribution, where they facilitate "lactate-shuttling" between glycolytic and oxidative cells within tissues and across blood-tissue barriers. However, the role of MCTs 1-4 is not confined to cellular metabolism. By interacting with bicarbonate transport proteins and carbonic anhydrases, MCTs participate in the regulation of pH homeostasis and fluid transport in renal proximal tubule and corneal endothelium, respectively. Here, we provide a comprehensive review of MCTs 1-4, linking their cellular distribution to their functions in various parts of the human body, so that we can better understand the physiological roles of MCTs at the systemic level.
Collapse
Affiliation(s)
- Jeffrey Adijanto
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| | | |
Collapse
|
416
|
Butler K, Teng R. Evaluation and Characterization of the Effects of Ticagrelor on Serum and Urinary Uric Acid in Healthy Volunteers. Clin Pharmacol Ther 2011; 91:264-71. [DOI: 10.1038/clpt.2011.223] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
417
|
Abstract
Elevated serum levels of uric acid consistently correlate with hypertension, but the directionality of the association remains debated. To help define this relationship, we used a controlled setting within a homogeneous Amish community and the Mendelian randomization of a nonsynonymous coding single-nucleotide polymorphism, rs16890979 (Val253Ile), in the SLC2A9 gene. This gene expresses the GLUT9 transporter that also transports uric acid and is associated with lower serum uric acid levels. We studied the unconfounded association between genotype and blood pressure in 516 Amish adults, each placed for 6 days on standardized diets, first with high sodium, followed by low sodium, with an intervening washout period. Blood pressure, measured using 24-h ambulatory monitoring, during both diet periods was used as the primary outcome. All participants were free of diuretic or other antihypertensive medications and the relationships between GLUT9 genotype and both serum uric acid and blood pressure were assessed. Each copy of the GLUT9 minor Ile allele was found to confer a significant 0.44 mg/dl reduction in serum uric acid and was associated with a significant mean decrease in the systolic blood pressure of 2.2 and 1.5 mm Hg on the high- and low-sodium diet, respectively. Thus, a Mendelian randomization analysis using variants in the GLUT9 gene indicates that a decrease in serum uric acid has a causal effect of lowering blood pressure.
Collapse
|
418
|
Rees MG, Ng D, Ruppert S, Turner C, Beer NL, Swift AJ, Morken MA, Below JE, Blech I, Mullikin JC, McCarthy MI, Biesecker LG, Gloyn AL, Collins FS. Correlation of rare coding variants in the gene encoding human glucokinase regulatory protein with phenotypic, cellular, and kinetic outcomes. J Clin Invest 2011; 122:205-17. [PMID: 22182842 DOI: 10.1172/jci46425] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 11/09/2011] [Indexed: 01/24/2023] Open
Abstract
Defining the genetic contribution of rare variants to common diseases is a major basic and clinical science challenge that could offer new insights into disease etiology and provide potential for directed gene- and pathway-based prevention and treatment. Common and rare nonsynonymous variants in the GCKR gene are associated with alterations in metabolic traits, most notably serum triglyceride levels. GCKR encodes glucokinase regulatory protein (GKRP), a predominantly nuclear protein that inhibits hepatic glucokinase (GCK) and plays a critical role in glucose homeostasis. The mode of action of rare GCKR variants remains unexplored. We identified 19 nonsynonymous GCKR variants among 800 individuals from the ClinSeq medical sequencing project. Excluding the previously described common missense variant p.Pro446Leu, all variants were rare in the cohort. Accordingly, we functionally characterized all variants to evaluate their potential phenotypic effects. Defects were observed for the majority of the rare variants after assessment of cellular localization, ability to interact with GCK, and kinetic activity of the encoded proteins. Comparing the individuals with functional rare variants to those without such variants showed associations with lipid phenotypes. Our findings suggest that, while nonsynonymous GCKR variants, excluding p.Pro446Leu, are rare in individuals of mixed European descent, the majority do affect protein function. In sum, this study utilizes computational, cell biological, and biochemical methods to present a model for interpreting the clinical significance of rare genetic variants in common disease.
Collapse
Affiliation(s)
- Matthew G Rees
- National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
419
|
Umeno J, Asano K, Matsushita T, Matsumoto T, Kiyohara Y, Iida M, Nakamura Y, Kamatani N, Kubo M. Meta-analysis of published studies identified eight additional common susceptibility loci for Crohn's disease and ulcerative colitis. Inflamm Bowel Dis 2011; 17:2407-15. [PMID: 21351207 DOI: 10.1002/ibd.21651] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 12/17/2010] [Indexed: 12/21/2022]
Abstract
BACKGROUND Both ulcerative colitis (UC) and Crohn's disease (CD) have a complex etiology involving multiple genetic and environmental factors. Many genome-wide association studies (GWAS) and subsequent replication studies revealed that both diseases share some of the susceptibility loci; however, common genetic factors for both diseases are not fully elucidated. This study is aimed to identify the common genetic factors for CD and UC by a meta-analysis of published studies. METHODS We first reviewed the 10 GWAS for CD to select candidate single nucleotide polymorphisms (SNPs). Next, we performed a PubMed literature search up to June 30, 2010 and carried out a systemic review of published studies that examined the association of CD susceptibility loci in UC patients. Meta-analysis was carried out using the inverse variance-weighted method or the DerSimonian-Laird method after estimating the heterogeneity among the studies. The data for highly linked SNPs were combined. Finally, we performed a meta-analysis of 43 published studies in 45 SNPs located at 33 loci by using a total of 4852 to 31,125 subjects. RESULTS We confirmed the association of 17 reported common susceptibility loci. Moreover, we found associations at eight additional loci: GCKR, ATG16L1, CDKAL1, ZNF365, LRRK2-MUC19, C13orf31, PTPN2, and SBNO2. The genetic risk of each locus was modest (odds ratios ranged from 1.05-1.22) except IL23R. CONCLUSIONS These results indicate that CD and UC share many susceptibility loci with small genetic effect. Our data provide further understanding of the common pathogenesis between CD and UC.
Collapse
Affiliation(s)
- Junji Umeno
- Laboratory for Genotyping Development, Center for Genomic Medicine, RIKEN, Yokohama Institute, Japan; Department of Medicine and Clinical Science, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
420
|
Arden C, Petrie JL, Tudhope SJ, Al-Oanzi Z, Claydon AJ, Beynon RJ, Towle HC, Agius L. Elevated glucose represses liver glucokinase and induces its regulatory protein to safeguard hepatic phosphate homeostasis. Diabetes 2011; 60:3110-20. [PMID: 22013014 PMCID: PMC3219956 DOI: 10.2337/db11-0061] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE The induction of hepatic glucose 6-phosphatase (G6pc) by glucose presents a paradox of glucose-induced glucose intolerance. We tested whether glucose regulation of liver gene expression is geared toward intracellular homeostasis. RESEARCH DESIGN AND METHODS The effect of glucose-induced accumulation of phosphorylated intermediates on expression of glucokinase (Gck) and its regulator Gckr was determined in hepatocytes. Cell ATP and uric acid production were measured as indices of cell phosphate homeostasis. RESULTS Accumulation of phosphorylated intermediates in hepatocytes incubated at elevated glucose induced rapid and inverse changes in Gck (repression) and Gckr (induction) mRNA concomitantly with induction of G6pc, but had slower effects on the Gckr-to-Gck protein ratio. Dynamic metabolic labeling in mice and liver proteome analysis confirmed that Gckr and Gck are low-turnover proteins. Involvement of Max-like protein X in glucose-mediated Gck-repression was confirmed by chromatin immunoprecipitation analysis. Elevation of the Gck-to-Gckr ratio in hepatocytes was associated with glucose-dependent ATP depletion and elevated urate production confirming compromised phosphate homeostasis. CONCLUSIONS The lowering by glucose of the Gck-to-Gckr ratio provides a potential explanation for the impaired hepatic glucose uptake in diabetes. Elevated uric acid production at an elevated Gck-to-Gckr ratio supports a role for glucose regulation of gene expression in hepatic phosphate homeostasis.
Collapse
Affiliation(s)
- Catherine Arden
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - John L. Petrie
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Susan J. Tudhope
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Ziad Al-Oanzi
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
- Department of Laboratory Medicine, Al-Jouf University, Sakaka, Saudi Arabia
| | - Amy J. Claydon
- Protein Function Group, Institute of Integrative Biology, University of Liverpool, Liverpool, U.K
| | - Robert J. Beynon
- Protein Function Group, Institute of Integrative Biology, University of Liverpool, Liverpool, U.K
| | - Howard C. Towle
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Loranne Agius
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
- Corresponding author: Loranne Agius,
| |
Collapse
|
421
|
Abstract
Drug-drug interactions are a serious clinical issue. An important mechanism underlying drug-drug interactions is induction or inhibition of drug transporters that mediate the cellular uptake and efflux of xenobiotics. Especially drug transporters of the small intestine, liver and kidney are major determinants of the pharmacokinetic profile of drugs. Transporter-mediated drug-drug interactions in these three organs can considerably influence the pharmacokinetics and clinical effects of drugs. In this article, we focus on probe drugs lacking significant metabolism to highlight mechanisms of interactions of selected intestinal, hepatic and renal drug transporters (e.g., organic anion transporting polypeptide [OATP] 1A2, OATP2B1, OATP1B1, OATP1B3, P-gp, organic anion transporter [OAT] 1, OAT3, breast cancer resistance protein [BCRP], organic cation transporter [OCT] 2 and multidrug and toxin extrusion protein [MATE] 1). Genotype-dependent drug-drug interactions are also discussed.
Collapse
Affiliation(s)
- Fabian Müller
- Institute of Experimental & Clinical Pharmacology & Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | | |
Collapse
|
422
|
Benyamin B, Middelberg RP, Lind PA, Valle AM, Gordon S, Nyholt DR, Medland SE, Henders AK, Heath AC, Madden PA, Visscher PM, O'Connor DT, Montgomery GW, Martin NG, Whitfield JB. GWAS of butyrylcholinesterase activity identifies four novel loci, independent effects within BCHE and secondary associations with metabolic risk factors. Hum Mol Genet 2011; 20:4504-14. [PMID: 21862451 PMCID: PMC3196893 DOI: 10.1093/hmg/ddr375] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 08/18/2011] [Indexed: 11/13/2022] Open
Abstract
Serum butyrylcholinesterase (BCHE) activity is associated with obesity, blood pressure and biomarkers of cardiovascular and diabetes risk. We have conducted a genome-wide association scan to discover genetic variants affecting BCHE activity, and to clarify whether the associations between BCHE activity and cardiometabolic risk factors are caused by variation in BCHE or whether BCHE variation is secondary to the metabolic abnormalities. We measured serum BCHE in adolescents and adults from three cohorts of Australian twin and family studies. The genotypes from ∼2.4 million single-nucleotide polymorphisms (SNPs) were available in 8791 participants with BCHE measurements. We detected significant associations with BCHE activity at three independent groups of SNPs at the BCHE locus (P = 5.8 × 10(-262), 7.8 × 10(-47), 2.9 × 10(-12)) and at four other loci: RNPEP (P = 9.4 × 10(-16)), RAPH1-ABI2 (P = 4.1 × 10(-18)), UGT1A1 (P = 4.0 × 10(-8)) and an intergenic region on chromosome 8 (P = 1.4 × 10(-8)). These loci affecting BCHE activity were not associated with metabolic risk factors. On the other hand, SNPs in genes previously associated with metabolic risk had effects on BCHE activity more often than can be explained by chance. In particular, SNPs within FTO and GCKR were associated with BCHE activity, but their effects were partly mediated by body mass index and triglycerides, respectively. We conclude that variation in BCHE activity is due to multiple variants across the spectrum from uncommon/large effect to common/small effect, and partly results from (rather than causes) metabolic abnormalities.
Collapse
Affiliation(s)
- Beben Benyamin
- Queensland Institute of Medical Research, Brisbane 4006, Australia
| | | | - Penelope A. Lind
- Queensland Institute of Medical Research, Brisbane 4006, Australia
| | - Anne M. Valle
- Department of Medicine, Division of Nephrology-Hypertension, University of California at San Diego, La Jolla, CA 92093, USA and
| | - Scott Gordon
- Queensland Institute of Medical Research, Brisbane 4006, Australia
| | - Dale R. Nyholt
- Queensland Institute of Medical Research, Brisbane 4006, Australia
| | - Sarah E. Medland
- Queensland Institute of Medical Research, Brisbane 4006, Australia
| | | | - Andrew C. Heath
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Pamela A.F. Madden
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | - Daniel T. O'Connor
- Department of Medicine, Division of Nephrology-Hypertension, University of California at San Diego, La Jolla, CA 92093, USA and
| | | | | | | |
Collapse
|
423
|
Abstract
Gout is a chronic arthritic disease associated with high levels of urate in blood. Recent advances in research have permitted the identification of several new and common genetic factors underlying the disease. Among them, a polymorphism in the ABC transporter gene ATP-binding cassette transporter isoform G2 has been highlighted. ATP-binding cassette transporter isoform G2 was found to be involved in renal urate elimination, and the presence of the Q141K polymorphism to induce a 2-fold decrease in urate efflux. The Q141K variant has been shown to have impaired trafficking, leading to its intracellular retention, whereas the wild type protein is expressed on the cell surface. Several agents are being studied for the purpose of improving folding, trafficking and function of various ABC transporters, including ATP-binding cassette transporter isoform G2. If successful, this strategy opens doors to potential new therapies for gout.
Collapse
Affiliation(s)
- Agnès Basseville
- Molecular therapeutics section, Medical Oncology Branch, National Cancer Institute, Building 10 - Room 13N248, National Institutes of Health 9000 Rockville Pike, Bethesda, MD 20892 USA
| | | |
Collapse
|
424
|
Anzai N, Jutabha P, Amonpatumrat-Takahashi S, Sakurai H. Recent advances in renal urate transport: characterization of candidate transporters indicated by genome-wide association studies. Clin Exp Nephrol 2011; 16:89-95. [PMID: 22038265 DOI: 10.1007/s10157-011-0532-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Accepted: 08/22/2011] [Indexed: 01/08/2023]
Abstract
Humans have higher serum uric acid levels than other mammalian species owing to the genetic silencing of the hepatic enzyme uricase that metabolizes uric acid into allantoin. Urate (the ionized form of uric acid) is generated from purine metabolism and it may provide antioxidant defense in the human body. Despite its potential advantage, sustained hyperuricemia has pathogenetic causes in gout and renal diseases, and putative roles in hypertension and cardiovascular diseases. Since the kidney plays a dominant role in maintaining plasma urate levels through the excretion process, it is important to understand the molecular mechanism of renal urate handling. Although the molecular identification of a kidney-specific urate/anion exchanger URAT1 in 2002 paved the way for successive identification of several urate transport-related proteins, the entire picture of effective renal urate handling in humans has not yet been clarified. Recently, several genome-wide association studies identified a substantial association between uric acid concentration and single nucleotide polymorphisms in at least ten genetic loci including eight transporter-coding genes. In 2008, we functionally characterized the facilitatory glucose transporter family member SLC2A9 (GLUT9), one of the candidate genes for urate handling, as a voltage-driven urate transporter URATv1 at the basolateral side of renal proximal tubules that comprises the main route of the urate reabsorption pathway, in tandem with URAT1 at the apical side. In this review, recent findings concerning these candidate molecules are presented.
Collapse
Affiliation(s)
- Naohiko Anzai
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi, 321-0293, Japan.
| | | | | | | |
Collapse
|
425
|
Shimizu T, Sugiura T, Wakayama T, Kijima A, Nakamichi N, Iseki S, Silver DL, Kato Y. PDZK1 regulates breast cancer resistance protein in small intestine. Drug Metab Dispos 2011; 39:2148-54. [PMID: 21816982 DOI: 10.1124/dmd.111.040295] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Transporter adaptor protein PDZK1 regulates several influx transporters for xenobiotics and nutrients in small intestine, and their expression on the apical membrane is diminished in pdzk1 gene knockout [pdzk1(-/-)] mice. In the present study, we initially attempted to use pdzk1(-/-) mice to functionally identify influx transporters responsible for intestinal absorption of cimetidine. Contrary to our expectation, the plasma concentration of cimetidine after oral administration to pdzk1(-/-) mice was higher than that in wild-type mice, and the double peaks of plasma concentration found in wild-type mice were not observed in pdzk1(-/-) mice. Western blot analysis of intestinal brush-border membranes revealed that expression of breast cancer resistance protein (BCRP) but not of P-glycoprotein is reduced in pdzk1(-/-) mice. This result was compatible with the reduction of apical localization of BCRP in pdzk1(-/-) mice assessed by immunohistochemical analysis. Transcellular transport of cimetidine in the basal-to-apical direction in Madin-Darby canine kidney II (MDCKII) cells stably expressing both BCRP and PDZK1 (MDCKII/BCRP/PDZK1) was higher than that in MDCKII cells stably expressing BCRP (MDCKII/BCRP) cells. Moreover, MDCKII/BCRP/PDZK1 cells are more resistant than MDCKII/BCRP cells to the cytotoxicity of the anticancer agent 7-ethyl-10-hydroxycamptothecin (SN-38), which is a substrate of BCRP. These results were consistent with the higher expression of BCRP on apical membranes in MDCKII/BCRP/PDZK1 cells. Pull-down and immunoprecipitation studies revealed a physical interaction between BCRP and PDZK1. Taken together, these findings demonstrate that PDZK1 plays a pivotal role in the apical localization of BCRP. This is the first identification of a regulatory protein that physically interacts with and regulates BCRP in small intestine in vivo.
Collapse
Affiliation(s)
- Takuya Shimizu
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
426
|
Tin A, Woodward OM, Kao WHL, Liu CT, Lu X, Nalls MA, Shriner D, Semmo M, Akylbekova EL, Wyatt SB, Hwang SJ, Yang Q, Zonderman AB, Adeyemo AA, Palmer C, Meng Y, Reilly M, Shlipak MG, Siscovick D, Evans MK, Rotimi CN, Flessner MF, Köttgen M, Cupples LA, Fox CS, Köttgen A. Genome-wide association study for serum urate concentrations and gout among African Americans identifies genomic risk loci and a novel URAT1 loss-of-function allele. Hum Mol Genet 2011; 20:4056-68. [PMID: 21768215 PMCID: PMC3177647 DOI: 10.1093/hmg/ddr307] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/25/2011] [Accepted: 07/11/2011] [Indexed: 12/20/2022] Open
Abstract
Serum urate concentrations are highly heritable and elevated serum urate is a key risk factor for gout. Genome-wide association studies (GWAS) of serum urate in African American (AA) populations are lacking. We conducted a meta-analysis of GWAS of serum urate levels and gout among 5820 AA and a large candidate gene study among 6890 AA and 21 708 participants of European ancestry (EA) within the Candidate Gene Association Resource Consortium. Findings were tested for replication among 1996 independent AA individuals, and evaluated for their association among 28 283 EA participants of the CHARGE Consortium. Functional studies were conducted using (14)C-urate transport assays in mammalian Chinese hamster ovary cells. In the discovery GWAS of serum urate, three loci achieved genome-wide significance (P< 5.0 × 10(-8)): a novel locus near SGK1/SLC2A12 on chromosome 6 (rs9321453, P= 1.0 × 10(-9)), and two loci previously identified in EA participants, SLC2A9 (P= 3.8 × 10(-32)) and SLC22A12 (P= 2.1 × 10(-10)). A novel rare non-synonymous variant of large effect size in SLC22A12, rs12800450 (minor allele frequency 0.01, G65W), was identified and replicated (beta -1.19 mg/dl, P= 2.7 × 10(-16)). (14)C-urate transport assays showed reduced urate transport for the G65W URAT1 mutant. Finally, in analyses of 11 loci previously associated with serum urate in EA individuals, 10 of 11 lead single-nucleotide polymorphisms showed direction-consistent association with urate among AA. In summary, we identified and replicated one novel locus in association with serum urate levels and experimentally characterize the novel G65W variant in URAT1 as a functional allele. Our data support the importance of multi-ethnic GWAS in the identification of novel risk loci as well as functional variants.
Collapse
Affiliation(s)
- Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Owen M. Woodward
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wen Hong Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Ave, CT3, Boston, MA 02118, USA
| | - Xiaoning Lu
- Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Ave, CT3, Boston, MA 02118, USA
| | - Michael A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, Building 12A, Room 4047, 12 South Dr., MSC 5635, Bethesda, MD 20892-5635, USA
| | - Mariam Semmo
- Renal Division, University Hospital of Freiburg, Freiburg, Germany
| | - Ermeg L. Akylbekova
- Jackson State University, 350 West Woodrow Wilson Drive, Suite 701, Jackson, MS 39213, USA
| | - Sharon B. Wyatt
- School of Nursing and
- Department of Medicine, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, USA
| | - Shih-Jen Hwang
- NHLBI's Framingham Heart Study and the Center for Population Studies, 73 Mt Wayte Ave Suite #2, Framingham, MA 01702, USA
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, 715 Albany Street, Boston, MA 02118, USA
| | - Alan B. Zonderman
- Laboratory of Personality and Cognition, NIA, Gerontology Research Center, 5600 Nathan Shock Drive MSC 6825, Baltimore, MD 21224-6825, USA
| | - Adebowale A. Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, Building 12A, Room 4047, 12 South Dr., MSC 5635, Bethesda, MD 20892-5635, USA
| | - Cameron Palmer
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Divisions of Genetics and Endocrinology and Program in Genomics, Children's Hospital Boston, Boston, MA, USA
- Metabolic Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Yan Meng
- Metabolic Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Muredach Reilly
- University of Pennsylvania Medical Center, Cardiovascular Institute, 609 BRB, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Michael G. Shlipak
- San Francisco VA Medical Center; Departments of Medicine, Epidemiology & Biostatistics, University of California, San Francisco, CA, USA
| | - David Siscovick
- Department of Epidemiology and
- Department of Medicine, University of Washington, 1730 Minor Avenure, Suite 1360, Seattle, WA 98101, USA
| | - Michele K. Evans
- Health Disparities Research Section, Clinical Research Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Charles N. Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, Building 12A, Room 4047, 12 South Dr., MSC 5635, Bethesda, MD 20892-5635, USA
| | - Michael F. Flessner
- KUH, NIDDK, NIH, 6707 Democracy Blvd, Room 641, 2 Democracy MS 5458, Bethesda, MD 20892-5458, USA and
| | - Michael Köttgen
- Renal Division, University Hospital of Freiburg, Freiburg, Germany
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Ave, CT3, Boston, MA 02118, USA
- NHLBI's Framingham Heart Study and the Center for Population Studies, 73 Mt Wayte Ave Suite #2, Framingham, MA 01702, USA
| | - Caroline S. Fox
- NHLBI's Framingham Heart Study and the Center for Population Studies, 73 Mt Wayte Ave Suite #2, Framingham, MA 01702, USA
- Brigham and Women's Hospital Department of Endocrinology and Harvard Medical School, Boston, MA, USA
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
- Renal Division, University Hospital of Freiburg, Freiburg, Germany
| |
Collapse
|
427
|
Sulem P, Gudbjartsson DF, Walters GB, Helgadottir HT, Helgason A, Gudjonsson SA, Zanon C, Besenbacher S, Bjornsdottir G, Magnusson OT, Magnusson G, Hjartarson E, Saemundsdottir J, Gylfason A, Jonasdottir A, Holm H, Karason A, Rafnar T, Stefansson H, Andreassen OA, Pedersen JH, Pack AI, de Visser MCH, Kiemeney LA, Geirsson AJ, Eyjolfsson GI, Olafsson I, Kong A, Masson G, Jonsson H, Thorsteinsdottir U, Jonsdottir I, Stefansson K. Identification of low-frequency variants associated with gout and serum uric acid levels. Nat Genet 2011; 43:1127-30. [DOI: 10.1038/ng.972] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 09/16/2011] [Indexed: 12/18/2022]
|
428
|
Pfister R, Barnes D, Luben R, Forouhi NG, Bochud M, Khaw KT, Wareham NJ, Langenberg C. No evidence for a causal link between uric acid and type 2 diabetes: a Mendelian randomisation approach. Diabetologia 2011; 54:2561-9. [PMID: 21717115 DOI: 10.1007/s00125-011-2235-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 06/03/2011] [Indexed: 01/17/2023]
Abstract
AIMS/HYPOTHESIS Epidemiological and experimental evidence suggests that uric acid has a role in the aetiology of type 2 diabetes. Using a Mendelian randomisation approach, we investigated whether there is evidence for a causal role of serum uric acid for development of type 2 diabetes. METHODS We examined the associations of serum-uric-acid-raising alleles of eight common variants recently identified in genome-wide association studies and summarised this in a genetic score with type 2 diabetes in case-control studies including 7,504 diabetes patients and 8,560 non-diabetic controls. We compared the observed effect size to that expected based on: (1) the association between the genetic score and uric acid levels in non-diabetic controls; and (2) the meta-analysed uric acid level to diabetes association. RESULTS The genetic score showed a linear association with uric acid levels, with a difference of 12.2 μmol/l (95% CI 9.3, 15.1) by score tertile. No significant associations were observed between the genetic score and potential confounders. No association was observed between the genetic score and type 2 diabetes with an OR of 0.99 (95% CI 0.94, 1.04) per score tertile, significantly different (p = 0.046) from that expected (1.04 [95% CI 1.03, 1.05]) based on the observed uric acid difference by score tertile and the uric acid to diabetes association of 1.21 (95% CI 1.14, 1.29) per 60 μmol/l. CONCLUSIONS/INTERPRETATION Our results do not support a causal role of serum uric acid for the development of type 2 diabetes and limit the expectation that uric-acid-lowering drugs will be effective in the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- R Pfister
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, Box 285, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | | | | | | | | | | | | | | |
Collapse
|
429
|
Avery CL, He Q, North KE, Ambite JL, Boerwinkle E, Fornage M, Hindorff LA, Kooperberg C, Meigs JB, Pankow JS, Pendergrass SA, Psaty BM, Ritchie MD, Rotter JI, Taylor KD, Wilkens LR, Heiss G, Lin DY. A phenomics-based strategy identifies loci on APOC1, BRAP, and PLCG1 associated with metabolic syndrome phenotype domains. PLoS Genet 2011; 7:e1002322. [PMID: 22022282 PMCID: PMC3192835 DOI: 10.1371/journal.pgen.1002322] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 08/11/2011] [Indexed: 01/11/2023] Open
Abstract
Despite evidence of the clustering of metabolic syndrome components, current approaches for identifying unifying genetic mechanisms typically evaluate clinical categories that do not provide adequate etiological information. Here, we used data from 19,486 European American and 6,287 African American Candidate Gene Association Resource Consortium participants to identify loci associated with the clustering of metabolic phenotypes. Six phenotype domains (atherogenic dyslipidemia, vascular dysfunction, vascular inflammation, pro-thrombotic state, central obesity, and elevated plasma glucose) encompassing 19 quantitative traits were examined. Principal components analysis was used to reduce the dimension of each domain such that >55% of the trait variance was represented within each domain. We then applied a statistically efficient and computational feasible multivariate approach that related eight principal components from the six domains to 250,000 imputed SNPs using an additive genetic model and including demographic covariates. In European Americans, we identified 606 genome-wide significant SNPs representing 19 loci. Many of these loci were associated with only one trait domain, were consistent with results in African Americans, and overlapped with published findings, for instance central obesity and FTO. However, our approach, which is applicable to any set of interval scale traits that is heritable and exhibits evidence of phenotypic clustering, identified three new loci in or near APOC1, BRAP, and PLCG1, which were associated with multiple phenotype domains. These pleiotropic loci may help characterize metabolic dysregulation and identify targets for intervention.
Collapse
Affiliation(s)
- Christy L Avery
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
430
|
|
431
|
Bis JC, Kavousi M, Franceschini N, Isaacs A, Abecasis GR, Schminke U, Post WS, Smith AV, Cupples LA, Markus HS, Schmidt R, Huffman JE, Lehtimäki T, Baumert J, Münzel T, Heckbert SR, Dehghan A, North K, Oostra B, Bevan S, Stoegerer EM, Hayward C, Raitakari O, Meisinger C, Schillert A, Sanna S, Völzke H, Cheng YC, Thorsson B, Fox CS, Rice K, Rivadeneira F, Nambi V, Halperin E, Petrovic KE, Peltonen L, Wichmann HE, Schnabel RB, Dörr M, Parsa A, Aspelund T, Demissie S, Kathiresan S, Reilly MP, Taylor K, Uitterlinden A, Couper DJ, Sitzer M, Kähönen M, Illig T, Wild PS, Orru M, Lüdemann J, Shuldiner AR, Eiriksdottir G, White CC, Rotter JI, Hofman A, Seissler J, Zeller T, Usala G, Ernst F, Launer LJ, D'Agostino RB, O'Leary DH, Ballantyne C, Thiery J, Ziegler A, Lakatta EG, Chilukoti RK, Harris TB, Wolf PA, Psaty BM, Polak JF, Li X, Rathmann W, Uda M, Boerwinkle E, Klopp N, Schmidt H, Wilson JF, Viikari J, Koenig W, Blankenberg S, Newman AB, Witteman J, Heiss G, Duijn CV, Scuteri A, Homuth G, Mitchell BD, Gudnason V, O'Donnell CJ. Meta-analysis of genome-wide association studies from the CHARGE consortium identifies common variants associated with carotid intima media thickness and plaque. Nat Genet 2011; 43:940-7. [PMID: 21909108 PMCID: PMC3257519 DOI: 10.1038/ng.920] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 08/02/2011] [Indexed: 01/17/2023]
Abstract
Carotid intima media thickness (cIMT) and plaque determined by ultrasonography are established measures of subclinical atherosclerosis that each predicts future cardiovascular disease events. We conducted a meta-analysis of genome-wide association data in 31,211 participants of European ancestry from nine large studies in the setting of the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium. We then sought additional evidence to support our findings among 11,273 individuals using data from seven additional studies. In the combined meta-analysis, we identified three genomic regions associated with common carotid intima media thickness and two different regions associated with the presence of carotid plaque (P < 5 × 10(-8)). The associated SNPs mapped in or near genes related to cellular signaling, lipid metabolism and blood pressure homeostasis, and two of the regions were associated with coronary artery disease (P < 0.006) in the Coronary Artery Disease Genome-Wide Replication and Meta-Analysis (CARDIoGRAM) consortium. Our findings may provide new insight into pathways leading to subclinical atherosclerosis and subsequent cardiovascular events.
Collapse
Affiliation(s)
- Joshua C Bis
- Cardiovascular Health Research Unit and Department of Medicine, University of Washington, Seattle, Washington, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
432
|
Large-scale genome-wide association studies in east Asians identify new genetic loci influencing metabolic traits. Nat Genet 2011; 43:990-5. [DOI: 10.1038/ng.939] [Citation(s) in RCA: 226] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 08/18/2011] [Indexed: 12/13/2022]
|
433
|
Huffman JE, Knežević A, Vitart V, Kattla J, Adamczyk B, Novokmet M, Igl W, Pučić M, Zgaga L, Johannson Å, Redžić I, Gornik O, Zemunik T, Polašek O, Kolčić I, Pehlić M, Koeleman CA, Campbell S, Wild SH, Hastie ND, Campbell H, Gyllensten U, Wuhrer M, Wilson JF, Hayward C, Rudan I, Rudd PM, Wright AF, Lauc G. Polymorphisms in B3GAT1, SLC9A9 and MGAT5 are associated with variation within the human plasma N-glycome of 3533 European adults. Hum Mol Genet 2011; 20:5000-11. [DOI: 10.1093/hmg/ddr414] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
434
|
Abstract
ATP-binding cassette (ABC) transporters form a large family of transmembrane proteins that facilitate the transport of specific substrates across membranes in an ATP-dependent manner. Transported substrates include lipids, lipopolysaccharides, amino acids, peptides, proteins, inorganic ions, sugars and xenobiotics. Despite this broad array of substrates, the physiological substrate of many ABC transporters has remained elusive. ABC transporters are divided into seven subfamilies, A-G, based on sequence similarity and domain organization. Here we review the role of members of the ABCG subfamily in human disease and how the identification of disease genes helped to determine physiological substrates for specific ABC transporters. We focus on the recent discovery of mutations in ABCG2 causing hyperuricemia and gout, which has led to the identification of urate as a physiological substrate for ABCG2.
Collapse
Affiliation(s)
- Owen M Woodward
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
435
|
|
436
|
Suhre K, Shin SY, Petersen AK, Mohney RP, Meredith D, Wägele B, Altmaier E, CARDIoGRAM, Deloukas P, Erdmann J, Grundberg E, Hammond CJ, de Angelis MH, Kastenmüller G, Köttgen A, Kronenberg F, Mangino M, Meisinger C, Meitinger T, Mewes HW, Milburn MV, Prehn C, Raffler J, Ried JS, Römisch-Margl W, Samani NJ, Small KS, Wichmann HE, Zhai G, Illig T, Spector TD, Adamski J, Soranzo N, Gieger C. Human metabolic individuality in biomedical and pharmaceutical research. Nature 2011; 477:54-60. [PMID: 21886157 PMCID: PMC3832838 DOI: 10.1038/nature10354] [Citation(s) in RCA: 834] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 06/30/2011] [Indexed: 01/08/2023]
Abstract
Genome-wide association studies (GWAS) have identified many risk loci for complex diseases, but effect sizes are typically small and information on the underlying biological processes is often lacking. Associations with metabolic traits as functional intermediates can overcome these problems and potentially inform individualized therapy. Here we report a comprehensive analysis of genotype-dependent metabolic phenotypes using a GWAS with non-targeted metabolomics. We identified 37 genetic loci associated with blood metabolite concentrations, of which 25 show effect sizes that are unusually high for GWAS and account for 10-60% differences in metabolite levels per allele copy. Our associations provide new functional insights for many disease-related associations that have been reported in previous studies, including those for cardiovascular and kidney disorders, type 2 diabetes, cancer, gout, venous thromboembolism and Crohn's disease. The study advances our knowledge of the genetic basis of metabolic individuality in humans and generates many new hypotheses for biomedical and pharmaceutical research.
Collapse
Affiliation(s)
- Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Faculty of Biology, Ludwig-Maximilians-Universität, Planegg-Martinsried, Germany
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Education City - Qatar Foundation, Doha, Qatar
| | - So-Youn Shin
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton UK
| | - Ann-Kristin Petersen
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | - David Meredith
- School of Life Sciences, Oxford Brookes University, Headington, Oxford, UK
| | - Brigitte Wägele
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Genome-oriented Bioinformatics, Life and Food Science Center Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Elisabeth Altmaier
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - CARDIoGRAM
- The member list of the CARDIoGRAM consortium is provided as Supplemental Information
| | - Panos Deloukas
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton UK
| | | | - Elin Grundberg
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton UK
- Department of Twin Research & Genetic Epidemiology, King’s College London, UK
| | | | - Martin Hrabé de Angelis
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Life and Food Science Center Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Anna Köttgen
- Renal Division, University Hospital Freiburg, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King’s College London, UK
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Hans-Werner Mewes
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Genome-oriented Bioinformatics, Life and Food Science Center Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | | | - Cornelia Prehn
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Faculty of Biology, Ludwig-Maximilians-Universität, Planegg-Martinsried, Germany
| | - Janina S. Ried
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton UK
| | - Werner Römisch-Margl
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, and Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, UK
| | - Kerrin S. Small
- Department of Twin Research & Genetic Epidemiology, King’s College London, UK
| | - H.-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Klinikum Grosshadern, Munich, Germany
| | - Guangju Zhai
- Department of Twin Research & Genetic Epidemiology, King’s College London, UK
| | - Thomas Illig
- Unit for Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Tim D. Spector
- Department of Twin Research & Genetic Epidemiology, King’s College London, UK
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Nicole Soranzo
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton UK
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
437
|
Castorino JJ, Gallagher-Colombo SM, Levin AV, Fitzgerald PG, Polishook J, Kloeckener-Gruissem B, Ostertag E, Philp NJ. Juvenile cataract-associated mutation of solute carrier SLC16A12 impairs trafficking of the protein to the plasma membrane. Invest Ophthalmol Vis Sci 2011; 52:6774-84. [PMID: 21778275 DOI: 10.1167/iovs.10-6579] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE SLC16A12 encodes an orphan member of the monocarboxylate transporter family, MCT12. A nonsense mutation in SLC16A12 (c.643C>T; p.Q215X) causes juvenile cataract with a dominant inheritance pattern. In the present study, in vitro and in vivo experimental models were used to gain insight into how the SLC16A12 (c.643C>T) mutation leads to cataract formation. METHODS MCT12 peptide antibodies were generated and used to examine the expression of MCT12 in the lens using immuno-confocal microscopy. To determine whether loss of Slc16a12 resulted in cataract formation, a Slc16a12 hypomorphic rat generated by transposon insertional mutagenesis was characterized using RT-PCR, slit lamp microscopy and histologic methods. Exogenous expression of MCT12 and MCT12:214Δ, a mimic of the mutant allele, were used to assess protein expression and trafficking. RESULTS MCT12 protein was detected in the lens epithelium and secondary fiber cells at postnatal day 1. In the Slc16a12(TKO) rat, complete loss of MCT12 did not result in any detectable ocular phenotype. Exogenous expression of MCT12-GFP and MCT12:214Δ-GFP revealed that the full-length protein was trafficked to the plasma membrane (PM), whereas the truncated protein was retained in the endoplasmic reticulum (ER). When both MCT12 and MCT12:214Δ were coexpressed, to mimic the heterozygous patient genotype, the truncated protein was retained in the ER whereas full-length MCT12 was trafficked to the PM. Furthermore, MCT12 was identified as another MCT isoform that requires CD147 for trafficking to the cell surface. CONCLUSIONS These data support a model whereby the SLC16A12 (c.643C>T) mutation causes juvenile cataract by a defect in protein trafficking rather than by haploinsufficiency.
Collapse
Affiliation(s)
- John J Castorino
- Department of Pathology, Anatomy, and Cell Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
438
|
Wei W, Hemani G, Hicks AA, Vitart V, Cabrera-Cardenas C, Navarro P, Huffman J, Hayward C, Knott SA, Rudan I, Pramstaller PP, Wild SH, Wilson JF, Campbell H, Dunlop MG, Hastie N, Wright AF, Haley CS. Characterisation of genome-wide association epistasis signals for serum uric acid in human population isolates. PLoS One 2011; 6:e23836. [PMID: 21886828 PMCID: PMC3158795 DOI: 10.1371/journal.pone.0023836] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 07/26/2011] [Indexed: 01/05/2023] Open
Abstract
Genome-wide association (GWA) studies have identified a number of loci underlying variation in human serum uric acid (SUA) levels with the SLC2A9 gene having the largest effect identified so far. Gene-gene interactions (epistasis) are largely unexplored in these GWA studies. We performed a full pair-wise genome scan in the Italian MICROS population (n = 1201) to characterise epistasis signals in SUA levels. In the resultant epistasis profile, no SNP pairs reached the Bonferroni adjusted threshold for the pair-wise genome-wide significance. However, SLC2A9 was found interacting with multiple loci across the genome, with NFIA-SLC2A9 and SLC2A9-ESRRAP2 being significant based on a threshold derived for interactions between GWA significant SNPs and the genome and jointly explaining 8.0% of the phenotypic variance in SUA levels (3.4% by interaction components). Epistasis signal replication in a CROATIAN population (n = 1772) was limited at the SNP level but improved dramatically at the gene ontology level. In addition, gene ontology terms enriched by the epistasis signals in each population support links between SUA levels and neurological disorders. We conclude that GWA epistasis analysis is useful despite relatively low power in small isolated populations.
Collapse
Affiliation(s)
- Wenhua Wei
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Gibran Hemani
- The Roslin Institute and R(D)SVS, University of Edinburgh, Roslin, Midlothian, United Kingdom
| | - Andrew A. Hicks
- Institute of Genetic Medicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Claudia Cabrera-Cardenas
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Jennifer Huffman
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Sara A. Knott
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Igor Rudan
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Croatian Centre for Global Health, University of Split, Split, Croatia
| | - Peter P. Pramstaller
- Institute of Genetic Medicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany
- Department of Neurology, General Central Hospital, Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Sarah H. Wild
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - James F. Wilson
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Harry Campbell
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Malcolm G. Dunlop
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Nicholas Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Chris S. Haley
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
- The Roslin Institute and R(D)SVS, University of Edinburgh, Roslin, Midlothian, United Kingdom
| |
Collapse
|
439
|
Dinour D, Gray NK, Ganon L, Knox AJS, Shalev H, Sela BA, Campbell S, Sawyer L, Shu X, Valsamidou E, Landau D, Wright AF, Holtzman EJ. Two novel homozygous SLC2A9 mutations cause renal hypouricemia type 2. Nephrol Dial Transplant 2011; 27:1035-41. [DOI: 10.1093/ndt/gfr419] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
440
|
Hamajima N, Okada R, Kawai S, Hishida A, Morita E, Yin G, Wakai K, Matsuo H, Inoue H, Takada Y, Asai Y, Mori A, Naito M. Significant association of serum uric acid levels with SLC2A9 rs11722228 among a Japanese population. Mol Genet Metab 2011; 103:378-82. [PMID: 21511506 DOI: 10.1016/j.ymgme.2011.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 04/03/2011] [Accepted: 04/03/2011] [Indexed: 12/13/2022]
Abstract
Genome-wide association studies identified that SLC2A9 (GLUT9) gene polymorphisms were associated with serum uric acid (SUA) levels. Among the Japanese, a C/T polymorphism in intron 8 (rs11722228) was reported to be highly significant, though the function and strength of association were unknown. This study aimed to confirm the association, estimating the means of SUA according to the genotype, as well as OR of the genotype. Subjects were 5024 health checkup examinees (3413 males and 1611 females) aged 35 to 69 years with creatinine <2.0 mg/dL. Since SLC22A12 258X allele and ABCG2 126X allele are known to influence SUA levels strongly, the subjects with SLC22A12 258WW and ABCG2 126QQ (3082 males and 1453 females, in total 4535 subjects) were selected. The genotype frequency of SLC2A9 rs11722228 was 2184 for CC, 1947 for CT, and 404 for TT, being in Hardy-Weinberg equilibrium (p=0.312). Mean SUA was 6.10 mg/dL for CC, 6.25 mg/dL for CT, and 6.45 mg/dL for TT among males (p=1.5E-6), and 4.34 mg/dL, 4.59 mg/dL, and 4.87 mg/dL among females (p=4.6E-11), respectively. Males with SUA less than 5.0 mg/dL were 14.7% for CC, 10.6% for CT, and 7.8% for TT (p=2.3E-4), and females with SUA less than 4.0 mg/dL were 34.1%, 25.5%, and 15.4% (p=3.7E-6), respectively. This study was the first report to estimate the impact of SLC2A9 rs11722228 on SUA levels. Since the allele frequency of rs11722228 is similar among different ethnic groups, the impact remains to be examined in other ethnic groups.
Collapse
Affiliation(s)
- Nobuyuki Hamajima
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
441
|
Bardin T, Richette P. [The epidemiology and genetic of gout]. Presse Med 2011; 40:830-5. [PMID: 21752574 DOI: 10.1016/j.lpm.2011.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 04/03/2011] [Accepted: 04/07/2011] [Indexed: 02/06/2023] Open
Abstract
Gout affects 1-2% of adults in developed countries, where it is the most common inflammatory arthritis in men. Epidemiological data are consistent with a rise in the prevalence of gout. Diet and genetic predisposition seem to be the main causal factors of primary gout. Gout and hyperuricaemia are associated with hypertension, diabetes mellitus, metabolic syndrome, and renal and cardiovascular diseases. Genetic studies have found that variants of GLUT9 and ABCG2 were associated with urate levels.
Collapse
Affiliation(s)
- Thomas Bardin
- Université Paris 7, Assistance-publique-Hôpitaux de Paris, hôpital Lariboisière, UFR médicale, 2, rue Ambroise-Paré, 75475 Paris cedex 10, France.
| | | |
Collapse
|
442
|
Yu Z, Kastenmüller G, He Y, Belcredi P, Möller G, Prehn C, Mendes J, Wahl S, Roemisch-Margl W, Ceglarek U, Polonikov A, Dahmen N, Prokisch H, Xie L, Li Y, Wichmann HE, Peters A, Kronenberg F, Suhre K, Adamski J, Illig T, Wang-Sattler R. Differences between human plasma and serum metabolite profiles. PLoS One 2011; 6:e21230. [PMID: 21760889 PMCID: PMC3132215 DOI: 10.1371/journal.pone.0021230] [Citation(s) in RCA: 343] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 05/24/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Human plasma and serum are widely used matrices in clinical and biological studies. However, different collecting procedures and the coagulation cascade influence concentrations of both proteins and metabolites in these matrices. The effects on metabolite concentration profiles have not been fully characterized. METHODOLOGY/PRINCIPAL FINDINGS We analyzed the concentrations of 163 metabolites in plasma and serum samples collected simultaneously from 377 fasting individuals. To ensure data quality, 41 metabolites with low measurement stability were excluded from further analysis. In addition, plasma and corresponding serum samples from 83 individuals were re-measured in the same plates and mean correlation coefficients (r) of all metabolites between the duplicates were 0.83 and 0.80 in plasma and serum, respectively, indicating significantly better stability of plasma compared to serum (p = 0.01). Metabolite profiles from plasma and serum were clearly distinct with 104 metabolites showing significantly higher concentrations in serum. In particular, 9 metabolites showed relative concentration differences larger than 20%. Despite differences in absolute concentration between the two matrices, for most metabolites the overall correlation was high (mean r = 0.81±0.10), which reflects a proportional change in concentration. Furthermore, when two groups of individuals with different phenotypes were compared with each other using both matrices, more metabolites with significantly different concentrations could be identified in serum than in plasma. For example, when 51 type 2 diabetes (T2D) patients were compared with 326 non-T2D individuals, 15 more significantly different metabolites were found in serum, in addition to the 25 common to both matrices. CONCLUSIONS/SIGNIFICANCE Our study shows that reproducibility was good in both plasma and serum, and better in plasma. Furthermore, as long as the same blood preparation procedure is used, either matrix should generate similar results in clinical and biological studies. The higher metabolite concentrations in serum, however, make it possible to provide more sensitive results in biomarker detection.
Collapse
Affiliation(s)
- Zhonghao Yu
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ying He
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Shanghai Center for Bioinformation Technology, Shanghai, China
- Bioinformatics Center, Key Lab of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Petra Belcredi
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Gabriele Möller
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Cornelia Prehn
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Joaquim Mendes
- Solid State NMR Spectroscopy and Center for Integrated Protein Science, Department Chemie, Technische Universität München, Garching, Germany
- Institute of Structural Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Werner Roemisch-Margl
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
| | - Norbert Dahmen
- Department for Psychiatry, University of Mainz, Mainz, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Lu Xie
- Shanghai Center for Bioinformation Technology, Shanghai, China
| | - Yixue Li
- Shanghai Center for Bioinformation Technology, Shanghai, China
- Bioinformatics Center, Key Lab of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - H. -Erich Wichmann
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Faculty of Biology, Ludwig-Maximilians-Universität, Planegg-Martinsried, Germany
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Education City - Qatar Foundation, Doha, Qatar
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Munich, Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Rui Wang-Sattler
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
443
|
Serum Urate as a Soluble Biomarker in Chronic Gout—Evidence that Serum Urate Fulfills the OMERACT Validation Criteria for Soluble Biomarkers. Semin Arthritis Rheum 2011; 40:483-500. [DOI: 10.1016/j.semarthrit.2010.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 09/23/2010] [Accepted: 09/24/2010] [Indexed: 02/07/2023]
|
444
|
Abstract
Many forms of cardiovascular disease (CVD) demonstrate heritability and thus a genetic contribution is likely. This is most evident when considering the "simple" Mendelian traits such as hypertrophic cardiomyopathy. However, family history also influences our assessment of patients with complex traits such as coronary artery disease, hypertension, and common forms of hypercholesterolemia, as observed in clinical practice. Recent research has led to advances in our understanding of the genetic basis of both the simple and complex forms of CVD. This review presents the current state of knowledge regarding major gene disorders, as well as more common, complex forms of CVD such as coronary artery disease. It discusses the fundamental approaches being used to identify the genetic basis of the various disease states, as well as the practical implications of the discoveries to clinicians. It also focuses on our need to assess the extent by which genetic analysis can alter our calculation of an individual's risk of disease, and our ability to successfully target treatment that will modify this process.
Collapse
|
445
|
Hyperuricemia cosegregating with osteogenesis imperfecta is associated with a mutation in GPATCH8. Hum Genet 2011; 130:671-83. [DOI: 10.1007/s00439-011-1006-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 05/09/2011] [Indexed: 10/18/2022]
|
446
|
Nakagawa H, Toyoda Y, Wakabayashi-Nakao K, Tamaki H, Osumi M, Ishikawa T. Ubiquitin-mediated proteasomal degradation of ABC transporters: a new aspect of genetic polymorphisms and clinical impacts. J Pharm Sci 2011; 100:3602-19. [PMID: 21567408 DOI: 10.1002/jps.22615] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/11/2011] [Accepted: 04/20/2011] [Indexed: 01/11/2023]
Abstract
The interindividual variation in the rate of drug metabolism and disposition has been known for many years. Pharmacogenomics dealing with heredity and response to drugs is a part of science that attempts to explain variability of drug responses and to search for the genetic basis of such variations or differences. Genetic polymorphisms of drug metabolizing enzymes and drug transporters have been found to play a significant role in the patients' responses to medication. Accumulating evidence demonstrates that certain nonsynonymous polymorphisms have great impacts on the protein stability and degradation, as well as the function of drug metabolizing enzymes and transporters. The aim of this review article is to address a new aspect of protein quality control in the endoplasmic reticulum and to present examples regarding the impact of nonsynonymous single-nucleotide polymorphisms on the protein stability of thiopurine S-methyltransferase as well as ATP-binding cassette (ABC) transporters including ABCC4, cystic fibrosis transmembrane conductance regulator (CFTR, ABCC7), ABCC11, and ABCG2. Furthermore, we will discuss the molecular mechanisms underlying posttranslational modifications (intramolecular and intermolecular disulfide bond formation and N-linked glycosylation) and ubiquitin-mediated proteasomal degradation of ABCG2, one of the major drug transporter proteins in humans.
Collapse
Affiliation(s)
- Hiroshi Nakagawa
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | | | | | | | | | | |
Collapse
|
447
|
Wu W, Dnyanmote AV, Nigam SK. Remote communication through solute carriers and ATP binding cassette drug transporter pathways: an update on the remote sensing and signaling hypothesis. Mol Pharmacol 2011; 79:795-805. [PMID: 21325265 PMCID: PMC3082935 DOI: 10.1124/mol.110.070607] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 02/11/2011] [Indexed: 01/12/2023] Open
Abstract
Recent data from knockouts, human disease, and transport studies suggest that solute carrier (SLC) and ATP binding cassette (ABC) multispecific "drug" transporters maintain effective organ and body fluid concentrations of key nutrients, signaling molecules, and antioxidants. These processes involve transcellular movement of solutes across epithelial barriers and fluid compartments (e.g., blood, cerebrospinal fluid, urine, bile) via "matching" or homologous sets of SLC (e.g., SLC21, SLC22, SLC47) and ABC transporters. As described in the "Remote Sensing and Signaling Hypothesis" (Biochem Biophys Res Commun 323:429-436, 2004; Biochem Biophys Res Commun 351:872-876, 2006; J Biol Chem 282:23841-23853, 2007; Nat Clin Pract Nephrol 3:443-448, 2007; Mol Pharmacol 76:481-490, 2009), highly regulated transporter networks with overlapping substrate preferences are involved in sensing and signaling to maintain homeostasis in response to environmental changes (e.g., substrate imbalance and injury). They function in parallel with (and interact with) the endocrine and autonomic systems. Uric acid (urate), carnitine, prostaglandins, conjugated sex steroids, cGMP, odorants, and enterobiome metabolites are discussed here as examples. Xenobiotics hitchhike on endogenous carrier systems, sometimes leading to toxicity and side effects. By regulation of the expression and/or function of various remote organ multispecific transporters after injury, the overall transport capacity of the remote organ to handle endogenous toxins, metabolites, and signaling molecules may change, aiding in recovery. Moreover, these transporters may play a role in communication between organisms. The specific cellular components involved in sensing and altering transporter abundance or functionality depend upon the metabolite in question and probably involve different types of sensors as well as epigenetic regulation.
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
448
|
Sugiura T, Shimizu T, Kijima A, Minakata S, Kato Y. PDZ adaptors: their regulation of epithelial transporters and involvement in human diseases. J Pharm Sci 2011; 100:3620-35. [PMID: 21538352 DOI: 10.1002/jps.22575] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Revised: 03/28/2011] [Accepted: 03/31/2011] [Indexed: 12/17/2022]
Abstract
Homeostasis in the body is at least partially maintained by mechanisms that control membrane permeability, and thereby serve to control the uptake of essential substances (e.g., nutrients) and the efflux of unwanted substances (e.g., xenobiotics and metabolites) in epithelial cells. Various transporters play fundamental roles in such bidirectional transport, but little is known about how they are organized on plasma membranes. Protein-protein interactions may play a key role: several transporters in epithelial cells interact with the so-called adaptor proteins, which are membrane anchored and interact with both transporters and other membranous proteins. Although most of the evidences for transporter-adaptor interaction has been obtained in vitro, recent studies suggest that adaptor-mediated transporter regulation does occur in vivo and could be relevant to human diseases. Thus, protein-protein interaction is not only associated with the formation of macromolecular complexes but is also involved in various cellular events, and may provide transporters with additional functionality by forming transporter networks on plasma membranes. Interactions between xenobiotic transporters and PSD95/Dlg/ZO1 (PDZ) adaptors were previously reviewed by Kato and Tsuji (2006. Eur J Pharm Sci 27:487-500); the present review focuses on the latest findings about PDZ adaptors as regulators of transporter networks and their potential role in human diseases.
Collapse
Affiliation(s)
- Tomoko Sugiura
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | | | | | | | | |
Collapse
|
449
|
The rs1014290 polymorphism of the SLC2A9 gene is associated with type 2 diabetes mellitus in Han Chinese. EXPERIMENTAL DIABETES RESEARCH 2011; 2011:527520. [PMID: 21584282 PMCID: PMC3092507 DOI: 10.1155/2011/527520] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 02/22/2011] [Accepted: 02/23/2011] [Indexed: 11/18/2022]
Abstract
Aims. The SLC2A9 gene encodes the glucose transporter 9, with the abilities of transporting both glucose and uric acid and is involved in the pancreatic glucose-stimulated insulin secretion. The single nucleotide polymorphisms (SNPs) of SLC2A9 accounted for 5% variance of serum uric acid (UA). UA was identified as a risk factor for type 2 diabetes mellitus (DM). We investigated whether the SLC2A9 gene variations are associated with type 2 DM in Han Chinese.
Methods. Three common SNPs of the SLC2A9, rs1014290, rs2280205, and rs3733591, were genotyped in 1003 Han Chinese randomly selected from Kaohsiung, Taiwan.
Results. The variant SNP rs1014290 is associated with decreased 0.12-fold risk of type 2 DM (P = .002). Per-copy increase in the minor C-allele results in 0.13 mmol/L (P = .037) and 10.03 μmol/L (P = .016) decrease in serum glucose and UA, respectively.
Conclusions. The SNP rs1014290 within the SLC2A9 gene is associated with type 2 DM in Han Chinese.
Collapse
|
450
|
Robey RW, Ierano C, Zhan Z, Bates SE. The challenge of exploiting ABCG2 in the clinic. Curr Pharm Biotechnol 2011; 12:595-608. [PMID: 21118093 PMCID: PMC3091815 DOI: 10.2174/138920111795163913] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 04/15/2010] [Indexed: 01/16/2023]
Abstract
ABCG2, or breast cancer resistance protein (BCRP), is an ATP-binding cassette half transporter that has been shown to transport a wide range of substrates including chemotherapeutics, antivirals, antibiotics and flavonoids. Given its wide range of substrates, much work has been dedicated to developing ABCG2 as a clinical target. But where can we intervene clinically and how can we avoid the mistakes made in past clinical trials targeting P-glycoprotein? This review will summarize the normal tissue distribution, cancer tissue expression, substrates and inhibitors of ABCG2, and highlight the challenges presented in exploiting ABCG2 in the clinic. We discuss the possibility of inhibiting ABCG2, so as to increase oral bioavailability or increase drug penetration into sanctuary sites, especially the central nervous system; and at the other end of the spectrum, the possibility of improving ABCG2 function, in the case of gout caused by a single nucleotide polymphism. Together, these aspects of ABCG2/BCRP make the protein a target of continuing interest for oncologists, biologists, and pharmacologists.
Collapse
Affiliation(s)
- Robert W Robey
- Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|