501
|
Genetic background and transplantation outcomes: insights from genome-wide association studies. Curr Opin Organ Transplant 2020; 25:35-41. [PMID: 31815792 DOI: 10.1097/mot.0000000000000718] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The current review summarizes recent advances in the genetic studies of transplantation outcomes, including new genome-wide association studies for acute rejection, allograft survival, pharmacogenomics, and common transplant comorbidities. RECENT FINDINGS Genetic studies of kidney transplantation outcomes have begun to address the question of genetic compatibility beyond human leukocyte antigens, including the role of genome-wide mismatches in missense variants, and the 'genomic collision' hypothesis under which the risk of rejection may be increased in recipients homozygous for loss-of-function variants with grafts from nonhomozygous donors. In recent pilot studies, missense mismatch scores for transmembrane and secreted proteins were associated with antibodies against the mismatched peptides and reduced allograft survival. A 'genomic collision' at the LIMS1 locus involving a common deletion near LIMS1 gene was associated with anti-LIMS1 antibody response and increased risk of rejection. Additional genetic factors under active investigation include genome-wide polygenic risk scores for renal function and apolipoprotein L1 risk genotypes in African-American kidney donors. Due to the heterogeneity and complexity of clinical outcomes, new genome-wide association studies for rejection, allograft survival, and specific transplant comorbidities will require larger multicenter meta-analyses. SUMMARY Genetic compatibilities between donor and recipient represent an important determinant of rejection and long-term allograft survival. Genetic background of transplant donors may be additionally predictive of allograft function, while recipient's genomes are likely determinant of a wide range of transplantation outcomes, from rejection susceptibility to pharmacogenetics and various comorbidities related to prolonged immunosuppression.
Collapse
|
502
|
Causal associations between urinary sodium with body mass, shape and composition: a Mendelian randomization study. Sci Rep 2020; 10:17475. [PMID: 33060734 PMCID: PMC7562909 DOI: 10.1038/s41598-020-74657-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/06/2020] [Indexed: 12/26/2022] Open
Abstract
Observational studies have found associations between urinary sodium (UNa) with obesity, body shape and composition; but the findings may be biased by residual confounding. The objective of this two-sample Mendelian randomization (MR) study was to analyze their causal associations in both sex-combined and sex-specific models. Genome-wide association studies of UNa, body mass index (BMI), BMI-adjusted waist-to-hip ratio (WHR), body fat (BF) percentage and estimated glomerular filtration rate (eGFR) were identified. We initially extracted fifty SNPs associated with UNa at significance level of 5 × 10–8, but further removed those SNPs with potential horizontal pleiotropy. Univariable and multivariable MR with adjustment for eGFR were performed. Inverse-variance weighted MR was performed as the primary analysis, with MR-Egger methods as sensitivity analysis. The potential bidirectional association between BMI and UNa was investigated. All exposure and outcomes were continuous, and the effect measure was regression coefficients (beta) and their 95% confidence intervals (95% CI). The total sample size was up to 322 154. UNa was causally associated with increased BMI in both men [eGFR-adjusted beta 0.443 (0.163–0.724)] and women [0.594 (0.333–0.855)]. UNa caused BF percentage increase in men [0.622 (0.268–0.976)] and women [0.334 (0.007–0.662)]. UNa significantly elevated BMI-adjusted WHR in men [0.321 (0.094–0.548)], but not in women [0.170 (− 0.052 to 0.391)]. Additionally, we found that BMI causally increased UNa [0.043 (0.023–0.063)]. UNa increased BMI and BF percentage. Salt intake affects male body shape by increasing BMI-adjusted WHR, but showed no effects on female body shape. The bidirectional association between BMI and UNa suggested that salt reduction measures and weight reduction measures should be implemented simultaneously to break the vicious cycle and gain more health benefits.
Collapse
|
503
|
Park S, Lee S, Kim Y, Lee Y, Kang MW, Kim K, Kim YC, Han SS, Lee H, Lee JP, Joo KW, Lim CS, Kim YS, Kim DK. Short or Long Sleep Duration and CKD: A Mendelian Randomization Study. J Am Soc Nephrol 2020; 31:2937-2947. [PMID: 33004418 DOI: 10.1681/asn.2020050666] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/17/2020] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Studies have found sleeping behaviors, such as sleep duration, to be associated with kidney function and cardiovascular disease risk. However, whether short or long sleep duration is a causative factor for kidney function impairment has been rarely studied. METHODS We studied data from participants aged 40-69 years in the UK Biobank prospective cohort, including 25,605 self-reporting short-duration sleep (<6 hours per 24 hours), 404,550 reporting intermediate-duration sleep (6-8 hours), and 35,659 reporting long-duration sleep (≥9 hours) in the clinical analysis. Using logistic regression analysis, we investigated the observational association between the sleep duration group and prevalent CKD stages 3-5, analyzed by logistic regression analysis. We performed Mendelian randomization (MR) analysis involving 321,260 White British individuals using genetic instruments (genetic variants linked with short- or long-duration sleep behavior as instrumental variables). We performed genetic risk score analysis as a one-sample MR and extended the finding with a two-sample MR analysis with CKD outcome information from the independent CKDGen Consortium genome-wide association study meta-analysis. RESULTS Short or long sleep duration clinically associated with higher prevalence of CKD compared with intermediate duration. The genetic risk score for short (but not long) sleep was significantly related to CKD (per unit reflecting a two-fold increase in the odds of the phenotype; adjusted odds ratio, 1.80; 95% confidence interval, 1.25 to 2.60). Two-sample MR analysis demonstrated causal effects of short sleep duration on CKD by the inverse variance weighted method, supported by causal estimates from MR-Egger regression. CONCLUSIONS These findings support an adverse effect of a short sleep duration on kidney function. Clinicians may encourage patients to avoid short-duration sleeping behavior to reduce CKD risk.
Collapse
Affiliation(s)
- Sehoon Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Armed Forces Capital Hospital, Gyeonggi-do, Korea
| | - Soojin Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yaerim Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Yeonhee Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Min Woo Kang
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kwangsoo Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yon Su Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea .,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea .,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
504
|
Abstract
PURPOSE OF REVIEW We summarize recent evidence on the shared genetics within and outside the musculoskeletal system (mostly related to bone density and osteoporosis). RECENT FINDINGS Osteoporosis is determined by an interplay between multiple genetic and environmental factors. Significant progress has been made regarding its genetic background revealing a number of robustly validated loci and respective pathways. However, pleiotropic factors affecting bone and other tissues are not well understood. The analytical methods proposed to test for potential associations between genetic variants and multiple phenotypes can be applied to bone-related data. A number of recent genetic studies have shown evidence of pleiotropy between bone density and other different phenotypes (traits, conditions, or diseases), within and outside the musculoskeletal system. Power benefits of combining correlated phenotypes, as well as unbiased discovery, make these studies promising. Studies in humans are supported by evidence from animal models. Drug development and repurposing should benefit from the pleiotropic approach. We believe that future studies should take into account shared genetics between the bone and related traits.
Collapse
Affiliation(s)
- M A Christou
- Clinical and Molecular Epidemiology Unit, Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, Ioannina, Greece
| | - E E Ntzani
- Clinical and Molecular Epidemiology Unit, Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, Ioannina, Greece
- Center for Research Synthesis in Health, Department of Health Services, Policy and Practice, School of Public Health, Brown University, Providence, RI, USA
| | - D Karasik
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA.
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel.
| |
Collapse
|
505
|
Nakamura R, Misawa K, Tohnai G, Nakatochi M, Furuhashi S, Atsuta N, Hayashi N, Yokoi D, Watanabe H, Watanabe H, Katsuno M, Izumi Y, Kanai K, Hattori N, Morita M, Taniguchi A, Kano O, Oda M, Shibuya K, Kuwabara S, Suzuki N, Aoki M, Ohta Y, Yamashita T, Abe K, Hashimoto R, Aiba I, Okamoto K, Mizoguchi K, Hasegawa K, Okada Y, Ishihara T, Onodera O, Nakashima K, Kaji R, Kamatani Y, Ikegawa S, Momozawa Y, Kubo M, Ishida N, Minegishi N, Nagasaki M, Sobue G. A multi-ethnic meta-analysis identifies novel genes, including ACSL5, associated with amyotrophic lateral sclerosis. Commun Biol 2020; 3:526. [PMID: 32968195 PMCID: PMC7511394 DOI: 10.1038/s42003-020-01251-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating progressive motor neuron disease that affects people of all ethnicities. Approximately 90% of ALS cases are sporadic and thought to have multifactorial pathogenesis. To understand the genetics of sporadic ALS, we conducted a genome-wide association study using 1,173 sporadic ALS cases and 8,925 controls in a Japanese population. A combined meta-analysis of our Japanese cohort with individuals of European ancestry revealed a significant association at the ACSL5 locus (top SNP p = 2.97 × 10−8). We validated the association with ACSL5 in a replication study with a Chinese population and an independent Japanese population (1941 ALS cases, 3821 controls; top SNP p = 1.82 × 10−4). In the combined meta-analysis, the intronic ACSL5 SNP rs3736947 showed the strongest association (p = 7.81 × 10−11). Using a gene-based analysis of the full multi-ethnic dataset, we uncovered additional genes significantly associated with ALS: ERGIC1, RAPGEF5, FNBP1, and ATXN3. These results advance our understanding of the genetic basis of sporadic ALS. Gen Sobue, Masao Nagasaki and colleagues report a genome-wide association study for amyotrophic lateral sclerosis (ALS) in a large, multi-ethnic cohort comprising Japanese, Chinese, and European ancestry populations. They find a significant association to variants within the ACSL5 gene and identify novel associations with 4 additional genes using a gene-based approach.
Collapse
Affiliation(s)
- Ryoichi Nakamura
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazuharu Misawa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.,Department of Molecular Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Genki Tohnai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masahiro Nakatochi
- Division of Data Science, Department of Nursing, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Sho Furuhashi
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Naoki Atsuta
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Naoki Hayashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Daichi Yokoi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.,Department of Neurology, Kakeyu-Misayama Rehabilitation Center Kakeyu Hospital, Ueda, Nagano, Japan
| | - Hazuki Watanabe
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.,Department of Neurology, Japanese Red Cross Nagoya Daiichi Hospital, Nagoya, Aichi, Japan
| | - Hirohisa Watanabe
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan.,Department of Neurology, Fujita Health University, Toyoake, Aichi, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuishin Izumi
- Department of Neurology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kazuaki Kanai
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Neurology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Akira Taniguchi
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Osamu Kano
- Division of Neurology, Department of Internal Medicine, Toho University Faculty of Medicine, Tokyo, Japan
| | - Masaya Oda
- Department of Neurology, Vihara Hananosato Hospital, Miyoshi, Hiroshima, Japan
| | - Kazumoto Shibuya
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naoki Suzuki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Yasuyuki Ohta
- Department of Neurology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Rina Hashimoto
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Aichi, Japan
| | - Ikuko Aiba
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Aichi, Japan
| | - Koichi Okamoto
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Gunma, Japan
| | - Kouichi Mizoguchi
- Department of Neurology, National Hospital Organization Shizuoka Medical Center, Shizuoka, Japan
| | - Kazuko Hasegawa
- Division of Neurology, National Hospital Organization, Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kenji Nakashima
- Department of Neurology, National Hospital Organization, Matsue Medical Center, Matsue, Shimane, Japan
| | - Ryuji Kaji
- Department of Neurology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Noriko Ishida
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Naoko Minegishi
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Masao Nagasaki
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan. .,Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Sakyo-ku, Kyoto, Japan. .,Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan.
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan. .,Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan. .,Aichi Medical University, Nagakute, Aichi, Japan.
| |
Collapse
|
506
|
Akiyama M. Multi-omics study for interpretation of genome-wide association study. J Hum Genet 2020; 66:3-10. [PMID: 32948838 DOI: 10.1038/s10038-020-00842-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/30/2020] [Accepted: 08/31/2020] [Indexed: 12/23/2022]
Abstract
Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with complex traits, including a wide variety of diseases. Despite the successful identification of associated loci, interpreting GWAS findings remains challenging and requires other biological resources. Omics, including genomics, transcriptomics, proteomics, metabolomics, and epigenomics, are increasingly used in a broad range of research fields. Integrative analyses applying GWAS with these omics data are expected to expand our knowledge of complex traits and provide insight into the pathogenesis of complex diseases and their causative factors. Recently, associations between genetic variants and omics data have been comprehensively evaluated, providing new information on the influence of genetic variants on omics. Furthermore, recent advances in analytic methods, including single-cell technologies, have revealed previously unknown disease mechanisms. To advance our understanding of complex traits, integrative analysis using GWAS with multi-omics data is needed. In this review, I describe successful examples of integrative analyses based on omics and GWAS, discuss the limitations of current multi-omics analyses, and provide a perspective on future integrative studies.
Collapse
Affiliation(s)
- Masato Akiyama
- Department of Ocular Pathology and Imaging Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan. .,Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.
| |
Collapse
|
507
|
Mutie PM, Pomares-Millan H, Atabaki-Pasdar N, Jordan N, Adams R, Daly NL, Tajes JF, Giordano GN, Franks PW. An investigation of causal relationships between prediabetes and vascular complications. Nat Commun 2020; 11:4592. [PMID: 32929089 PMCID: PMC7490420 DOI: 10.1038/s41467-020-18386-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 07/31/2020] [Indexed: 12/13/2022] Open
Abstract
Prediabetes is a state of glycaemic dysregulation below the diagnostic threshold of type 2 diabetes (T2D). Globally, ~352 million people have prediabetes, of which 35-50% develop full-blown diabetes within five years. T2D and its complications are costly to treat, causing considerable morbidity and early mortality. Whether prediabetes is causally related to diabetes complications is unclear. Here we report a causal inference analysis investigating the effects of prediabetes in coronary artery disease, stroke and chronic kidney disease, complemented by a systematic review of relevant observational studies. Although the observational studies suggest that prediabetes is broadly associated with diabetes complications, the causal inference analysis revealed that prediabetes is only causally related with coronary artery disease, with no evidence of causal effects on other diabetes complications. In conclusion, prediabetes likely causes coronary artery disease and its prevention is likely to be most effective if initiated prior to the onset of diabetes.
Collapse
Affiliation(s)
- Pascal M Mutie
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, Skåne University Hospital, Jan Waldenströms gata 35, Malmö, SE-20502, Sweden
| | - Hugo Pomares-Millan
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, Skåne University Hospital, Jan Waldenströms gata 35, Malmö, SE-20502, Sweden
| | - Naeimeh Atabaki-Pasdar
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, Skåne University Hospital, Jan Waldenströms gata 35, Malmö, SE-20502, Sweden
| | - Nina Jordan
- Regulatory Affairs Intelligence, Novo Nordisk A/S, Copenhagen, Denmark
| | - Rachel Adams
- Regulatory Affairs-Neuroscience and Cardiovascular Metabolism, Janssen, High Wycombe, UK
| | - Nicole L Daly
- Regulatory Affairs-Neuroscience and Cardiovascular Metabolism, Janssen, High Wycombe, UK
| | - Juan Fernandes Tajes
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, Skåne University Hospital, Jan Waldenströms gata 35, Malmö, SE-20502, Sweden
| | - Giuseppe N Giordano
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, Skåne University Hospital, Jan Waldenströms gata 35, Malmö, SE-20502, Sweden
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, Skåne University Hospital, Jan Waldenströms gata 35, Malmö, SE-20502, Sweden.
- Department of Public Health and Clinical Medicine, Section for Medicine, Umeå University, Umeå, Sweden.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
508
|
Geurtsen ML, Jaddoe VWV, Gaillard R, Felix JF. Associations of maternal early-pregnancy blood glucose and insulin concentrations with DNA methylation in newborns. Clin Epigenetics 2020; 12:134. [PMID: 32894192 PMCID: PMC7487846 DOI: 10.1186/s13148-020-00924-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 08/25/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Intrauterine exposure to a disturbed maternal glucose metabolism is associated with adverse offspring outcomes. DNA methylation is a potential mechanism underlying these associations. We examined whether maternal early-pregnancy glucose and insulin concentrations are associated with newborn DNA methylation. In a population-based prospective cohort study among 935 pregnant women, maternal plasma concentrations of non-fasting glucose and insulin were measured at a median of 13.1 weeks of gestation (95% range 9.4-17.4). DNA methylation was measured using the Infinium HumanMethylation450 BeadChip (Ilumina). We analyzed associations of maternal early-pregnancy glucose and insulin concentrations with single-CpG DNA methylation using robust linear regression models. Differentially methylated regions were analyzed using the dmrff package in R. We stratified the analyses on normal weight versus overweight or obese women. We also performed a look-up of CpGs and differently methylated regions from previous studies to be associated with maternal gestational diabetes, hyperglycemia or hyperinsulinemia, or with type 2 diabetes in adults. RESULTS Maternal early-pregnancy glucose and insulin concentrations were not associated with DNA methylation at single CpGs nor with differentially methylated regions in the total group. In analyses stratified on maternal BMI, maternal early-pregnancy glucose concentrations were associated with DNA methylation at one CpG (cg03617420, XKR6) among normal weight women and at another (cg12081946, IL17D) among overweight or obese women. No stratum-specific associations were found for maternal early-pregnancy insulin concentrations. The two CpGs were not associated with birth weight or childhood glycemic measures (p values > 0.1). Maternal early-pregnancy insulin concentrations were associated with one CpG known to be related to adult type 2 diabetes. Enrichment among nominally significant findings in our maternal early-pregnancy glucose concentrations was found for CpGs identified in a previous study on adult type 2 diabetes. CONCLUSIONS Maternal early-pregnancy glucose concentrations, but not insulin concentrations, were associated with DNA methylation at one CpG each in the subgroups of normal weight and of overweight or obese women. No associations were present in the full group. The role of these CpGs in mechanisms underlying offspring health outcomes needs further study. Future studies should replicate our results in larger samples with early-pregnancy information on maternal fasting glucose metabolism.
Collapse
Affiliation(s)
- Madelon L Geurtsen
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Romy Gaillard
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands.
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
509
|
Reilly DF, Breyer MD. The Use of Genomics to Drive Kidney Disease Drug Discovery and Development. Clin J Am Soc Nephrol 2020; 15:1342-1351. [PMID: 32193173 PMCID: PMC7480559 DOI: 10.2215/cjn.11070919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
As opposed to diseases such as cancer, autoimmune disease, and diabetes, identifying drugs to treat CKD has proven significantly more challenging. Over the past 2 decades, new potential therapeutic targets have been identified as genetically altered proteins involved in rare monogenetic kidney diseases. Other possible target genes have been implicated through common genetic polymorphisms associated with CKD in the general population. Significant challenges remain before translating these genetic insights into clinical therapies for CKD. This paper will discuss how genetic variants may be leveraged to develop drugs and will especially focus on those genes associated with CKD to exemplify the value and challenges in including genetic information in the drug development pipeline.
Collapse
Affiliation(s)
- Dermot F Reilly
- Cardiovascular and Metabolism Discovery Research, Janssen Research and Development LLC, Boston, Massachusetts
| | | |
Collapse
|
510
|
Leong A, Cole J, Brenner LN, Meigs JB, Florez JC, Mercader JM. Cardiometabolic Risk Factors for COVID-19 Susceptibility and Severity: A Mendelian Randomization Analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 32909013 DOI: 10.1101/2020.08.26.20182709] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
IMPORTANCE Early epidemiological studies report associations of diverse cardiometabolic conditions especially body mass index (BMI), with COVID-19 susceptibility and severity, but causality has not been established. Identifying causal risk factors is critical to inform preventive strategies aimed at modifying disease risk. OBJECTIVE We sought to evaluate the causal associations of cardiometabolic conditions with COVID-19 susceptibility and severity. DESIGN Two-sample Mendelian Randomization (MR) Study. SETTING Population-based cohorts that contributed to the genome-wide association study (GWAS) meta-analysis by the COVID-19 Host Genetics Initiative. PARTICIPANTS Patients hospitalized with COVID-19 diagnosed by RNA PCR, serologic testing, or clinician diagnosis. Population controls defined as anyone who was not a case in the cohorts. Exposures: Selected genetic variants associated with 17 cardiometabolic diseases, including diabetes, coronary artery disease, stroke, chronic kidney disease, and BMI, at p<5 x 10-8 from published largescale GWAS. MAIN OUTCOMES We performed an inverse-variance weighted averages of variant-specific causal estimates for susceptibility, defined as people who tested positive for COVID-19 vs. population controls, and severity, defined as patients hospitalized with COVID-19 vs. population controls, and repeated the analysis for BMI using effect estimates from UKBB. To estimate direct and indirect causal effects of BMI through obesity-related cardiometabolic diseases, we performed pairwise multivariable MR. We used p<0.05/17 exposure/2 outcomes=0.0015 to declare statistical significance. RESULTS Genetically increased BMI was causally associated with testing positive for COVID-19 [6,696 cases / 1,073,072 controls; p=6.7 x 10-4, odds ratio and 95% confidence interval 1.08 (1.03, 1.13) per kg/m2] and a higher risk of COVID-19 hospitalization [3,199 cases/897,488 controls; p=8.7 x 10-4, 1.12 (1.04, 1.21) per kg/m2]. In the multivariable MR, the direct effect of BMI was abolished upon conditioning on the effect on type 2 diabetes but persisted when conditioning on the effects on coronary artery disease, stroke, chronic kidney disease, and c-reactive protein. No other cardiometabolic exposures tested were associated with a higher risk of poorer COVID-19 outcomes. CONCLUSIONS AND RELEVANCE Genetic evidence supports BMI as a causal risk factor for COVID-19 susceptibility and severity. This relationship may be mediated via type 2 diabetes. Obesity may have amplified the disease burden of the COVID-19 pandemic either single-handedly or through its metabolic consequences.
Collapse
|
511
|
Fan L, Gao W, Nguyen BV, Jefferson JR, Liu Y, Fan F, Roman RJ. Impaired renal hemodynamics and glomerular hyperfiltration contribute to hypertension-induced renal injury. Am J Physiol Renal Physiol 2020; 319:F624-F635. [PMID: 32830539 DOI: 10.1152/ajprenal.00239.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Recently, we reported a mutation in γ-adducin (ADD3) was associated with an impaired myogenic response of the afferent arteriole and hypertension-induced chronic kidney disease (CKD) in fawn hooded hypertensive (FHH) rats. However, the mechanisms by which altered renal blood flow (RBF) autoregulation promotes hypertension-induced renal injury remain to be determined. The present study compared the time course of changes in renal hemodynamics and the progression of CKD during the development of DOCA-salt hypertension in FHH 1BN congenic rats [wild-type (WT)] with an intact myogenic response versus FHH 1BN Add3KO (Add3KO) rats, which have impaired myogenic response. RBF was well autoregulated in WT rats but not in Add3KO rats. Glomerular capillary pressure rose by 6 versus 14 mmHg in WT versus Add3KO rats when blood pressure increased from 100 to 150 mmHg. After 1 wk of hypertension, glomerular filtration rate increased by 38% and glomerular nephrin expression decreased by 20% in Add3KO rats. Neither were altered in WT rats. Proteinuria doubled in WT rats versus a sixfold increase in Add3KO rats. The degree of renal injury was greater in Add3KO than WT rats after 3 wk of hypertension. RBF, glomerular filtration rate, and glomerular capillary pressure were lower by 20%, 28%, and 19% in Add3KO rats than in WT rats, which was associated with glomerular matrix expansion and loss of capillary filtration area. The results indicated that impaired RBF autoregulation and eutrophic remodeling of preglomerular arterioles increase the transmission of pressure to glomeruli, which induces podocyte loss and accelerates the progression of CKD in hypertensive Add3KO rats.
Collapse
Affiliation(s)
- Letao Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Wenjun Gao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bond V Nguyen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joshua R Jefferson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
512
|
Li Y, Haug S, Schlosser P, Teumer A, Tin A, Pattaro C, Köttgen A, Wuttke M. Integration of GWAS Summary Statistics and Gene Expression Reveals Target Cell Types Underlying Kidney Function Traits. J Am Soc Nephrol 2020; 31:2326-2340. [PMID: 32764137 DOI: 10.1681/asn.2020010051] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Genetic variants identified in genome-wide association studies (GWAS) are often not specific enough to reveal complex underlying physiology. By integrating RNA-seq data and GWAS summary statistics, novel computational methods allow unbiased identification of trait-relevant tissues and cell types. METHODS The CKDGen consortium provided GWAS summary data for eGFR, urinary albumin-creatinine ratio (UACR), BUN, and serum urate. Genotype-Tissue Expression Project (GTEx) RNA-seq data were used to construct the top 10% specifically expressed genes for each of 53 tissues followed by linkage disequilibrium (LD) score-based enrichment testing for each trait. Similar procedures were performed for five kidney single-cell RNA-seq datasets from humans and mice and for a microdissected tubule RNA-seq dataset from rat. Gene set enrichment analyses were also conducted for genes implicated in Mendelian kidney diseases. RESULTS Across 53 tissues, genes in kidney function-associated GWAS loci were enriched in kidney (P=9.1E-8 for eGFR; P=1.2E-5 for urate) and liver (P=6.8·10-5 for eGFR). In the kidney, proximal tubule was enriched in humans (P=8.5E-5 for eGFR; P=7.8E-6 for urate) and mice (P=0.0003 for eGFR; P=0.0002 for urate) and confirmed as the primary cell type in microdissected tubules and organoids. Gene set enrichment analysis supported this and showed enrichment of genes implicated in monogenic glomerular diseases in podocytes. A systematic approach generated a comprehensive list of GWAS genes prioritized by cell type-specific expression. CONCLUSIONS Integration of GWAS statistics of kidney function traits and gene expression data identified relevant tissues and cell types, as a basis for further mechanistic studies to understand GWAS loci.
Collapse
Affiliation(s)
- Yong Li
- Institute of Genetic Epidemiology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Stefan Haug
- Institute of Genetic Epidemiology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany.,German Center for Cardiovascular Research, Partner Site Greifswald, Greifswald, Germany
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Medical Center-University of Freiburg, Freiburg, Germany.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Medical Center-University of Freiburg, Freiburg, Germany
| |
Collapse
|
513
|
Scholz M, Henger S, Beutner F, Teren A, Baber R, Willenberg A, Ceglarek U, Pott J, Burkhardt R, Thiery J. Cohort Profile: The Leipzig Research Center for Civilization Diseases–Heart Study (LIFE-Heart). Int J Epidemiol 2020; 49:1439-1440h. [DOI: 10.1093/ije/dyaa075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Affiliation(s)
- Markus Scholz
- Institute for Medical Informatics, Statistic and Epidemiology, University of Leipzig, Leipzig, Germany
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
| | - Sylvia Henger
- Institute for Medical Informatics, Statistic and Epidemiology, University of Leipzig, Leipzig, Germany
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
| | - Frank Beutner
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- Heart Center Leipzig, Leipzig, Germany
| | - Andrej Teren
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- Heart Center Leipzig, Leipzig, Germany
| | - Ronny Baber
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Anja Willenberg
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Janne Pott
- Institute for Medical Informatics, Statistic and Epidemiology, University of Leipzig, Leipzig, Germany
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
| | - Ralph Burkhardt
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Joachim Thiery
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
514
|
Zhao JV, Schooling CM. Sex-specific associations of insulin resistance with chronic kidney disease and kidney function: a bi-directional Mendelian randomisation study. Diabetologia 2020; 63:1554-1563. [PMID: 32409868 DOI: 10.1007/s00125-020-05163-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/24/2020] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Reasons for the sexual disparity in chronic kidney disease (CKD) are unclear. To provide insight we contextualised these differences within evolutionary biology, and explored sex-specific effects of insulin resistance because it may have sex-specific effects on the reproductive axis. Impaired kidney function may also cause insulin resistance. We assessed these possibilities using bi-directional, sex-specific, two-sample Mendelian randomisation (MR). METHODS Given that fasting insulin, fasting glucose and HbA1c are related, we used MR-Bayesian model averaging (MR-BMA) to identify the best-fitting model and most influential exposure. Genetic associations with glycaemic traits were obtained from genome-wide association studies (GWAS) in Europeans without diabetes (n = 108,557 for fasting insulin, as a proxy for insulin resistance, and for fasting glucose, n = 123,665 for HbA1c in the Meta-Analyses of Glucose and Insulin-related traits Consortium [MAGIC]), and applied to GWAS of 480,698 Europeans for overall associations with CKD (cases n = 41,395) and eGFR. We also used sex-specific individual information in white British (179,917 men, 6016 CKD cases; 212,079 women, 5958 CKD cases) from the UK Biobank. Univariable or multivariable MR was used to assess the role of glycaemic trait(s) selected by MR-BMA in CKD and kidney function. Genetic variants predicting eGFR were used to assess the role of kidney function in the most influential exposure(s). RESULTS Fasting insulin was selected as the most likely exposure by both overall and sex-specific MR-BMA. It increased CKD in men (OR 7.23 per pmol/l higher fasting insulin [95% CI 2.46, 21.2]) but not in women (OR 1.05 [95% CI 0.21, 5.21]), and reduced eGFR in men (-0.04 [95% CI -0.07, -0.01]) but not in women (0.01 [95% CI -0.02, 0.03]). Genetically predicted eGFR was unrelated to fasting insulin. CONCLUSIONS/INTERPRETATION Genetically predicted fasting insulin was sex-specifically associated with CKD and unhealthier kidney function but was not affected by kidney function. Graphical abstract.
Collapse
Affiliation(s)
- Jie V Zhao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Hong Kong, SAR, China.
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Hong Kong, SAR, China
- City University of New York, School of Public Health and Health Policy, New York, NY, USA
| |
Collapse
|
515
|
Cheng Y, Li Y, Benkowitz P, Lamina C, Köttgen A, Sekula P. The relationship between blood metabolites of the tryptophan pathway and kidney function: a bidirectional Mendelian randomization analysis. Sci Rep 2020; 10:12675. [PMID: 32728058 PMCID: PMC7391729 DOI: 10.1038/s41598-020-69559-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
Blood metabolites of the tryptophan pathway were found to be associated with kidney function and disease in observational studies. In order to evaluate causal relationship and direction, we designed a study using a bidirectional Mendelian randomization approach. The analyses were based on published summary statistics with study sizes ranging from 1,960 to 133,413. After correction for multiple testing, results provided no evidence of an effect of metabolites of the tryptophan pathway on estimated glomerular filtration rate (eGFR). Conversely, lower eGFR was related to higher levels of four metabolites: C-glycosyltryptophan (effect estimate = − 0.16, 95% confidence interval [CI] (− 0.22; − 0.1); p = 9.2e−08), kynurenine (effect estimate = − 0.18, 95% CI (− 0.25; − 0.11); p = 1.1e−06), 3-indoxyl sulfate (effect estimate = − 0.25, 95% CI (− 0.4; − 0.11); p = 6.3e−04) and indole-3-lactate (effect estimate = − 0.26, 95% CI (− 0.38; − 0.13); p = 5.4e−05). Our study supports that lower eGFR causes higher blood metabolite levels of the tryptophan pathway including kynurenine, C-glycosyltryptophan, 3-indoxyl sulfate, and indole-3-lactate. These findings aid the notion that metabolites of the tryptophan pathway are a consequence rather than a cause of reduced eGFR. Further research is needed to specifically examine relationships with respect to chronic kidney disease (CKD) progression among patients with existing CKD.
Collapse
Affiliation(s)
- Yurong Cheng
- Department of Biometry, Epidemiology and Medical Bioinformatics, Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Hugstetter Str. 49, 79106, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Yong Li
- Department of Biometry, Epidemiology and Medical Bioinformatics, Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Hugstetter Str. 49, 79106, Freiburg, Germany
| | - Paula Benkowitz
- Department of Biometry, Epidemiology and Medical Bioinformatics, Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Hugstetter Str. 49, 79106, Freiburg, Germany
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Köttgen
- Department of Biometry, Epidemiology and Medical Bioinformatics, Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Hugstetter Str. 49, 79106, Freiburg, Germany
| | - Peggy Sekula
- Department of Biometry, Epidemiology and Medical Bioinformatics, Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Hugstetter Str. 49, 79106, Freiburg, Germany.
| |
Collapse
|
516
|
Verma SS, Bergmeijer TO, Gong L, Reny JL, Lewis JP, Mitchell BD, Alexopoulos D, Aradi D, Altman RB, Bliden K, Bradford Y, Campo G, Chang K, Cleator JH, Déry JP, Dridi NP, Fernandez-Cadenas I, Fontana P, Gawaz M, Geisler T, Gensini GF, Giusti B, Gurbel PA, Hochholzer W, Holmvang L, Kim EY, Kim HS, Marcucci R, Montaner J, Backman JD, Pakyz RE, Roden DM, Schaeffeler E, Schwab M, Shin JG, Siller-Matula JM, Ten Berg JM, Trenk D, Valgimigli M, Wallace J, Wen MS, Kubo M, Lee MTM, Whaley R, Winter S, Klein TE, Shuldiner AR, Ritchie MD. Genomewide Association Study of Platelet Reactivity and Cardiovascular Response in Patients Treated With Clopidogrel: A Study by the International Clopidogrel Pharmacogenomics Consortium. Clin Pharmacol Ther 2020; 108:1067-1077. [PMID: 32472697 PMCID: PMC7689744 DOI: 10.1002/cpt.1911] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/08/2020] [Indexed: 01/07/2023]
Abstract
Antiplatelet response to clopidogrel shows wide variation, and poor response is correlated with adverse clinical outcomes. CYP2C19 loss‐of‐function alleles play an important role in this response, but account for only a small proportion of variability in response to clopidogrel. An aim of the International Clopidogrel Pharmacogenomics Consortium (ICPC) is to identify other genetic determinants of clopidogrel pharmacodynamics and clinical response. A genomewide association study (GWAS) was performed using DNA from 2,750 European ancestry individuals, using adenosine diphosphate‐induced platelet reactivity and major cardiovascular and cerebrovascular events as outcome parameters. GWAS for platelet reactivity revealed a strong signal for CYP2C19*2 (P value = 1.67e−33). After correction for CYP2C19*2 no other single‐nucleotide polymorphism reached genomewide significance. GWAS for a combined clinical end point of cardiovascular death, myocardial infarction, or stroke (5.0% event rate), or a combined end point of cardiovascular death or myocardial infarction (4.7% event rate) showed no significant results, although in coronary artery disease, percutaneous coronary intervention, and acute coronary syndrome subgroups, mutations in SCOS5P1, CDC42BPA, and CTRAC1 showed genomewide significance (lowest P values: 1.07e−09, 4.53e−08, and 2.60e−10, respectively). CYP2C19*2 is the strongest genetic determinant of on‐clopidogrel platelet reactivity. We identified three novel associations in clinical outcome subgroups, suggestive for each of these outcomes.
Collapse
Affiliation(s)
- Shefali Setia Verma
- Department of Genetics and Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas O Bergmeijer
- Department of Cardiology, St. Antonius Center for Platelet Function Research, Nieuwegein, The Netherlands
| | - Li Gong
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Jean-Luc Reny
- Internal Medicine, Béziers Hospital, Béziers, France.,Geneva Platelet Group, School of Medicine, University of Geneva, Geneva, Switzerland.,Department of Internal Medicine, Rehabilitation and Geriatrics, University Hospitals of Geneva, Geneva, Switzerland.,Geneva Platelet Group and Division of Angiology and Haemostasis, University Hospitals of Geneva, Geneva, Switzerland
| | - Joshua P Lewis
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Braxton D Mitchell
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, Maryland, USA.,Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, Maryland, USA
| | - Dimitrios Alexopoulos
- National and Kapodistrian University of Athens Medical School, Attikon University Hospital, Athens, Greece
| | - Daniel Aradi
- Department of Cardiology, Heart Center Balatonfüred, Balatonfüred, Hungary
| | - Russ B Altman
- Department of Bioengineering, Genetics and Medicine, Stanford University, Stanford, California, USA
| | - Kevin Bliden
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, Maryland, USA
| | - Yuki Bradford
- Department of Genetics and Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gianluca Campo
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara and Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Kiyuk Chang
- Department of Internal Medicine, Cardiology Division, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - John H Cleator
- Division of Cardiology and Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jean-Pierre Déry
- Quebec Heart and Lung Institute, University Laval, Quebec City, QC, Canada
| | - Nadia P Dridi
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Israel Fernandez-Cadenas
- Neurology, Stroke Pharmacogenomics and Genetics Group, Sant Pau Institute of Research, Barcelona, Spain
| | - Pierre Fontana
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA.,Geneva Platelet Group and Division of Angiology and Haemostasis, University Hospitals of Geneva, Geneva, Switzerland
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University of Tübingen, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, Medizinische Klinik III, University Hospital Tübingen, Tübingen, Germany
| | - Gian Franco Gensini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, Maryland, USA
| | - Willibald Hochholzer
- Department of Cardiology and Angiology II, University Heart Center Freiburg Bad Krozingen, Bad Krozingen, Germany
| | - Lene Holmvang
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Eun-Young Kim
- Department of Clinical Pharmacology, Inje University, Busan Paik Hospital, Busan, South Korea
| | - Ho-Sook Kim
- Department of Clinical Pharmacology, Inje University, Busan Paik Hospital, Busan, South Korea
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Barcelona, Spain
| | - Joshua D Backman
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Ruth E Pakyz
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Dan M Roden
- Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Tübingen, Germany.,Department of Clinical Pharmacology, and Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany
| | - Jae Gook Shin
- Department of Clinical Pharmacology, Inje University, Busan Paik Hospital, Busan, South Korea.,Department of Pharmacology and Pharmacogenomics Research Center, Inje University, Busan Paik Hospital, Busan, South Korea
| | - Jolanta M Siller-Matula
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.,Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| | - Jurriën M Ten Berg
- Department of Cardiology, St. Antonius Center for Platelet Function Research, Nieuwegein, The Netherlands
| | - Dietmar Trenk
- Department of Cardiology and Angiology II, University Heart Center Freiburg Bad Krozingen, Bad Krozingen, Germany.,Department of Clinical Pharmacology, University Heart Centre Freiburg, Bad Krozingen, Germany
| | - Marco Valgimigli
- Department of Cardiology, Swiss Cardiovascular Center Bern, Bern University Hospital, Bern, Switzerland
| | - John Wallace
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA
| | - Ming-Shien Wen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou and School of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Michiaki Kubo
- Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | | | - Ryan Whaley
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Tübingen, Germany
| | - Teri E Klein
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA.,Department of Medicine, Stanford University, Stanford, California, USA
| | - Alan R Shuldiner
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Marylyn D Ritchie
- Department of Genetics and Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
517
|
Zhang YM, Zheng J, Gaunt TR, Zhang H. Mendelian Randomization Analysis Reveals a Causal Effect of Urinary Sodium/Urinary Creatinine Ratio on Kidney Function in Europeans. Front Bioeng Biotechnol 2020; 8:662. [PMID: 32733862 PMCID: PMC7358605 DOI: 10.3389/fbioe.2020.00662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/28/2020] [Indexed: 11/13/2022] Open
Abstract
Salt restriction was recommended in clinical practice guideline for chronic kidney disease (CKD) treatment, but its effect on kidney outcomes remains conflicting. We aimed to test the causal effect of salt intake, using estimated 24-h sodium excretion from spot urinary sodium/urinary creatinine (UNa/UCr) ratio as a surrogate, on renal function using two-sample Mendelian randomization (MR). Genetic instruments for UNa/UCr were derived from a recent genome-wide association study of 218,450 European-descent individuals in the UK Biobank. Kidney outcomes were creatinine-based estimated glomerular filtration rate (eGFRcrea) (N = 567,460) and CKD (eGFRcrea < 60 ml/min/1.73 m2, N cases = 41,395, N controls = 439,303) from the CKDGen consortium. Cystatin C-based eGFR (eGFRcys) and eGFRcrea single-nucleotide polymorphisms associated with blood urea nitrogen (BUN) were used for sensitivity analyses. MR revealed a causal effect of UNa/UCr on higher eGFRcrea [β = 0.14, unit change in log ml/min/1.73 m2 per UNa/UCr ratio; 95% confidence interval (CI) = 0.07 - 0.20, P = 2.15 × 10-5] and a protective effect against CKD risk (odds ratio = 0.24, 95% CI = 0.14 to 0.41, P = 1.20 × 10-7). The MR findings were confirmed by MR-Egger regression, weighted median MR, and mode estimate MR, with less evidence of existence of horizontal pleiotropy. Consistent positive causal effect of UNa/UCr on eGFRcys was also detected. On the other hand, bidirectional MR suggested inconclusive results of CKD, eGFRcrea, eGFRcrea (BUN associated), and eGFRcys on UNa/UCr. The average 24-h sodium excretion was estimated to be approximately 2.6 g per day for women and 3.7 g per day for men. This study provides evidence that sodium excretion, well above the recommendation of <2 g per day of sodium intake, might not have a harmful effect on kidney function. Clinical trials are warranted to evaluate the sodium restriction target on kidney function.
Collapse
Affiliation(s)
- Yue-Miao Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Jie Zheng
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Bristol, United Kingdom
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Bristol, United Kingdom.,National Institute for Health Research (NIHR), Bristol Biomedical Research Centre, Bristol, United Kingdom
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
518
|
Lin J, Susztak K. Complexities of Understanding Function from CKD-Associated DNA Variants. Clin J Am Soc Nephrol 2020; 15:1028-1040. [PMID: 32513823 PMCID: PMC7341770 DOI: 10.2215/cjn.15771219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Genome-wide association studies (GWASs) have facilitated the unbiased discovery of hundreds of genomic loci associated with CKD and kidney function. The vast majority of disease-associated DNA variants are noncoding. Those that are causal in CKD pathogenesis likely modulate transcription of target genes in a cell type-specific manner. To gain novel biological insights into mechanisms driving the development of CKD, the causal variants (which are usually not the most significant variant reported in a GWAS), their target genes, and causal cell types need to be identified. This functional validation requires a large number of new data sets, complex bioinformatics analyses, and experimental cellular and in vivo studies. Here, we review the basic principles and some of the current approaches being leveraged to assign functional significance to a genotype-phenotype association.
Collapse
Affiliation(s)
- Jennie Lin
- Division of Nephrology and Hypertension, Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Northwestern University, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Genetics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
519
|
Abstract
Diabetes is one of the fastest growing diseases worldwide, projected to affect 693 million adults by 2045. Devastating macrovascular complications (cardiovascular disease) and microvascular complications (such as diabetic kidney disease, diabetic retinopathy and neuropathy) lead to increased mortality, blindness, kidney failure and an overall decreased quality of life in individuals with diabetes. Clinical risk factors and glycaemic control alone cannot predict the development of vascular complications; numerous genetic studies have demonstrated a clear genetic component to both diabetes and its complications. Early research aimed at identifying genetic determinants of diabetes complications relied on familial linkage analysis suited to strong-effect loci, candidate gene studies prone to false positives, and underpowered genome-wide association studies limited by sample size. The explosion of new genomic datasets, both in terms of biobanks and aggregation of worldwide cohorts, has more than doubled the number of genetic discoveries for both diabetes and diabetes complications. We focus herein on genetic discoveries for diabetes and diabetes complications, empowered primarily through genome-wide association studies, and emphasize the gaps in research for taking genomic discovery to the next level.
Collapse
Affiliation(s)
- Joanne B Cole
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA, USA
| | - Jose C Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
520
|
Tofte N, Vogelzangs N, Mook-Kanamori D, Brahimaj A, Nano J, Ahmadizar F, van Dijk KW, Frimodt-Møller M, Arts I, Beulens JWJ, Rutters F, van der Heijden AA, Kavousi M, Stehouwer CDA, Nijpels G, van Greevenbroek MMJ, van der Kallen CJH, Rossing P, Ahluwalia TS, 't Hart LM. Plasma Metabolomics Identifies Markers of Impaired Renal Function: A Meta-analysis of 3089 Persons with Type 2 Diabetes. J Clin Endocrinol Metab 2020; 105:5818360. [PMID: 32271379 DOI: 10.1210/clinem/dgaa173] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/08/2020] [Indexed: 01/10/2023]
Abstract
CONTEXT There is a need for novel biomarkers and better understanding of the pathophysiology of diabetic kidney disease. OBJECTIVE To investigate associations between plasma metabolites and kidney function in people with type 2 diabetes (T2D). DESIGN 3089 samples from individuals with T2D, collected between 1999 and 2015, from 5 independent Dutch cohort studies were included. Up to 7 years follow-up was available in 1100 individuals from 2 of the cohorts. MAIN OUTCOME MEASURES Plasma metabolites (n = 149) were measured by nuclear magnetic resonance spectroscopy. Associations between metabolites and estimated glomerular filtration rate (eGFR), urinary albumin-to-creatinine ratio (UACR), and eGFR slopes were investigated in each study followed by random effect meta-analysis. Adjustments included traditional cardiovascular risk factors and correction for multiple testing. RESULTS In total, 125 metabolites were significantly associated (PFDR = 1.5×10-32 - 0.046; β = -11.98-2.17) with eGFR. Inverse associations with eGFR were demonstrated for branched-chain and aromatic amino acids (AAAs), glycoprotein acetyls, triglycerides (TGs), lipids in very low-density lipoproteins (VLDL) subclasses, and fatty acids (PFDR < 0.03). We observed positive associations with cholesterol and phospholipids in high-density lipoproteins (HDL) and apolipoprotein A1 (PFDR < 0.05). Albeit some metabolites were associated with UACR levels (P < 0.05), significance was lost after correction for multiple testing. Tyrosine and HDL-related metabolites were positively associated with eGFR slopes before adjustment for multiple testing (PTyr = 0.003; PHDLrelated < 0.05), but not after. CONCLUSIONS This study identified metabolites associated with impaired kidney function in T2D, implying involvement of lipid and amino acid metabolism in the pathogenesis. Whether these processes precede or are consequences of renal impairment needs further investigation.
Collapse
Affiliation(s)
- Nete Tofte
- Steno Diabetes Center, Copenhagen, Denmark
| | - Nicole Vogelzangs
- Department of Epidemiology & Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Dennis Mook-Kanamori
- Departments of Clinical Epidemiology and Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Adela Brahimaj
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of General Practice, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jana Nano
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Institute of Epidemiology, Helmholtz Zentrum, Munich, Germany
- German Center for Diabetes Research, Munich, Germany
| | - Fariba Ahmadizar
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ko Willems van Dijk
- Departments of Human Genetics and Internal Medicine/Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Ilja Arts
- Department of Epidemiology & Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Joline W J Beulens
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Femke Rutters
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Amber A van der Heijden
- Department of General Practice and Elderly Care Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Coen D A Stehouwer
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Giel Nijpels
- Department of General Practice and Elderly Care Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Marleen M J van Greevenbroek
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Carla J H van der Kallen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Peter Rossing
- Steno Diabetes Center, Copenhagen, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | | | - Leen M 't Hart
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Center, Amsterdam, the Netherlands
- Department of Cell and Chemical Biology & Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
521
|
Mahootchi E, Cannon Homaei S, Kleppe R, Winge I, Hegvik TA, Megias-Perez R, Totland C, Mogavero F, Baumann A, Glennon JC, Miletic H, Kursula P, Haavik J. GADL1 is a multifunctional decarboxylase with tissue-specific roles in β-alanine and carnosine production. SCIENCE ADVANCES 2020; 6:eabb3713. [PMID: 32733999 PMCID: PMC7367687 DOI: 10.1126/sciadv.abb3713] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/05/2020] [Indexed: 05/26/2023]
Abstract
Carnosine and related β-alanine-containing peptides are believed to be important antioxidants, pH buffers, and neuromodulators. However, their biosynthetic routes and therapeutic potential are still being debated. This study describes the first animal model lacking the enzyme glutamic acid decarboxylase-like 1 (GADL1). We show that Gadl1-/- mice are deficient in β-alanine, carnosine, and anserine, particularly in the olfactory bulb, cerebral cortex, and skeletal muscle. Gadl1-/- mice also exhibited decreased anxiety, increased levels of oxidative stress markers, alterations in energy and lipid metabolism, and age-related changes. Examination of the GADL1 active site indicated that the enzyme may have multiple physiological substrates, including aspartate and cysteine sulfinic acid. Human genetic studies show strong associations of the GADL1 locus with plasma levels of carnosine, subjective well-being, and muscle strength. Together, this shows the multifaceted and organ-specific roles of carnosine peptides and establishes Gadl1 knockout mice as a versatile model to explore carnosine biology and its therapeutic potential.
Collapse
Affiliation(s)
| | - Selina Cannon Homaei
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Rune Kleppe
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Ingeborg Winge
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Tor-Arne Hegvik
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Christian Totland
- Department of Chemistry, University of Bergen, Bergen, Norway
- Norwegian Geotechnical Institute, Oslo, Norway
| | - Floriana Mogavero
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anne Baumann
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Jeffrey Colm Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Petri Kursula
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
522
|
Pietzner M, Wheeler E, Carrasco-Zanini J, Raffler J, Kerrison ND, Oerton E, Auyeung VP, Luan J, Finan C, Casas JP, Ostroff R, Williams SA, Kastenmüller G, Ralser M, Gamazon ER, Wareham NJ, Hingorani AD, Langenberg C. Genetic architecture of host proteins interacting with SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.01.182709. [PMID: 32637948 PMCID: PMC7337378 DOI: 10.1101/2020.07.01.182709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Strategies to develop therapeutics for SARS-CoV-2 infection may be informed by experimental identification of viral-host protein interactions in cellular assays and measurement of host response proteins in COVID-19 patients. Identification of genetic variants that influence the level or activity of these proteins in the host could enable rapid 'in silico' assessment in human genetic studies of their causal relevance as molecular targets for new or repurposed drugs to treat COVID-19. We integrated large-scale genomic and aptamer-based plasma proteomic data from 10,708 individuals to characterize the genetic architecture of 179 host proteins reported to interact with SARS-CoV-2 proteins or to participate in the host response to COVID-19. We identified 220 host DNA sequence variants acting in cis (MAF 0.01-49.9%) and explaining 0.3-70.9% of the variance of 97 of these proteins, including 45 with no previously known protein quantitative trait loci (pQTL) and 38 encoding current drug targets. Systematic characterization of pQTLs across the phenome identified protein-drug-disease links, evidence that putative viral interaction partners such as MARK3 affect immune response, and establish the first link between a recently reported variant for respiratory failure of COVID-19 patients at the ABO locus and hypercoagulation, i.e. maladaptive host response. Our results accelerate the evaluation and prioritization of new drug development programmes and repurposing of trials to prevent, treat or reduce adverse outcomes. Rapid sharing and dynamic and detailed interrogation of results is facilitated through an interactive webserver ( https://omicscience.org/apps/covidpgwas/ ).
Collapse
Affiliation(s)
- Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Erin Oerton
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - Jian’an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, UK
- UCL BHF Research Accelerator centre
| | - Juan P. Casas
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, USA
| | | | | | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Markus Ralser
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
- Department of Biochemistry, Charité University Medicine, Berlin, Germany
| | - Eric R. Gamazon
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, UK
| | - Aroon D. Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, UK
- UCL BHF Research Accelerator centre
- Health Data Research UK, Institute of Health Informatics, University College London, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, UK
| |
Collapse
|
523
|
Schmidt AF, Finan C, Gordillo-Marañón M, Asselbergs FW, Freitag DF, Patel RS, Tyl B, Chopade S, Faraway R, Zwierzyna M, Hingorani AD. Genetic drug target validation using Mendelian randomisation. Nat Commun 2020; 11:3255. [PMID: 32591531 PMCID: PMC7320010 DOI: 10.1038/s41467-020-16969-0] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Mendelian randomisation (MR) analysis is an important tool to elucidate the causal relevance of environmental and biological risk factors for disease. However, causal inference is undermined if genetic variants used to instrument a risk factor also influence alternative disease-pathways (horizontal pleiotropy). Here we report how the 'no horizontal pleiotropy assumption' is strengthened when proteins are the risk factors of interest. Proteins are typically the proximal effectors of biological processes encoded in the genome. Moreover, proteins are the targets of most medicines, so MR studies of drug targets are becoming a fundamental tool in drug development. To enable such studies, we introduce a mathematical framework that contrasts MR analysis of proteins with that of risk factors located more distally in the causal chain from gene to disease. We illustrate key model decisions and introduce an analytical framework for maximising power and evaluating the robustness of analyses.
Collapse
Affiliation(s)
- Amand F Schmidt
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK.
- UCL BHF Research Accelerator Centre, London, UK.
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK
- UCL BHF Research Accelerator Centre, London, UK
| | - Maria Gordillo-Marañón
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK
| | - Folkert W Asselbergs
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Health Data Research UK, 222 Euston Road, London, UK
| | - Daniel F Freitag
- Bayer AG Pharmaceuticals, Open Innovation & Digital Technologies, Aprather Weg 18a, Wuppertal, 42096, Germany
| | - Riyaz S Patel
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK
- UCL BHF Research Accelerator Centre, London, UK
| | - Benoît Tyl
- Center for Therapeutic Innovation, Cardiovascular and Metabolic Disease, Institut de Recherches Internationales Servier, 50 Rue Carnot, 92284, Suresnes Cedex, France
| | - Sandesh Chopade
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK
- UCL BHF Research Accelerator Centre, London, UK
| | - Rupert Faraway
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK
- UCL BHF Research Accelerator Centre, London, UK
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
| | - Magdalena Zwierzyna
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK
- UCL BHF Research Accelerator Centre, London, UK
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT, UK
- UCL BHF Research Accelerator Centre, London, UK
- Health Data Research UK, 222 Euston Road, London, UK
| |
Collapse
|
524
|
Butler-Laporte G, Harroud A, Forgetta V, Richards JB. Elevated body mass index is associated with an increased risk of infectious disease admissions and mortality: a mendelian randomization study. Clin Microbiol Infect 2020; 27:S1198-743X(20)30356-6. [PMID: 32592749 DOI: 10.1016/j.cmi.2020.06.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The effect of body mass index (BMI) on the risk of infectious diseases admissions and mortality is unclear and is difficult to study given the risks of confounding variables. METHODS We used genome-wide association studies (GWASs) with mendelian randomization (MR) to obtain causal inference of BMI on the following infectious diseases outcomes: hospital admissions for pneumonia, sepsis, urinary tract infections, skin and soft tissue infections (SSTIs) or all-cause infections. For patients with pneumonia and sepsis, we also analysed their 28-day and 90-day mortalities. The UK Biobank (UKB) cohort (n > 500 000) provided data for GWASs on infectious diseases. The GIANT consortium (n = 681 265) GWAS was used to identify single-nucleotide polymorphisms (SNPs) associated with BMI. RESULTS Genetically increased BMI, by one standard deviation, was associated with higher rates of admission due to all infectious disease. The effect was most important for SSTIs (OR: 1.11, 95%CI: 1.09, 1.12). Increasing BMI by one standard deviation was associated with higher pneumonia mortality, especially at 28 days (OR: 1.03, 95%CI: 1.01, 1.05). BMI was not clearly associated with sepsis mortality, although interpretation of the results was limited by a small sample size. There were consistent findings in sensitivity analysis performed by removing highly pleiotropic SNPs and multivariate MR including type-2 diabetes mellitus, estimated glomerular filtration rate, high-density lipoprotein, educational attainment, and a history of smoking. CONCLUSIONS Increased BMI was associated with increased risk of admission for infectious disease and mortality. While the pathophysiology behind this phenomenon remains unknown, increasing BMI may influence immune dysregulation.
Collapse
Affiliation(s)
- G Butler-Laporte
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada; Division of Infectious Diseases and Medical Microbiology, McGill University, Montréal, Québec, Canada.
| | - A Harroud
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - V Forgetta
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - J B Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada; Department of Human Genetics, McGill University, Montréal, Québec, Canada; Department of Twin Research, King's College London, London, UK.
| |
Collapse
|
525
|
Casanova F, Tyrrell J, Beaumont RN, Ji Y, Jones SE, Hattersley AT, Weedon MN, Murray A, Shore AC, Frayling TM, Wood AR. A genome-wide association study implicates multiple mechanisms influencing raised urinary albumin-creatinine ratio. Hum Mol Genet 2020; 28:4197-4207. [PMID: 31630189 PMCID: PMC7246045 DOI: 10.1093/hmg/ddz243] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/27/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022] Open
Abstract
Raised albumin–creatinine ratio (ACR) is an indicator of microvascular damage and renal disease. We aimed to identify genetic variants associated with raised ACR and study the implications of carrying multiple ACR-raising alleles with metabolic and vascular-related disease. We performed a genome-wide association study of ACR using 437 027 individuals from the UK Biobank in the discovery phase, 54 527 more than previous studies, and followed up our findings in independent studies. We identified 62 independent associations with ACR across 56 loci (P < 5 × 10–8), of which 20 were not previously reported. Pathway analyses and the identification of 20 of the 62 variants (at r2 > 0.8) coinciding with signals for at least 16 related metabolic and vascular traits, suggested multiple pathways leading to raised ACR levels. After excluding variants at the CUBN locus, known to alter ACR via effects on renal absorption, an ACR genetic risk score was associated with a higher risk of hypertension, and less strongly, type 2 diabetes and stroke. For some rare genotype combinations at the CUBN locus, most individuals had ACR levels above the microalbuminuria clinical threshold. Contrary to our hypothesis, individuals carrying more CUBN ACR-raising alleles, and above the clinical threshold, had a higher frequency of vascular disease. The CUBN allele effects on ACR were twice as strong in people with diabetes—a result robust to an optimization-algorithm approach to simulating interactions, validating previously reported gene–diabetes interactions (P ≤ 4 × 10–5). In conclusion, a variety of genetic mechanisms and traits contribute to variation
in ACR.
Collapse
Affiliation(s)
- Francesco Casanova
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and College of Medicine and Health, University of Exeter, Exeter, UK
| | - Jessica Tyrrell
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and College of Medicine and Health, University of Exeter, Exeter, UK
| | - Robin N Beaumont
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Yingjie Ji
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Samuel E Jones
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Andrew T Hattersley
- Institute of Biomedical & Clinical Science, University of Exeter, Exeter, UK
| | - Michael N Weedon
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Anna Murray
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Angela C Shore
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and College of Medicine and Health, University of Exeter, Exeter, UK
| | - Timothy M Frayling
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Andrew R Wood
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, UK
| |
Collapse
|
526
|
Xie T, Wang B, Nolte IM, van der Most PJ, Oldehinkel AJ, Hartman CA, Snieder H. Genetic Risk Scores for Complex Disease Traits in Youth. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 13:e002775. [PMID: 32527150 PMCID: PMC7439939 DOI: 10.1161/circgen.119.002775] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND For most disease-related traits the magnitude of the contribution of genetic factors in adolescents remains unclear. METHODS Twenty continuous traits related to anthropometry, cardiovascular and renal function, metabolism, and inflammation were selected from the ongoing prospective Tracking Adolescents' Individual Lives Survey cohort in the Netherlands with measurements of up to 5 waves from age 11 to 22 years (n=1354, 47.6% males) and all traits available at the third wave (mean age [SD]=16.22 [0.66]). For each trait, unweighted and weighted genetic risk scores (GRSs) were generated based on significantly associated single nucleotide polymorphisms identified from literature. The variance explained by the GRSs in adolescents were estimated by linear regression after adjustment for covariates. RESULTS Except for ALT (alanine transaminase), all GRSs were significantly associated with their traits. The trait variance explained by the GRSs was highest for lipoprotein[a] (39.59%) and varied between 0.09% (ALT) and 18.49% (LDL [low-density lipoprotein]) for the other traits. For most traits, the variances explained in adolescents were comparable with or slightly smaller than those in adults. Significant increases of trait levels (except ALT) and increased risks for overweight/obesity (odds ratio, 6.41 [95% CI, 2.95-15.56]) and hypertension (odds ratio, 2.86 [95% CI, 1.39-6.17]) were found in individuals in the top GRS decile compared with those at the bottom decile. CONCLUSIONS Variances explained by adult-based GRSs for disease-related traits in adolescents, although still relatively modest, were comparable with or slightly smaller than in adults offering promise for improved risk prediction at early ages.
Collapse
Affiliation(s)
- Tian Xie
- Department of Epidemiology (T.X., B.W., I.M.N., P.J.v.d.M., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Bin Wang
- Department of Epidemiology (T.X., B.W., I.M.N., P.J.v.d.M., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Ilja M Nolte
- Department of Epidemiology (T.X., B.W., I.M.N., P.J.v.d.M., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Peter J van der Most
- Department of Epidemiology (T.X., B.W., I.M.N., P.J.v.d.M., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Albertine J Oldehinkel
- Department of Psychiatry (A.J.O., C.A.H.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Catharina A Hartman
- Department of Psychiatry (A.J.O., C.A.H.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Harold Snieder
- Department of Epidemiology (T.X., B.W., I.M.N., P.J.v.d.M., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| |
Collapse
|
527
|
Zhao JV, Schooling CM. The role of testosterone in chronic kidney disease and kidney function in men and women: a bi-directional Mendelian randomization study in the UK Biobank. BMC Med 2020; 18:122. [PMID: 32493397 PMCID: PMC7271464 DOI: 10.1186/s12916-020-01594-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) has an apparent sex disparity, with a more rapid progress in men than in women. Whether the well-established sex-specific evolutionary biology trade-off between reproduction and longevity might inform CKD has not previously been considered. Relevant evidence from randomized controlled trials (RCTs) is not available. METHODS We used a bi-directional Mendelian randomization study to obtain unconfounded estimates using the UK Biobank. Single nucleotide polymorphisms (SNPs) that strongly (p value < 5 × 10-8) predicted testosterone in a sex-specific manner were applied to 179,916 white British men (6016 CKD cases) and 212,079 white British women (5958 CKD cases) to obtain sex-specific associations with CKD, albuminuria, and estimated glomerular filtration rate (eGFR). We also used multivariable MR to control for sex hormone binding globulin (SHBG). For validation, we similarly examined their role in hemoglobin and high-density lipoprotein cholesterol (HDL-c). We also assessed the role of kidney function in serum testosterone, by applying eGFR-related SNPs to testosterone in the UK Biobank. RESULTS Genetically predicted testosterone was associated with CKD in men (odds ratio (OR) for bioavailable testosterone 1.17 per standard deviation, 95% confidence interval (CI) 1.03 to 1.33) based on 125 SNPs but not in women (OR 1.02, 95% CI 0.92 to 1.14 for total testosterone) based on 254 SNPs. Multivariable MR allowing for SHBG showed consistent patterns. Genetically predicted bioavailable testosterone in men and women and genetically predicted total testosterone in women increased hemoglobin and lowered HDL-c, as seen in RCTs. Genetically predicted eGFR was not related to serum testosterone in men or in women. CONCLUSIONS Genetically predicted testosterone was associated with CKD and worse kidney function in men, whilst not affected by kidney function. Identifying drivers of testosterone and the underlying pathways could provide new insights into CKD prevention and treatment.
Collapse
Affiliation(s)
- Jie V Zhao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Pokfulam, Hong Kong SAR, China.
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Pokfulam, Hong Kong SAR, China.,School of Public Health and Health Policy, City University of New York, New York, NY, USA
| |
Collapse
|
528
|
Genetics of Biochemical Phenotypes. Twin Res Hum Genet 2020; 23:77-79. [PMID: 32482194 DOI: 10.1017/thg.2020.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Biomarkers diagnose, predict or assess the risk of disease, and studies of the effects of genetic variation on biomarker phenotypes in the general population complement studies on patients diagnosed with disease. This paper traces the evolution of studies on biomarker genetics over the past 40 years through examples drawn from the work of Professor Martin and his colleagues.
Collapse
|
529
|
Yu Z, Coresh J, Qi G, Grams M, Boerwinkle E, Snieder H, Teumer A, Pattaro C, Köttgen A, Chatterjee N, Tin A. A bidirectional Mendelian randomization study supports causal effects of kidney function on blood pressure. Kidney Int 2020; 98:708-716. [PMID: 32454124 DOI: 10.1016/j.kint.2020.04.044] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/12/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
Blood pressure and kidney function have a bidirectional relation. Hypertension has long been considered as a risk factor for kidney function decline. However, whether intensive blood pressure control could promote kidney health has been uncertain. The kidney is known to have a major role in affecting blood pressure through sodium extraction and regulating electrolyte balance. This bidirectional relation makes causal inference between these two traits difficult. Therefore, to examine the causal relations between these two traits, we performed two-sample Mendelian randomization analyses using summary statistics of large-scale genome-wide association studies. We selected genetic instruments more likely to be specific for kidney function using meta-analyses of complementary kidney function biomarkers (glomerular filtration rate estimated from serum creatinine [eGFRcr], and blood urea nitrogen from the CKDGen Consortium). Systolic and diastolic blood pressure summary statistics were from the International Consortium for Blood Pressure and UK Biobank. Significant evidence supported the causal effects of higher kidney function on lower blood pressure. Based on the mode-based Mendelian randomization method, the effect estimates for one standard deviation (SD) higher in log-transformed eGFRcr was -0.17 SD unit (95 % confidence interval: -0.09 to -0.24) in systolic blood pressure and -0.15 SD unit (95% confidence interval: -0.07 to -0.22) in diastolic blood pressure. In contrast, the causal effects of blood pressure on kidney function were not statistically significant. Thus, our results support causal effects of higher kidney function on lower blood pressure and suggest preventing kidney function decline can reduce the public health burden of hypertension.
Collapse
Affiliation(s)
- Zhi Yu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Guanghao Qi
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Morgan Grams
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Harold Snieder
- Department of Epidemiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Centre - University of Freiburg, Freiburg, Germany
| | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA; Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| |
Collapse
|
530
|
Levey AS, Titan SM, Powe NR, Coresh J, Inker LA. Kidney Disease, Race, and GFR Estimation. Clin J Am Soc Nephrol 2020; 15:1203-1212. [PMID: 32393465 PMCID: PMC7409747 DOI: 10.2215/cjn.12791019] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Assessment of GFR is central to clinical practice, research, and public health. Current Kidney Disease Improving Global Outcomes guidelines recommend measurement of serum creatinine to estimate GFR as the initial step in GFR evaluation. Serum creatinine is influenced by creatinine metabolism as well as GFR; hence, all equations to estimate GFR from serum creatinine include surrogates for muscle mass, such as age, sex, race, height, or weight. The guideline-recommended equation in adults (the 2009 Chronic Kidney Disease Epidemiology Collaboration creatinine equation) includes a term for race (specified as black versus nonblack), which improves the accuracy of GFR estimation by accounting for differences in non-GFR determinants of serum creatinine by race in the study populations used to develop the equation. In that study, blacks had a 16% higher average measured GFR compared with nonblacks with the same age, sex, and serum creatinine. The reasons for this difference are only partly understood, and the use of race in GFR estimation has limitations. Some have proposed eliminating the race coefficient, but this would induce a systematic underestimation of measured GFR in blacks, with potential unintended consequences at the individual and population levels. We propose a more cautious approach that maintains and improves accuracy of GFR estimates and avoids disadvantaging any racial group. We suggest full disclosure of use of race in GFR estimation, accommodation of those who decline to identify their race, and shared decision making between health care providers and patients. We also suggest mindful use of cystatin C as a confirmatory test as well as clearance measurements. It would be preferable to avoid specification of race in GFR estimation if there was a superior, evidence-based substitute. The goal of future research should be to develop more accurate methods for GFR estimation that do not require use of race or other demographic characteristics.
Collapse
Affiliation(s)
- Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts;
| | - Silvia M Titan
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Neil R Powe
- Department of Medicine, Priscilla Chan and Mark Zuckerberg San Francisco General Hospital and University of California, San Francisco, California; and
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
531
|
Casanova F, Wood AR, Yaghootkar H, Beaumont RN, Jones SE, Gooding KM, Aizawa K, Strain WD, Hattersley AT, Khan F, Shore AC, Frayling TM, Tyrrell J. A Mendelian Randomization Study Provides Evidence That Adiposity and Dyslipidemia Lead to Lower Urinary Albumin-to-Creatinine Ratio, a Marker of Microvascular Function. Diabetes 2020; 69:1072-1082. [PMID: 31915152 PMCID: PMC7611011 DOI: 10.2337/db19-0862] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/23/2019] [Indexed: 01/22/2023]
Abstract
Urinary albumin-to-creatinine ratio (ACR) is a marker of diabetic nephropathy and microvascular damage. Metabolic-related traits are observationally associated with ACR, but their causal role is uncertain. Here, we confirmed ACR as a marker of microvascular damage and tested whether metabolic-related traits have causal relationships with ACR. The association between ACR and microvascular function (responses to acetylcholine [ACH] and sodium nitroprusside) was tested in the SUMMIT study. Two-sample Mendelian randomization (MR) was used to infer the causal effects of 11 metabolic risk factors, including glycemic, lipid, and adiposity traits, on ACR. MR was performed in up to 440,000 UK Biobank and 54,451 CKDGen participants. ACR was robustly associated with microvascular function measures in SUMMIT. Using MR, we inferred that higher triglyceride (TG) and LDL cholesterol (LDL-C) levels caused elevated ACR. A 1 SD higher TG and LDL-C level caused a 0.062 (95% CI 0.040, 0.083) and a 0.026 (95% CI 0.008, 0.044) SD higher ACR, respectively. There was evidence that higher body fat and visceral body fat distribution caused elevated ACR, while a metabolically "favorable adiposity" phenotype lowered ACR. ACR is a valid marker for microvascular function. MR suggested that seven traits have causal effects on ACR, highlighting the role of adiposity-related traits in causing lower microvascular function.
Collapse
Affiliation(s)
- Francesco Casanova
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Andrew R Wood
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - Robert N Beaumont
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Samuel E Jones
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Kim M Gooding
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Kunihiko Aizawa
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - W David Strain
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Andrew T Hattersley
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Faisel Khan
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, U.K
| | - Angela C Shore
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Timothy M Frayling
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Jessica Tyrrell
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K.
| |
Collapse
|
532
|
Garrett MR, Korstanje R. Using Genetic and Species Diversity to Tackle Kidney Disease. Trends Genet 2020; 36:499-509. [PMID: 32362446 DOI: 10.1016/j.tig.2020.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/26/2020] [Accepted: 04/03/2020] [Indexed: 12/18/2022]
Abstract
Progress in the identification of causal genes and understanding of the mechanism underlying kidney disease is hindered by the almost exclusive use of a few animal models with restrictive monogenic backgrounds that may be more resistant to kidney disease compared with humans and, therefore, poor models. Exploring the large genetic diversity in classical animal models, such as mice and rats, and leveraging species diversity will allow us to use the genetic advantages of zebrafish, Drosophila, and other species, to develop both new animal models that are more relevant to the study of human kidney disease and potential therapies.
Collapse
Affiliation(s)
- Michael R Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS, USA; Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, MS, USA; Department of Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Ron Korstanje
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, Maine, ME, USA; Mount Desert Island Biological Laboratory, Bar Harbor, Maine, ME, USA.
| |
Collapse
|
533
|
Fazzini F, Lamina C, Raschenberger J, Schultheiss UT, Kotsis F, Schönherr S, Weissensteiner H, Forer L, Steinbrenner I, Meiselbach H, Bärthlein B, Wanner C, Eckardt KU, Köttgen A, Kronenberg F. Results from the German Chronic Kidney Disease (GCKD) study support association of relative telomere length with mortality in a large cohort of patients with moderate chronic kidney disease. Kidney Int 2020; 98:488-497. [PMID: 32641227 DOI: 10.1016/j.kint.2020.02.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 02/08/2023]
Abstract
Telomere length is known to be inversely associated with aging and has been proposed as a marker for aging-related diseases. Telomere attrition can be accelerated by oxidative stress and inflammation, both commonly present in patients with chronic kidney disease. Here, we investigated whether relative telomere length is associated with mortality in a large cohort of patients with chronic kidney disease stage G3 and A1-3 or G1-2 with overt proteinuria (A3) at enrollment. Relative telomere length was quantified in peripheral blood by a quantitative PCR method in 4,955 patients from the GCKD study, an ongoing prospective observational cohort. Complete four-year follow-up was available from 4,926 patients in whom we recorded 354 deaths. Relative telomere length was a strong and independent predictor of all-cause mortality. Each decrease of 0.1 relative telomere length unit was highly associated with a 14% increased risk of death (hazard ratio1.14 [95% confidence interval 1.06-1.22]) in a model adjusted for age, sex, baseline eGFR, urine albumin/creatinine ratio, diabetes mellitus, prevalent cardiovascular disease, LDL-cholesterol, HDL-cholesterol, smoking, body mass index, systolic and diastolic blood pressure, C-reactive protein and serum albumin. This translated to a 75% higher risk for those in the lowest compared to the highest quartile of relative telomere length. The association was mainly driven by 117 cardiovascular deaths (1.20 [1.05-1.35]) as well as 67 deaths due to infections (1.27 [1.07-1.50]). Thus, our findings support an association of shorter telomere length with all-cause mortality, cardiovascular mortality and death due to infections in patients with moderate chronic kidney disease.
Collapse
Affiliation(s)
- Federica Fazzini
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Julia Raschenberger
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ulla T Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany; Renal Division, Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Fruzsina Kotsis
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany; Renal Division, Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hansi Weissensteiner
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Inga Steinbrenner
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Heike Meiselbach
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Barbara Bärthlein
- Medical Centre for Information and Communication Technology (MIK), University Hospital Erlangen, Erlangen, Germany
| | - Christoph Wanner
- Division of Nephrology, Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria.
| | | |
Collapse
|
534
|
Stewart BJ, Clatworthy MR. Applying single-cell technologies to clinical pathology: progress in nephropathology. J Pathol 2020; 250:693-704. [PMID: 32125696 PMCID: PMC8651001 DOI: 10.1002/path.5417] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 12/13/2022]
Abstract
Cells represent the basic building blocks of living organisms. Accurate characterisation of cellular phenotype, intercellular signalling networks, and the spatial organisation of cells within organs is crucial to deliver a better understanding of the processes underpinning physiology, and the perturbations that lead to disease. Single-cell methodologies have increased rapidly in scale and scope in recent years and are set to generate important insights into human disease. Here, we review current practices in nephropathology, which are dominated by relatively simple morphological descriptions of tissue biopsies based on their appearance using light microscopy. Bulk transcriptomics have more recently been used to explore glomerular and tubulointerstitial kidney disease, renal cancer, and the responses to injury and alloimmunity in kidney transplantation, generating novel disease insights and prognostic biomarkers. These studies set the stage for single-cell transcriptomic approaches that reveal cell-type-specific gene expression patterns in health and disease. These technologies allow genome-wide disease susceptibility genes to be interpreted with the knowledge of the specific cell populations within organs that express them, identifying candidate cell types for further study. Single-cell technologies are also moving beyond assaying individual cellular transcriptomes, to measuring the epigenetic landscape of single cells. Single-cell antigen-receptor gene sequencing also enables specific T- and B-cell clones to be tracked in different tissues and disease states. In the coming years these rich 'multi-omic' descriptions of kidney disease will enable histopathological descriptions to be comprehensively integrated with molecular phenotypes, enabling better disease classification and prognostication and the application of personalised treatment strategies. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Benjamin J Stewart
- Department of MedicineUniversity of CambridgeCambridgeUK
- Cellular GeneticsWellcome Sanger InstituteCambridgeUK
- Cambridge NIHR Biomedical Research CentreAddenbrooke's HospitalCambridgeUK
| | - Menna R Clatworthy
- Department of MedicineUniversity of CambridgeCambridgeUK
- Cellular GeneticsWellcome Sanger InstituteCambridgeUK
- Cambridge NIHR Biomedical Research CentreAddenbrooke's HospitalCambridgeUK
| |
Collapse
|
535
|
Guan Y, Liu H, Ma Z, Li SY, Park J, Sheng X, Susztak K. Dnmt3a and Dnmt3b-Decommissioned Fetal Enhancers are Linked to Kidney Disease. J Am Soc Nephrol 2020; 31:765-782. [PMID: 32127410 PMCID: PMC7191927 DOI: 10.1681/asn.2019080797] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/24/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cytosine methylation is an epigenetic mark that dictates cell fate and response to stimuli. The timing and establishment of methylation logic during kidney development remains unknown. DNA methyltransferase 3a and 3b are the enzymes capable of establishing de novo methylation. METHODS We generated mice with genetic deletion of Dnmt3a and Dnmt3b in nephron progenitor cells (Six2CreDnmt3a/3b) and kidney tubule cells (KspCreDnmt3a/3b). We characterized KspCreDnmt3a/3b mice at baseline and after injury. Unbiased omics profiling, such as whole genome bisulfite sequencing, reduced representation bisulfite sequencing and RNA sequencing were performed on whole-kidney samples and isolated renal tubule cells. RESULTS KspCreDnmt3a/3b mice showed no obvious morphologic and functional alterations at baseline. Knockout animals exhibited increased resistance to cisplatin-induced kidney injury, but not to folic acid-induced fibrosis. Whole-genome bisulfite sequencing indicated that Dnmt3a and Dnmt3b play an important role in methylation of gene regulatory regions that act as fetal-specific enhancers in the developing kidney but are decommissioned in the mature kidney. Loss of Dnmt3a and Dnmt3b resulted in failure to silence developmental genes. We also found that fetal-enhancer regions methylated by Dnmt3a and Dnmt3b were enriched for kidney disease genetic risk loci. Methylation patterns of kidneys from patients with CKD showed defects similar to those in mice with Dnmt3a and Dnmt3b deletion. CONCLUSIONS Our results indicate a potential locus-specific convergence of genetic, epigenetic, and developmental elements in kidney disease development.
Collapse
Affiliation(s)
- Yuting Guan
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hongbo Liu
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ziyuan Ma
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Szu-Yuan Li
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jihwan Park
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xin Sheng
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
536
|
Viggiano D, Wagner CA, Martino G, Nedergaard M, Zoccali C, Unwin R, Capasso G. Mechanisms of cognitive dysfunction in CKD. Nat Rev Nephrol 2020; 16:452-469. [PMID: 32235904 DOI: 10.1038/s41581-020-0266-9] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2020] [Indexed: 02/07/2023]
Abstract
Cognitive impairment is an increasingly recognized major cause of chronic disability and is commonly found in patients with chronic kidney disease (CKD). Knowledge of the relationship between kidney dysfunction and impaired cognition may improve our understanding of other forms of cognitive dysfunction. Patients with CKD are at an increased risk (compared with the general population) of both dementia and its prodrome, mild cognitive impairment (MCI), which are characterized by deficits in executive functions, memory and attention. Brain imaging in patients with CKD has revealed damage to white matter in the prefrontal cortex and, in animal models, in the subcortical monoaminergic and cholinergic systems, accompanied by widespread macrovascular and microvascular damage. Unfortunately, current interventions that target cardiovascular risk factors (such as anti-hypertensive drugs, anti-platelet agents and statins) seem to have little or no effect on CKD-associated MCI, suggesting that the accumulation of uraemic neurotoxins may be more important than disturbed haemodynamic factors or lipid metabolism in MCI pathogenesis. Experimental models show that the brain monoaminergic system is susceptible to uraemic neurotoxins and that this system is responsible for the altered sleep pattern commonly observed in patients with CKD. Neural progenitor cells and the glymphatic system, which are important in Alzheimer disease pathogenesis, may also be involved in CKD-associated MCI. More detailed study of CKD-associated MCI is needed to fully understand its clinical relevance, underlying pathophysiology, possible means of early diagnosis and prevention, and whether there may be novel approaches and potential therapies with wider application to this and other forms of cognitive decline.
Collapse
Affiliation(s)
- Davide Viggiano
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.,Biogem Scarl, Ariano Irpino, Italy
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland, and National Center of Competence in Research NCCR Kidney.CH, Zurich, Switzerland
| | - Gianvito Martino
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Maiken Nedergaard
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY, USA
| | - Carmine Zoccali
- Institute of Clinical Physiology, National Research Council (CNR), Reggio Calabria Unit, Reggio Calabria, Italy
| | - Robert Unwin
- Department of Renal Medicine, University College London (UCL), Royal Free Campus, London, UK.,Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Giovambattista Capasso
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy. .,Biogem Scarl, Ariano Irpino, Italy.
| |
Collapse
|
537
|
Xie J, Liu L, Mladkova N, Li Y, Ren H, Wang W, Cui Z, Lin L, Hu X, Yu X, Xu J, Liu G, Caliskan Y, Sidore C, Balderes O, Rosen RJ, Bodria M, Zanoni F, Zhang JY, Krithivasan P, Mehl K, Marasa M, Khan A, Ozay F, Canetta PA, Bomback AS, Appel GB, Sanna-Cherchi S, Sampson MG, Mariani LH, Perkowska-Ptasinska A, Durlik M, Mucha K, Moszczuk B, Foroncewicz B, Pączek L, Habura I, Ars E, Ballarin J, Mani LY, Vogt B, Ozturk S, Yildiz A, Seyahi N, Arikan H, Koc M, Basturk T, Karahan G, Akgul SU, Sever MS, Zhang D, Santoro D, Bonomini M, Londrino F, Gesualdo L, Reiterova J, Tesar V, Izzi C, Savoldi S, Spotti D, Marcantoni C, Messa P, Galliani M, Roccatello D, Granata S, Zaza G, Lugani F, Ghiggeri G, Pisani I, Allegri L, Sprangers B, Park JH, Cho B, Kim YS, Kim DK, Suzuki H, Amoroso A, Cattran DC, Fervenza FC, Pani A, Hamilton P, Harris S, Gupta S, Cheshire C, Dufek S, Issler N, Pepper RJ, Connolly J, Powis S, Bockenhauer D, Stanescu HC, Ashman N, Loos RJF, Kenny EE, Wuttke M, Eckardt KU, Köttgen A, Hofstra JM, Coenen MJH, Kiemeney LA, et alXie J, Liu L, Mladkova N, Li Y, Ren H, Wang W, Cui Z, Lin L, Hu X, Yu X, Xu J, Liu G, Caliskan Y, Sidore C, Balderes O, Rosen RJ, Bodria M, Zanoni F, Zhang JY, Krithivasan P, Mehl K, Marasa M, Khan A, Ozay F, Canetta PA, Bomback AS, Appel GB, Sanna-Cherchi S, Sampson MG, Mariani LH, Perkowska-Ptasinska A, Durlik M, Mucha K, Moszczuk B, Foroncewicz B, Pączek L, Habura I, Ars E, Ballarin J, Mani LY, Vogt B, Ozturk S, Yildiz A, Seyahi N, Arikan H, Koc M, Basturk T, Karahan G, Akgul SU, Sever MS, Zhang D, Santoro D, Bonomini M, Londrino F, Gesualdo L, Reiterova J, Tesar V, Izzi C, Savoldi S, Spotti D, Marcantoni C, Messa P, Galliani M, Roccatello D, Granata S, Zaza G, Lugani F, Ghiggeri G, Pisani I, Allegri L, Sprangers B, Park JH, Cho B, Kim YS, Kim DK, Suzuki H, Amoroso A, Cattran DC, Fervenza FC, Pani A, Hamilton P, Harris S, Gupta S, Cheshire C, Dufek S, Issler N, Pepper RJ, Connolly J, Powis S, Bockenhauer D, Stanescu HC, Ashman N, Loos RJF, Kenny EE, Wuttke M, Eckardt KU, Köttgen A, Hofstra JM, Coenen MJH, Kiemeney LA, Akilesh S, Kretzler M, Beck LH, Stengel B, Debiec H, Ronco P, Wetzels JFM, Zoledziewska M, Cucca F, Ionita-Laza I, Lee H, Hoxha E, Stahl RAK, Brenchley P, Scolari F, Zhao MH, Gharavi AG, Kleta R, Chen N, Kiryluk K. The genetic architecture of membranous nephropathy and its potential to improve non-invasive diagnosis. Nat Commun 2020; 11:1600. [PMID: 32231244 PMCID: PMC7105485 DOI: 10.1038/s41467-020-15383-w] [Show More Authors] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 03/03/2020] [Indexed: 02/06/2023] Open
Abstract
Membranous Nephropathy (MN) is a rare autoimmune cause of kidney failure. Here we report a genome-wide association study (GWAS) for primary MN in 3,782 cases and 9,038 controls of East Asian and European ancestries. We discover two previously unreported loci, NFKB1 (rs230540, OR = 1.25, P = 3.4 × 10-12) and IRF4 (rs9405192, OR = 1.29, P = 1.4 × 10-14), fine-map the PLA2R1 locus (rs17831251, OR = 2.25, P = 4.7 × 10-103) and report ancestry-specific effects of three classical HLA alleles: DRB1*1501 in East Asians (OR = 3.81, P = 2.0 × 10-49), DQA1*0501 in Europeans (OR = 2.88, P = 5.7 × 10-93), and DRB1*0301 in both ethnicities (OR = 3.50, P = 9.2 × 10-23 and OR = 3.39, P = 5.2 × 10-82, respectively). GWAS loci explain 32% of disease risk in East Asians and 25% in Europeans, and correctly re-classify 20-37% of the cases in validation cohorts that are antibody-negative by the serum anti-PLA2R ELISA diagnostic test. Our findings highlight an unusual genetic architecture of MN, with four loci and their interactions accounting for nearly one-third of the disease risk.
Collapse
Affiliation(s)
- Jingyuan Xie
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lili Liu
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Nikol Mladkova
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Yifu Li
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Hong Ren
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiming Wang
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhao Cui
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijng, China
| | - Li Lin
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofan Hu
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xialian Yu
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xu
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Liu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijng, China
| | - Yasar Caliskan
- Division of Nephrology, Department of Internal Medicine, Istanbul School of Medicine, Istanbul University, Istanbul, Turkey
| | - Carlo Sidore
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Olivia Balderes
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Raphael J Rosen
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Monica Bodria
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Francesca Zanoni
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
- Nephrology Dialysis and Kidney Transplant Unit, Fundazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli studi di Milano, Milan, Italy
| | - Jun Y Zhang
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Priya Krithivasan
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Karla Mehl
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Maddalena Marasa
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Atlas Khan
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Fatih Ozay
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Pietro A Canetta
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Andrew S Bomback
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Gerald B Appel
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Simone Sanna-Cherchi
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Matthew G Sampson
- Department of Pediatrics-Nephrology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Laura H Mariani
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI, USA
- Arbor Research Collaborative for Health, Ann Arbor, MI, USA
| | | | - Magdalena Durlik
- Department of Transplantology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Mucha
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Moszczuk
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Bartosz Foroncewicz
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Ireneusz Habura
- Department of Nephrology, University Hospital of Karol Marcinkowski in Zielona Góra, Zielona Góra, Poland
| | - Elisabet Ars
- Molecular Biology Laboratory and Nephrology Department, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau, Universitat Autònoma de Barcelona, REDINREN, IISCIII, Barcelona, Spain
| | - Jose Ballarin
- Molecular Biology Laboratory and Nephrology Department, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau, Universitat Autònoma de Barcelona, REDINREN, IISCIII, Barcelona, Spain
| | - Laila-Yasmin Mani
- Department of Nephrology and Hypertension, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Savas Ozturk
- Nephrology Clinic, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Abdülmecit Yildiz
- Department of Nephrology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Nurhan Seyahi
- Division of Nephrology, Department of Internal Medicine, Cerrahpasa Medical Faculty, Istanbul University - Cerrahpasa, Istanbul, Turkey
| | - Hakki Arikan
- Division of Nephrology, Department of Internal Medicine, Marmara University School of Medicine, Istanbul, Turkey
| | - Mehmet Koc
- Division of Nephrology, Department of Internal Medicine, Marmara University School of Medicine, Istanbul, Turkey
| | - Taner Basturk
- Department of Nephrology, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey
| | - Gonca Karahan
- Department of Medical Biology, Istanbul School of Medicine, Istanbul University, Istanbul, Turkey
| | - Sebahat Usta Akgul
- Department of Medical Biology, Istanbul School of Medicine, Istanbul University, Istanbul, Turkey
| | - Mehmet Sukru Sever
- Division of Nephrology, Department of Internal Medicine, Istanbul School of Medicine, Istanbul University, Istanbul, Turkey
| | - Dan Zhang
- Department of Nephrology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Domenico Santoro
- Department of Cinical and Experimental Medicine, Unit of Nephrology, University of Messina, Messina, Italy
| | - Mario Bonomini
- Department of Medicine, University of Chieti-Pescara, SS. Annunziata Hospital, Chieti, Italy
| | | | | | - Jana Reiterova
- Department of Nephrology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Vladimir Tesar
- Department of Nephrology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Claudia Izzi
- Second Division of Nephrology, ASST-Spedali Civili di Brescia Presidio di Montichiari, Brescia, Italy
- Department of Obstetrics and Gynecology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Silvana Savoldi
- Unit of Nephrology and Dialysis ASL TO4, Cirié, Turin, Italy
| | | | | | - Piergiorgio Messa
- Nephrology Dialysis and Kidney Transplant Unit, Fundazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli studi di Milano, Milan, Italy
| | | | - Dario Roccatello
- San Giovanni Bosco Hospital (ERK-net Member) and University of Turin, Turin, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Francesca Lugani
- Division of Nephrology, Dialysis, Transplantation, IRCCS Giannina Gaslini, Genoa, Italy
| | - GianMarco Ghiggeri
- Division of Nephrology, Dialysis, Transplantation, IRCCS Giannina Gaslini, Genoa, Italy
| | - Isabella Pisani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Landino Allegri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Ben Sprangers
- Department of Microbiology and Immunology, Laboratory of Molecular Immunology, Rega Institute, KU, Leuven, Belgium
- Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Jin-Ho Park
- Department of Family Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea
| | - BeLong Cho
- Department of Family Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea
- Institute on Aging, Seoul National University College of Medicine, Seoul, Korea
| | - Yon Su Kim
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ki Kim
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hitoshi Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Antonio Amoroso
- Department of Medical Sciences, University of Torino and Immunogenetics and Transplant Biology Service, University Hospital "Città della Salute e della Scienza di Torino", Turin, Italy
| | - Daniel C Cattran
- Department of Nephrology, University of Toronto, Toronto General Hospital, Toronto, ON, Canada
| | | | - Antonello Pani
- Department of Nephrology and Dialysis, G. Brotzu Hospital, Cagliari, Italy
| | - Patrick Hamilton
- Manchester Institute of Nephrology and Transplantation, Manchester University Hospitals NHS Trust, Manchester, UK
| | - Shelly Harris
- Manchester Institute of Nephrology and Transplantation, Manchester University Hospitals NHS Trust, Manchester, UK
| | - Sanjana Gupta
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - Chris Cheshire
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - Stephanie Dufek
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - Naomi Issler
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - Ruth J Pepper
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - John Connolly
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - Stephen Powis
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - Detlef Bockenhauer
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - Horia C Stanescu
- Department of Nephrology, Division of Medicine, University College London, London, UK
| | - Neil Ashman
- Renal Unit, Royal London Hospital, Barts Health, Whitechapel, London, UK
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eimear E Kenny
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New York, NY, USA
- Center for Population Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Dep. of Biometry, Epidemiology, and Medical Bioinformatics, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität, Erlangen, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Dep. of Biometry, Epidemiology, and Medical Bioinformatics, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Julia M Hofstra
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marieke J H Coenen
- Department of Human Genetics, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Lambertus A Kiemeney
- Department of Epidemiology, Biostatistics & HTA, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Shreeram Akilesh
- Department of Anatomic Pathology, University of Washington, Seattle, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lawrence H Beck
- Department of Medicine, Renal Section, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Benedicte Stengel
- Institut National de la Santé et de la Recherche Médicale, Centre for Research in Epidemiology and Population Health, Villejuif, France
- University Paris-Sud, Villejuif, France
| | - Hanna Debiec
- Sorbonne Université, Pierre and Marie Curie University Paris 06, Paris, France
| | - Pierre Ronco
- Sorbonne Université, Pierre and Marie Curie University Paris 06, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) 1155, Paris, France
| | - Jack F M Wetzels
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Magdalena Zoledziewska
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Iuliana Ionita-Laza
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Hajeong Lee
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Elion Hoxha
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rolf A K Stahl
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Brenchley
- Faculty of Biology, Medicine, Health, University of Manchester, Manchester, UK
| | - Francesco Scolari
- Second Division of Nephrology, ASST-Spedali Civili di Brescia Presidio di Montichiari, Brescia, Italy
- University of Brescia, Brescia, Italy
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijng, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Ali G Gharavi
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA
| | - Robert Kleta
- Department of Nephrology, Division of Medicine, University College London, London, UK.
| | - Nan Chen
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, USA.
| |
Collapse
|
538
|
Protecting the kidney in systemic lupus erythematosus: from diagnosis to therapy. Nat Rev Rheumatol 2020; 16:255-267. [PMID: 32203285 DOI: 10.1038/s41584-020-0401-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2020] [Indexed: 12/20/2022]
Abstract
Lupus nephritis (LN) is a common manifestation of systemic lupus erythematosus that can lead to irreversible renal impairment. Although the prognosis of LN has improved substantially over the past 50 years, outcomes have plateaued in the USA in the past 20 years as immunosuppressive therapies have failed to reverse disease in more than half of treated patients. This failure might reflect disease complexity and heterogeneity, as well as social and economic barriers to health-care access that can delay intervention until after damage has already occurred. LN progression is still poorly understood and involves multiple cell types and both immune and non-immune mechanisms. Single-cell analysis of intrinsic renal cells and infiltrating cells from patients with LN is a new approach that will help to define the pathways of renal injury at a cellular level. Although many new immune-modulating therapies are being tested in the clinic, the development of therapies to improve regeneration of the injured kidney and to prevent fibrosis requires a better understanding of the mechanisms of LN progression. This mechanistic understanding, together with the development of clinical measures to evaluate risk and detect early disease and better access to expert health-care providers, should improve outcomes for patients with LN.
Collapse
|
539
|
Siedlinski M, Jozefczuk E, Xu X, Teumer A, Evangelou E, Schnabel RB, Welsh P, Maffia P, Erdmann J, Tomaszewski M, Caulfield MJ, Sattar N, Holmes MV, Guzik TJ. White Blood Cells and Blood Pressure: A Mendelian Randomization Study. Circulation 2020; 141:1307-1317. [PMID: 32148083 PMCID: PMC7176352 DOI: 10.1161/circulationaha.119.045102] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND High blood pressure (BP) is a risk factor for cardiovascular morbidity and mortality. While BP is regulated by the function of kidney, vasculature, and sympathetic nervous system, recent experimental data suggest that immune cells may play a role in hypertension. METHODS We studied the relationship between major white blood cell types and blood pressure in the UK Biobank population and used Mendelian randomization (MR) analyses using the ≈750 000 UK-Biobank/International Consortium of Blood Pressure-Genome-Wide Association Studies to examine which leukocyte populations may be causally linked to BP. RESULTS A positive association between quintiles of lymphocyte, monocyte, and neutrophil counts, and increased systolic BP, diastolic BP, and pulse pressure was observed (eg, adjusted systolic BP mean±SE for 1st versus 5th quintile respectively: 140.13±0.08 versus 141.62±0.07 mm Hg for lymphocyte, 139.51±0.08 versus 141.84±0.07 mm Hg for monocyte, and 137.96±0.08 versus 142.71±0.07 mm Hg for neutrophil counts; all P<10-50). Using 121 single nucleotide polymorphisms in MR, implemented through the inverse-variance weighted approach, we identified a potential causal relationship of lymphocyte count with systolic BP and diastolic BP (causal estimates: 0.69 [95% CI, 0.19-1.20] and 0.56 [95% CI, 0.23-0.90] of mm Hg per 1 SD genetically elevated lymphocyte count, respectively), which was directionally concordant to the observational findings. These inverse-variance weighted estimates were consistent with other robust MR methods. The exclusion of rs3184504 SNP in the SH2B3 locus attenuated the magnitude of the signal in some of the MR analyses. MR in the reverse direction found evidence of positive effects of BP indices on counts of monocytes, neutrophils, and eosinophils but not lymphocytes or basophils. Subsequent MR testing of lymphocyte count in the context of genetic correlation with renal function or resting and postexercise heart rate demonstrated a positive association of lymphocyte count with urine albumin-to-creatinine ratio. CONCLUSIONS Observational and genetic analyses demonstrate a concordant, positive and potentially causal relationship of lymphocyte count with systolic BP and diastolic BP.
Collapse
Affiliation(s)
- Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland (M.S., E.J., T.J.G.).,Institute of Cardiovascular and Medical Sciences (M.S., P.W., N.S., T.J.G.), University of Glasgow, United Kingdom
| | - Ewelina Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland (M.S., E.J., T.J.G.)
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom (X.X., M.T.)
| | - Alexander Teumer
- Department SHIP/Clinical-Epidemiological Research, Institute for Community Medicine, University Medicine Greifswald, Germany (A.T.).,German Centre for Cardiovascular Research partner site Greifswald, Germany (A.T.)
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (E.E.)
| | - Renate B Schnabel
- University Heart Center Hamburg Eppendorf, German Center for Cardiovascular Research partner site Hamburg/Kiel/Lübeck, Germany (R.B.S.)
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences (M.S., P.W., N.S., T.J.G.), University of Glasgow, United Kingdom
| | - Pasquale Maffia
- Institute of Infection, Immunity, and Inflammation (P.M.), University of Glasgow, United Kingdom.,Department of Pharmacy, University of Naples Federico II, Italy (P.M.)
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Germany (J.E.)
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom (X.X., M.T.)
| | - Mark J Caulfield
- William Harvey Research Institute, National Institute for Health Research Biomedical Research Centre at Barts, Queen Mary University of London, United Kingdom (M.J.C.)
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences (M.S., P.W., N.S., T.J.G.), University of Glasgow, United Kingdom
| | - Michael V Holmes
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom (M.V.H.)
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland (M.S., E.J., T.J.G.).,Institute of Cardiovascular and Medical Sciences (M.S., P.W., N.S., T.J.G.), University of Glasgow, United Kingdom
| |
Collapse
|
540
|
Dhande IS, Doris PA. Pulling the Hood off Genetic Susceptibility to Hypertensive Renal Disease. J Am Soc Nephrol 2020; 31:667-668. [PMID: 32123053 DOI: 10.1681/asn.2020020139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Isha S Dhande
- Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Peter A Doris
- Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
541
|
Abstract
Background: Uncertainty in the mechanism and directionality of observational associations between thyroid function and kidney function may be addressed by genetic analysis with an instrumental variable method termed bidirectional Mendelian randomization (MR). Methods: In the Women's Genome Health Study (WGHS), observational associations between thyroid measures and kidney function were evaluated. Genetic instruments for MR were from recent genome-wide association studies (GWAS) of hypothyroidism, thyrotropin (TSH), and free thyroxine (fT4) concentrations within the reference range, thyroid peroxidase antibodies (TPOAb), estimated glomerular filtration rate from creatinine (eGFRcrea), eGFR from cystatin C (eGFRcys), and chronic kidney disease (CKD). In WGHS individual-level data, these instruments were used for bidirectional MR between thyroid (N = 3336) and kidney (N = 23,186) functions. To increase power, MR was also performed using GWAS summary statistics from the Chronic Kidney Disease Genetics Consortium (CKDGen) for eGFRcrea (N = 567,460), eGFRcys (N = 24,063), CKD [N(total) = 480,698, N(cases) = 41,395], and urinary albumin/creatinine ratio (UACR/N = 54,450). Results: In the WGHS, hypothyroidism was observationally associated with decreased eGFRcrea [beta (standard error, SE): -0.024 (0.009) ln(mL/min/1.73 m2), p = 0.01]. By MR, hypothyroidism was associated with decreased eGFRcrea in the WGHS [beta (SE): -0.007 (0.002) per doubled odds hypothyroidism, p = 1.7 × 10-3] and in CKDGen [beta (SE): -0.004 (0.0005), p = 2.0 × 10-22], and robust to sensitivity analysis. Hypothyroidism was also associated by MR with increased CKD in CKDGen (odds ratio, OR [confidence interval, CI]: 1.05 [1.03-1.08], p = 3.3 × 10-5), but not in the WGHS (OR [CI]: 1.02 [0.95-1.10], p = 0.57). Increased TSH within the reference range had an MR association with increased eGFRcrea in the WGHS [beta (SE): -0.018 (0.007) ln(mL/min/1.73 m2)/standard deviation, SD, p = 6.5 × 10-3] and CKDGen [beta (SE): -0.008 (0.001) ln(mL/min/1.73 m2)/SD, p = 6.8 × 10-17], and with CKD in CKDGen (OR [CI]: 1.10 [1.04-1.15], p = 3.1 × 10-4). There were no MR associations of hypothyroidism or TSH with eGFRcys or UACR, and MR associations of fT4 in the reference range with kidney function were inconsistent in both the WGHS and CKDGen. However, by MR in CKDGen, TPOAb were robustly associated with decreased eGFRcrea [beta (SE): -0.041 (0.009), p = 6.2 × 10-6] and decreased eGFRcys [beta (SE): -0.294 (0.065), p = 6.2 × 10-6]. TPOAb were less robustly associated with CKD but not associated with UACR. In reverse MR in the WGHS, kidney function was not consistently associated with thyroid function. Conclusions: Bidirectional MR supports a directional association from hypothyroidism, increased TSH, and TPOAb, but not fT4, to decreased eGFRcrea and increased CKD.
Collapse
Affiliation(s)
- Christina Ellervik
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, Massachusetts
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Christina Ellervik, MD, PhD, DMSci, Department of Laboratory Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 ;
| | - Samia Mora
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Lipid Metabolomics, Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Paul M. Ridker
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Lipid Metabolomics, Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Epidemiology, T.H. Chan School of Public Health, Boston, Massachusetts
| | - Daniel I. Chasman
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Lipid Metabolomics, Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Address correspondence to: Daniel I. Chasman, PhD, Division of Preventive Medicine, Brigham and Women's Hospital, 900 Commonwealth Avenue, Boston, MA 02215
| |
Collapse
|
542
|
How many more data is required to give the kidney the attention it deserves? Time to act for the “Big Five” of cardiovascular risk. Atherosclerosis 2020; 297:146-148. [DOI: 10.1016/j.atherosclerosis.2020.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/28/2020] [Indexed: 11/20/2022]
|
543
|
Epidemiology research to foster improvement in chronic kidney disease care. Kidney Int 2020; 97:477-486. [DOI: 10.1016/j.kint.2019.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/12/2019] [Accepted: 11/15/2019] [Indexed: 11/24/2022]
|
544
|
Fan F, Geurts AM, Pabbidi MR, Ge Y, Zhang C, Wang S, Liu Y, Gao W, Guo Y, Li L, He X, Lv W, Muroya Y, Hirata T, Prokop J, Booz GW, Jacob HJ, Roman RJ. A Mutation in γ-Adducin Impairs Autoregulation of Renal Blood Flow and Promotes the Development of Kidney Disease. J Am Soc Nephrol 2020; 31:687-700. [PMID: 32029431 DOI: 10.1681/asn.2019080784] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/14/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The genes and mechanisms involved in the association between diabetes or hypertension and CKD risk are unclear. Previous studies have implicated a role for γ-adducin (ADD3), a cytoskeletal protein encoded by Add3. METHODS We investigated renal vascular function in vitro and in vivo and the susceptibility to CKD in rats with wild-type or mutated Add3 and in genetically modified rats with overexpression or knockout of ADD3. We also studied glomeruli and primary renal vascular smooth muscle cells isolated from these rats. RESULTS This study identified a K572Q mutation in ADD3 in fawn-hooded hypertensive (FHH) rats-a mutation previously reported in Milan normotensive (MNS) rats that also develop kidney disease. Using molecular dynamic simulations, we found that this mutation destabilizes a critical ADD3-ACTIN binding site. A reduction of ADD3 expression in membrane fractions prepared from the kidney and renal vascular smooth muscle cells of FHH rats was associated with the disruption of the F-actin cytoskeleton. Compared with renal vascular smooth muscle cells from Add3 transgenic rats, those from FHH rats had elevated membrane expression of BKα and BK channel current. FHH and Add3 knockout rats exhibited impairments in the myogenic response of afferent arterioles and in renal blood flow autoregulation, which were rescued in Add3 transgenic rats. We confirmed these findings in a genetic complementation study that involved crossing FHH and MNS rats that share the ADD3 mutation. Add3 transgenic rats showed attenuation of proteinuria, glomerular injury, and kidney fibrosis with aging and mineralocorticoid-induced hypertension. CONCLUSIONS This is the first report that a mutation in ADD3 that alters ACTIN binding causes renal vascular dysfunction and promotes the susceptibility to kidney disease.
Collapse
Affiliation(s)
- Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ying Ge
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Chao Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Wenjun Gao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ya Guo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Longyang Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Xiaochen He
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Wenshan Lv
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yoshikazu Muroya
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Takashi Hirata
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jeremy Prokop
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Howard J Jacob
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi;
| |
Collapse
|
545
|
Köttgen A, Pattaro C. The CKDGen Consortium: ten years of insights into the genetic basis of kidney function. Kidney Int 2020; 97:236-242. [DOI: 10.1016/j.kint.2019.10.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 01/08/2023]
|
546
|
Abstract
The kidney is subject to a wide range of abnormalities, many of which have a significant hereditable component. Next generation sequencing is increasingly bringing the genetic drivers of Mendelian disease into focus at the base pair level, whereas inexpensive genotyping arrays have surveyed hundreds of thousands of individuals to identify common variants that predispose to kidney dysfunction. In this first article in a CJASN series on kidney genomics, we review how both rare and common variants contribute to kidney disease, explore how evolution may influence the genetic variants that affect kidney function, consider how genetic information is and will be used in the clinic, and identify some of the most important future directions for kidney disease research. Forthcoming articles in the series will elaborate on many of these themes.
Collapse
Affiliation(s)
- Martin R Pollak
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - David J Friedman
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
547
|
Bedin M, Boyer O, Servais A, Li Y, Villoing-Gaudé L, Tête MJ, Cambier A, Hogan J, Baudouin V, Krid S, Bensman A, Lammens F, Louillet F, Ranchin B, Vigneau C, Bouteau I, Isnard-Bagnis C, Mache CJ, Schäfer T, Pape L, Gödel M, Huber TB, Benz M, Klaus G, Hansen M, Latta K, Gribouval O, Morinière V, Tournant C, Grohmann M, Kuhn E, Wagner T, Bole-Feysot C, Jabot-Hanin F, Nitschké P, Ahluwalia TS, Köttgen A, Andersen CBF, Bergmann C, Antignac C, Simons M. Human C-terminal CUBN variants associate with chronic proteinuria and normal renal function. J Clin Invest 2020; 130:335-344. [PMID: 31613795 PMCID: PMC6934218 DOI: 10.1172/jci129937] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/02/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUNDProteinuria is considered an unfavorable clinical condition that accelerates renal and cardiovascular disease. However, it is not clear whether all forms of proteinuria are damaging. Mutations in CUBN cause Imerslund-Gräsbeck syndrome (IGS), which is characterized by intestinal malabsorption of vitamin B12 and in some cases proteinuria. CUBN encodes for cubilin, an intestinal and proximal tubular uptake receptor containing 27 CUB domains for ligand binding.METHODSWe used next-generation sequencing for renal disease genes to genotype cohorts of patients with suspected hereditary renal disease and chronic proteinuria. CUBN variants were analyzed using bioinformatics, structural modeling, and epidemiological methods.RESULTSWe identified 39 patients, in whom biallelic pathogenic variants in the CUBN gene were associated with chronic isolated proteinuria and early childhood onset. Since the proteinuria in these patients had a high proportion of albuminuria, glomerular diseases such as steroid-resistant nephrotic syndrome or Alport syndrome were often the primary clinical diagnosis, motivating renal biopsies and the use of proteinuria-lowering treatments. However, renal function was normal in all cases. By contrast, we did not found any biallelic CUBN variants in proteinuric patients with reduced renal function or focal segmental glomerulosclerosis. Unlike the more N-terminal IGS mutations, 37 of the 41 proteinuria-associated CUBN variants led to modifications or truncations after the vitamin B12-binding domain. Finally, we show that 4 C-terminal CUBN variants are associated with albuminuria and slightly increased GFR in meta-analyses of large population-based cohorts.CONCLUSIONCollectively, our data suggest an important role for the C-terminal half of cubilin in renal albumin reabsorption. Albuminuria due to reduced cubilin function could be an unexpectedly common benign condition in humans that may not require any proteinuria-lowering treatment or renal biopsy.FUNDINGATIP-Avenir program, Fondation Bettencourt-Schueller (Liliane Bettencourt Chair of Developmental Biology), Agence Nationale de la Recherche (ANR) Investissements d'avenir program (ANR-10-IAHU-01) and NEPHROFLY (ANR-14-ACHN-0013, to MS), Steno Collaborative Grant 2018 (NNF18OC0052457, to TSA and MS), Heisenberg Professorship of the German Research Foundation (KO 3598/5-1, to AK), Deutsche Forschungsgemeinschaft (DFG) Collaborative Research Centre (SFB) KIDGEM 1140 (project 246781735, to CB), and Federal Ministry of Education and Research (BMB) (01GM1515C, to CB).
Collapse
Affiliation(s)
| | - Olivia Boyer
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
- Department of Pediatric Nephrology and
| | - Aude Servais
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
- Department of Nephrology, Necker Hospital, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | | | - Marie-Josephe Tête
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
| | - Alexandra Cambier
- Department of Pediatric Nephrology and Transplantation, Robert-Debré Hospital, APHP, Paris, France
| | - Julien Hogan
- Department of Pediatric Nephrology and Transplantation, Robert-Debré Hospital, APHP, Paris, France
| | - Veronique Baudouin
- Department of Pediatric Nephrology and Transplantation, Robert-Debré Hospital, APHP, Paris, France
| | | | | | - Florie Lammens
- Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | | | - Bruno Ranchin
- Department of Pediatric Nephrology, Hospices Civils de Lyon, Bron, France
| | - Cecile Vigneau
- Centre Hospitalier Universitaire de Rennes, INSERM U1085 IRSET-9, Rennes, France
| | - Iseline Bouteau
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | | | | | - Tobias Schäfer
- Renal Division, University Medical Center Freiburg, Freiburg, Germany
| | - Lars Pape
- Department of Pediatric Kidney, Liver and Metabolic Disease, Hannover Medical School, Hannover, Germany
| | - Markus Gödel
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B. Huber
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Günter Klaus
- Department of Child and Adolescent Medicine, University Medical Center Marburg-Giessen, Marburg, Germany
| | - Matthias Hansen
- KfH-Nierenzentrum für Kinder und Jugendliche und Clementine-Kinderhospital, Frankfurt, Germany
| | - Kay Latta
- KfH-Nierenzentrum für Kinder und Jugendliche und Clementine-Kinderhospital, Frankfurt, Germany
| | - Olivier Gribouval
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
| | | | - Carole Tournant
- Department of Genetics, Necker Hospital, APHP, Paris, France
| | - Maik Grohmann
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
- Center for Human Genetics, Mainz, Germany
| | - Elisa Kuhn
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
| | - Timo Wagner
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
| | - Christine Bole-Feysot
- Bioinformatic Platform, INSERM UMR 1163, Institut Imagine, Paris, France
- Bioinformatics Core Facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Université de Paris, Paris, France
| | - Fabienne Jabot-Hanin
- Bioinformatic Platform, INSERM UMR 1163, Institut Imagine, Paris, France
- Bioinformatics Core Facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Université de Paris, Paris, France
| | - Patrick Nitschké
- Bioinformatic Platform, INSERM UMR 1163, Institut Imagine, Paris, France
- Bioinformatics Core Facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Université de Paris, Paris, France
| | | | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | | | - Carsten Bergmann
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
- Center for Human Genetics, Mainz, Germany
- Renal Division, Department of Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
- Department of Genetics, Necker Hospital, APHP, Paris, France
| | | |
Collapse
|
548
|
Bedin M, Boyer O, Servais A, Li Y, Villoing-Gaudé L, Tête MJ, Cambier A, Hogan J, Baudouin V, Krid S, Bensman A, Lammens F, Louillet F, Ranchin B, Vigneau C, Bouteau I, Isnard-Bagnis C, Mache CJ, Schäfer T, Pape L, Gödel M, Huber TB, Benz M, Klaus G, Hansen M, Latta K, Gribouval O, Morinière V, Tournant C, Grohmann M, Kuhn E, Wagner T, Bole-Feysot C, Jabot-Hanin F, Nitschké P, Ahluwalia TS, Köttgen A, Andersen CBF, Bergmann C, Antignac C, Simons M. Human C-terminal CUBN variants associate with chronic proteinuria and normal renal function. J Clin Invest 2020. [PMID: 31613795 DOI: 10.1172/jci12937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023] Open
Abstract
BACKGROUNDProteinuria is considered an unfavorable clinical condition that accelerates renal and cardiovascular disease. However, it is not clear whether all forms of proteinuria are damaging. Mutations in CUBN cause Imerslund-Gräsbeck syndrome (IGS), which is characterized by intestinal malabsorption of vitamin B12 and in some cases proteinuria. CUBN encodes for cubilin, an intestinal and proximal tubular uptake receptor containing 27 CUB domains for ligand binding.METHODSWe used next-generation sequencing for renal disease genes to genotype cohorts of patients with suspected hereditary renal disease and chronic proteinuria. CUBN variants were analyzed using bioinformatics, structural modeling, and epidemiological methods.RESULTSWe identified 39 patients, in whom biallelic pathogenic variants in the CUBN gene were associated with chronic isolated proteinuria and early childhood onset. Since the proteinuria in these patients had a high proportion of albuminuria, glomerular diseases such as steroid-resistant nephrotic syndrome or Alport syndrome were often the primary clinical diagnosis, motivating renal biopsies and the use of proteinuria-lowering treatments. However, renal function was normal in all cases. By contrast, we did not found any biallelic CUBN variants in proteinuric patients with reduced renal function or focal segmental glomerulosclerosis. Unlike the more N-terminal IGS mutations, 37 of the 41 proteinuria-associated CUBN variants led to modifications or truncations after the vitamin B12-binding domain. Finally, we show that 4 C-terminal CUBN variants are associated with albuminuria and slightly increased GFR in meta-analyses of large population-based cohorts.CONCLUSIONCollectively, our data suggest an important role for the C-terminal half of cubilin in renal albumin reabsorption. Albuminuria due to reduced cubilin function could be an unexpectedly common benign condition in humans that may not require any proteinuria-lowering treatment or renal biopsy.FUNDINGATIP-Avenir program, Fondation Bettencourt-Schueller (Liliane Bettencourt Chair of Developmental Biology), Agence Nationale de la Recherche (ANR) Investissements d'avenir program (ANR-10-IAHU-01) and NEPHROFLY (ANR-14-ACHN-0013, to MS), Steno Collaborative Grant 2018 (NNF18OC0052457, to TSA and MS), Heisenberg Professorship of the German Research Foundation (KO 3598/5-1, to AK), Deutsche Forschungsgemeinschaft (DFG) Collaborative Research Centre (SFB) KIDGEM 1140 (project 246781735, to CB), and Federal Ministry of Education and Research (BMB) (01GM1515C, to CB).
Collapse
Affiliation(s)
| | - Olivia Boyer
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
- Department of Pediatric Nephrology and
| | - Aude Servais
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
- Department of Nephrology, Necker Hospital, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | | | - Marie-Josephe Tête
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
| | - Alexandra Cambier
- Department of Pediatric Nephrology and Transplantation, Robert-Debré Hospital, APHP, Paris, France
| | - Julien Hogan
- Department of Pediatric Nephrology and Transplantation, Robert-Debré Hospital, APHP, Paris, France
| | - Veronique Baudouin
- Department of Pediatric Nephrology and Transplantation, Robert-Debré Hospital, APHP, Paris, France
| | | | | | - Florie Lammens
- Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | | | - Bruno Ranchin
- Department of Pediatric Nephrology, Hospices Civils de Lyon, Bron, France
| | - Cecile Vigneau
- Centre Hospitalier Universitaire de Rennes, INSERM U1085 IRSET-9, Rennes, France
| | - Iseline Bouteau
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | | | | | - Tobias Schäfer
- Renal Division, University Medical Center Freiburg, Freiburg, Germany
| | - Lars Pape
- Department of Pediatric Kidney, Liver and Metabolic Disease, Hannover Medical School, Hannover, Germany
| | - Markus Gödel
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Günter Klaus
- Department of Child and Adolescent Medicine, University Medical Center Marburg-Giessen, Marburg, Germany
| | - Matthias Hansen
- KfH-Nierenzentrum für Kinder und Jugendliche und Clementine-Kinderhospital, Frankfurt, Germany
| | - Kay Latta
- KfH-Nierenzentrum für Kinder und Jugendliche und Clementine-Kinderhospital, Frankfurt, Germany
| | - Olivier Gribouval
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
| | | | - Carole Tournant
- Department of Genetics, Necker Hospital, APHP, Paris, France
| | - Maik Grohmann
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
- Center for Human Genetics, Mainz, Germany
| | - Elisa Kuhn
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
| | - Timo Wagner
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
| | - Christine Bole-Feysot
- Bioinformatic Platform, INSERM UMR 1163, Institut Imagine, Paris, France
- Bioinformatics Core Facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Université de Paris, Paris, France
| | - Fabienne Jabot-Hanin
- Bioinformatic Platform, INSERM UMR 1163, Institut Imagine, Paris, France
- Bioinformatics Core Facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Université de Paris, Paris, France
| | - Patrick Nitschké
- Bioinformatic Platform, INSERM UMR 1163, Institut Imagine, Paris, France
- Bioinformatics Core Facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Université de Paris, Paris, France
| | | | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | | | - Carsten Bergmann
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
- Center for Human Genetics, Mainz, Germany
- Renal Division, Department of Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Disease, Imagine Institute, INSERM U1163, Université de Paris, Paris, France
- Department of Genetics, Necker Hospital, APHP, Paris, France
| | | |
Collapse
|
549
|
The Urinary Excretion of Uromodulin is Regulated by the Potassium Channel ROMK. Sci Rep 2019; 9:19517. [PMID: 31863061 PMCID: PMC6925250 DOI: 10.1038/s41598-019-55771-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
Uromodulin, the most abundant protein in normal urine, is produced by cells lining the thick ascending limb (TAL) of the loop of Henle. Uromodulin regulates the activity of the potassium channel ROMK in TAL cells. Common variants in KCNJ1, the gene encoding ROMK, are associated with urinary levels of uromodulin in population studies. Here, we investigated the functional link between ROMK and uromodulin in Kcnj1 knock-out mouse models, in primary cultures of mouse TAL (mTAL) cells, and in patients with Bartter syndrome due to KCNJ1 mutations. Both global and kidney-specific Kcnj1 knock-out mice showed reduced urinary levels of uromodulin paralleled by increased levels in the kidney, compared to wild-type controls. Pharmacological inhibition and genetic deletion of ROMK in mTAL cells caused a reduction in apical uromodulin excretion, reflected by cellular accumulation. In contrast, NKCC2 inhibition showed no effect on uromodulin processing. Patients with Bartter syndrome type 2 showed reduced urinary uromodulin levels compared to age and gender matched controls. These results demonstrate that ROMK directly regulates processing and release of uromodulin by TAL cells, independently from NKCC2. They support the functional link between transport activity and uromodulin in the TAL, relevant for blood pressure control and urinary concentrating ability.
Collapse
|
550
|
Sedor JR, Freedman BI. Biologic Underpinnings of Type 1 Diabetic Kidney Disease. J Am Soc Nephrol 2019; 30:1782-1783. [PMID: 31570541 DOI: 10.1681/asn.2019080803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- John R Sedor
- Department of Nephrology and Hypertension, Glickman Urology and Kidney Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - Barry I Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|