51
|
Yang N, Shi L, Xu P, Ren F, Lv S, Li C, Qi X. Identification of potential drug targets for insomnia by Mendelian randomization analysis based on plasma proteomics. Front Neurol 2024; 15:1380321. [PMID: 38725646 PMCID: PMC11079244 DOI: 10.3389/fneur.2024.1380321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Insomnia, a common clinical disorder, significantly impacts the physical and mental well-being of patients. Currently, available hypnotic medications are unsatisfactory due to adverse reactions and dependency, necessitating the identification of new drug targets for the treatment of insomnia. Methods In this study, we utilized 734 plasma proteins as genetic instruments obtained from genome-wide association studies to conduct a Mendelian randomization analysis, with insomnia as the outcome variable, to identify potential drug targets for insomnia. Additionally, we validated our results externally using other datasets. Sensitivity analyses entailed reverse Mendelian randomization analysis, Bayesian co-localization analysis, and phenotype scanning. Furthermore, we constructed a protein-protein interaction network to elucidate potential correlations between the identified proteins and existing targets. Results Mendelian randomization analysis indicated that elevated levels of TGFBI (OR = 1.01; 95% CI, 1.01-1.02) and PAM ((OR = 1.01; 95% CI, 1.01-1.02) in plasma are associated with an increased risk of insomnia, with external validation supporting these findings. Moreover, there was no evidence of reverse causality for these two proteins. Co-localization analysis confirmed that PAM (coloc.abf-PPH4 = 0.823) shared the same variant with insomnia, further substantiating its potential role as a therapeutic target. There are interactive relationships between the potential proteins and existing targets of insomnia. Conclusion Overall, our findings suggested that elevated plasma levels of TGFBI and PAM are connected with an increased risk of insomnia and might be promising therapeutic targets, particularly PAM. However, further exploration is necessary to fully understand the underlying mechanisms involved.
Collapse
Affiliation(s)
- Ni Yang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liangyuan Shi
- Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital) Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Pengfei Xu
- Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital) Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Fang Ren
- Department of Laboratory, Jimo District Qingdao Hospital of Traditional Chinese Medicine, Qingdao, China
| | - Shimeng Lv
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunlin Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianghua Qi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
52
|
Schmit SL, Tsai YY, Bonner JD, Sanz-Pamplona R, Joshi AD, Ugai T, Lindsey SS, Melas M, McDonnell KJ, Idos GE, Walker CP, Qu C, Kast WM, Da Silva DM, Glickman JN, Chan AT, Giannakis M, Nowak JA, Rennert HS, Robins HS, Ogino S, Greenson JK, Moreno V, Rennert G, Gruber SB. Germline genetic regulation of the colorectal tumor immune microenvironment. BMC Genomics 2024; 25:409. [PMID: 38664626 PMCID: PMC11046907 DOI: 10.1186/s12864-024-10295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVE To evaluate the contribution of germline genetics to regulating the briskness and diversity of T cell responses in CRC, we conducted a genome-wide association study to examine the associations between germline genetic variation and quantitative measures of T cell landscapes in 2,876 colorectal tumors from participants in the Molecular Epidemiology of Colorectal Cancer Study (MECC). METHODS Germline DNA samples were genotyped and imputed using genome-wide arrays. Tumor DNA samples were extracted from paraffin blocks, and T cell receptor clonality and abundance were quantified by immunoSEQ (Adaptive Biotechnologies, Seattle, WA). Tumor infiltrating lymphocytes per high powered field (TILs/hpf) were scored by a gastrointestinal pathologist. Regression models were used to evaluate the associations between each variant and the three T-cell features, adjusting for sex, age, genotyping platform, and global ancestry. Three independent datasets were used for replication. RESULTS We identified a SNP (rs4918567) near RBM20 associated with clonality at a genome-wide significant threshold of 5 × 10- 8, with a consistent direction of association in both discovery and replication datasets. Expression quantitative trait (eQTL) analyses and in silico functional annotation for these loci provided insights into potential functional roles, including a statistically significant eQTL between the T allele at rs4918567 and higher expression of ADRA2A (P = 0.012) in healthy colon mucosa. CONCLUSIONS Our study suggests that germline genetic variation is associated with the quantity and diversity of adaptive immune responses in CRC. Further studies are warranted to replicate these findings in additional samples and to investigate functional genomic mechanisms.
Collapse
Affiliation(s)
- Stephanie L Schmit
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA.
- Population and Cancer Prevention Program, Case Comprehensive Cancer Center, Cleveland, OH, USA.
| | - Ya-Yu Tsai
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Joseph D Bonner
- Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Rebeca Sanz-Pamplona
- Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Amit D Joshi
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Tomotaka Ugai
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sidney S Lindsey
- Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Marilena Melas
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Kevin J McDonnell
- Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Gregory E Idos
- Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Christopher P Walker
- Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Chenxu Qu
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - W Martin Kast
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Diane M Da Silva
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | | | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Marios Giannakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hedy S Rennert
- B. Rappaport Faculty of Medicine, Technion and the Association for Promotion of Research in Precision Medicine (APRPM), Haifa, Israel
| | | | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Tokyo Medical and Dental University (Institute of Science Tokyo), Tokyo, Japan
| | - Joel K Greenson
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Victor Moreno
- Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Gad Rennert
- B. Rappaport Faculty of Medicine, Technion and the Association for Promotion of Research in Precision Medicine (APRPM), Haifa, Israel
| | - Stephen B Gruber
- Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
53
|
Weaver C, Nam A, Settle C, Overton M, Giddens M, Richardson KP, Piver R, Mysona DP, Rungruang B, Ghamande S, McIndoe R, Purohit S. Serum Proteomic Signatures in Cervical Cancer: Current Status and Future Directions. Cancers (Basel) 2024; 16:1629. [PMID: 38730581 PMCID: PMC11083044 DOI: 10.3390/cancers16091629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
In 2020, the World Health Organization (WHO) reported 604,000 new diagnoses of cervical cancer (CC) worldwide, and over 300,000 CC-related fatalities. The vast majority of CC cases are caused by persistent human papillomavirus (HPV) infections. HPV-related CC incidence and mortality rates have declined worldwide because of increased HPV vaccination and CC screening with the Papanicolaou test (PAP test). Despite these significant improvements, developing countries face difficulty implementing these programs, while developed nations are challenged with identifying HPV-independent cases. Molecular and proteomic information obtained from blood or tumor samples have a strong potential to provide information on malignancy progression and response to therapy in CC. There is a large amount of published biomarker data related to CC available but the extensive validation required by the FDA approval for clinical use is lacking. The ability of researchers to use the big data obtained from clinical studies and to draw meaningful relationships from these data are two obstacles that must be overcome for implementation into clinical practice. We report on identified multimarker panels of serum proteomic studies in CC for the past 5 years, the potential for modern computational biology efforts, and the utilization of nationwide biobanks to bridge the gap between multivariate protein signature development and the prediction of clinically relevant CC patient outcomes.
Collapse
Affiliation(s)
- Chaston Weaver
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (C.W.); (K.P.R.); (R.P.); (D.P.M.); (R.M.)
| | - Alisha Nam
- Department of Undergraduate Health Professions, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA; (A.N.); (C.S.); (M.O.); (M.G.)
| | - Caitlin Settle
- Department of Undergraduate Health Professions, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA; (A.N.); (C.S.); (M.O.); (M.G.)
| | - Madelyn Overton
- Department of Undergraduate Health Professions, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA; (A.N.); (C.S.); (M.O.); (M.G.)
| | - Maya Giddens
- Department of Undergraduate Health Professions, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA; (A.N.); (C.S.); (M.O.); (M.G.)
| | - Katherine P. Richardson
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (C.W.); (K.P.R.); (R.P.); (D.P.M.); (R.M.)
| | - Rachael Piver
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (C.W.); (K.P.R.); (R.P.); (D.P.M.); (R.M.)
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (B.R.); (S.G.)
| | - David P. Mysona
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (C.W.); (K.P.R.); (R.P.); (D.P.M.); (R.M.)
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (B.R.); (S.G.)
| | - Bunja Rungruang
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (B.R.); (S.G.)
| | - Sharad Ghamande
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (B.R.); (S.G.)
| | - Richard McIndoe
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (C.W.); (K.P.R.); (R.P.); (D.P.M.); (R.M.)
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (B.R.); (S.G.)
| | - Sharad Purohit
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (C.W.); (K.P.R.); (R.P.); (D.P.M.); (R.M.)
- Department of Undergraduate Health Professions, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA; (A.N.); (C.S.); (M.O.); (M.G.)
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (B.R.); (S.G.)
| |
Collapse
|
54
|
Su Z, Wan Q. Potential therapeutic targets for membranous nephropathy: proteome-wide Mendelian randomization and colocalization analysis. Front Immunol 2024; 15:1342912. [PMID: 38707900 PMCID: PMC11069303 DOI: 10.3389/fimmu.2024.1342912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/21/2024] [Indexed: 05/07/2024] Open
Abstract
Background The currently available medications for treating membranous nephropathy (MN) still have unsatisfactory efficacy in inhibiting disease recurrence, slowing down its progression, and even halting the development of end-stage renal disease. There is still a need to develop novel drugs targeting MN. Methods We utilized summary statistics of MN from the Kiryluk Lab and obtained plasma protein data from Zheng et al. We performed a Bidirectional Mendelian randomization analysis, HEIDI test, mediation analysis, Bayesian colocalization, phenotype scanning, drug bank analysis, and protein-protein interaction network. Results The Mendelian randomization analysis uncovered 8 distinct proteins associated with MN after multiple false discovery rate corrections. Proteins related to an increased risk of MN in plasma include ABO [(Histo-Blood Group Abo System Transferase) (WR OR = 1.12, 95%CI:1.05-1.19, FDR=0.09, PPH4 = 0.79)], VWF [(Von Willebrand Factor) (WR OR = 1.41, 95%CI:1.16-1.72, FDR=0.02, PPH4 = 0.81)] and CD209 [(Cd209 Antigen) (WR OR = 1.19, 95%CI:1.07-1.31, FDR=0.09, PPH4 = 0.78)], and proteins that have a protective effect on MN: HRG [(Histidine-Rich Glycoprotein) (WR OR = 0.84, 95%CI:0.76-0.93, FDR=0.02, PPH4 = 0.80)], CD27 [(Cd27 Antigen) (WR OR = 0.78, 95%CI:0.68-0.90, FDR=0.02, PPH4 = 0.80)], LRPPRC [(Leucine-Rich Ppr Motif-Containing Protein, Mitochondrial) (WR OR = 0.79, 95%CI:0.69-0.91, FDR=0.09, PPH4 = 0.80)], TIMP4 [(Metalloproteinase Inhibitor 4) (WR OR = 0.67, 95%CI:0.53-0.84, FDR=0.09, PPH4 = 0.79)] and MAP2K4 [(Dual Specificity Mitogen-Activated Protein Kinase Kinase 4) (WR OR = 0.82, 95%CI:0.72-0.92, FDR=0.09, PPH4 = 0.80)]. ABO, HRG, and TIMP4 successfully passed the HEIDI test. None of these proteins exhibited a reverse causal relationship. Bayesian colocalization analysis provided evidence that all of them share variants with MN. We identified type 1 diabetes, trunk fat, and asthma as having intermediate effects in these pathways. Conclusions Our comprehensive analysis indicates a causal effect of ABO, CD27, VWF, HRG, CD209, LRPPRC, MAP2K4, and TIMP4 at the genetically determined circulating levels on the risk of MN. These proteins can potentially be a promising therapeutic target for the treatment of MN.
Collapse
Affiliation(s)
| | - Qijun Wan
- Department of Nephrology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
55
|
Cao M, Cui B. Clinically relevant plasma proteome for adiposity depots: evidence from systematic mendelian randomization and colocalization analyses. Cardiovasc Diabetol 2024; 23:126. [PMID: 38614964 PMCID: PMC11016216 DOI: 10.1186/s12933-024-02222-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/31/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND The accumulation of visceral and ectopic fat comprise a major cause of cardiometabolic diseases. However, novel drug targets for reducing unnecessary visceral and ectopic fat are still limited. Our study aims to provide a comprehensive investigation of the causal effects of the plasma proteome on visceral and ectopic fat using Mendelian randomization (MR) approach. METHODS We performed two-sample MR analyses based on five large genome-wide association study (GWAS) summary statistics of 2656 plasma proteins, to screen for causal associations of these proteins with traits of visceral and ectopic fat in over 30,000 participants of European ancestry, as well as to assess mediation effects by risk factors of outcomes. The colocalization analysis was conducted to examine whether the identified proteins and outcomes shared casual variants. RESULTS Genetically predicted levels of 14 circulating proteins were associated with visceral and ectopic fat (P < 4.99 × 10- 5, at a Bonferroni-corrected threshold). Colocalization analysis prioritized ten protein targets that showed effect on outcomes, including FST, SIRT2, DNAJB9, IL6R, CTSA, RGMB, PNLIPRP1, FLT4, PPY and IL6ST. MR analyses revealed seven risk factors for visceral and ectopic fat (P < 0.0024). Furthermore, the associations of CTSA, DNAJB9 and IGFBP1 with primary outcomes were mediated by HDL-C and SHBG. Sensitivity analyses showed little evidence of pleiotropy. CONCLUSIONS Our study identified candidate proteins showing putative causal effects as potential therapeutic targets for visceral and ectopic fat accumulation and outlined causal pathways for further prevention of downstream cardiometabolic diseases.
Collapse
Affiliation(s)
- Min Cao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bin Cui
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
56
|
Kalnapenkis A, Jõeloo M, Lepik K, Kukuškina V, Kals M, Alasoo K, Mägi R, Esko T, Võsa U. Genetic determinants of plasma protein levels in the Estonian population. Sci Rep 2024; 14:7694. [PMID: 38565889 PMCID: PMC10987560 DOI: 10.1038/s41598-024-57966-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
The proteome holds great potential as an intermediate layer between the genome and phenome. Previous protein quantitative trait locus studies have focused mainly on describing the effects of common genetic variations on the proteome. Here, we assessed the impact of the common and rare genetic variations as well as the copy number variants (CNVs) on 326 plasma proteins measured in up to 500 individuals. We identified 184 cis and 94 trans signals for 157 protein traits, which were further fine-mapped to credible sets for 101 cis and 87 trans signals for 151 proteins. Rare genetic variation contributed to the levels of 7 proteins, with 5 cis and 14 trans associations. CNVs were associated with the levels of 11 proteins (7 cis and 5 trans), examples including a 3q12.1 deletion acting as a hub for multiple trans associations; and a CNV overlapping NAIP, a sensor component of the NAIP-NLRC4 inflammasome which is affecting pro-inflammatory cytokine interleukin 18 levels. In summary, this work presents a comprehensive resource of genetic variation affecting the plasma protein levels and provides the interpretation of identified effects.
Collapse
Affiliation(s)
- Anette Kalnapenkis
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia.
| | - Maarja Jõeloo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Kaido Lepik
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Viktorija Kukuškina
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mart Kals
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kaur Alasoo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
| | - Urmo Võsa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
| |
Collapse
|
57
|
Zhang W, Ma L, Zhou Q, Gu T, Zhang X, Xing H. Therapeutic Targets for Diabetic Kidney Disease: Proteome-Wide Mendelian Randomization and Colocalization Analyses. Diabetes 2024; 73:618-627. [PMID: 38211557 PMCID: PMC10958583 DOI: 10.2337/db23-0564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/23/2023] [Indexed: 01/13/2024]
Abstract
At present, safe and effective treatment drugs are urgently needed for diabetic kidney disease (DKD). Circulating protein biomarkers with causal genetic evidence represent promising drug targets, which provides an opportunity to identify new therapeutic targets. Summary data from two protein quantitative trait loci studies are presented, one involving 4,907 plasma proteins data from 35,559 individuals and the other encompassing 4,657 plasma proteins among 7,213 European Americans. Summary statistics for DKD were obtained from a large genome-wide association study (3,345 cases and 2,372 controls) and the FinnGen study (3,676 cases and 283,456 controls). Mendelian randomization (MR) analysis was conducted to examine the potential targets for DKD. The colocalization analysis was used to detect whether the potential proteins exist in the shared causal variants. To enhance the credibility of the results, external validation was conducted. Additionally, enrichment analysis, assessment of protein druggability, and the protein-protein interaction networks were used to further enrich the research findings. The proteome-wide MR analyses identified 21 blood proteins that may causally be associated with DKD. Colocalization analysis further supported a causal relationship between 12 proteins and DKD, with external validation confirming 4 of these proteins, and TGFBI was affirmed through two separate group data sets. These results indicate that targeting these four proteins could be a promising approach for treating DKD, and warrant further clinical investigations. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Wei Zhang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Leilei Ma
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qianyi Zhou
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Tianjiao Gu
- Department of Endocrinology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaotian Zhang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Haitao Xing
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
58
|
Goldberg DT, Yaskolka Meir A, Tsaban G, Rinott E, Kaplan A, Zelicha H, Klöting N, Ceglarek U, Iserman B, Shelef I, Rosen P, Blüher M, Stumvoll M, Etzion O, Stampfer MJ, Hu FB, Shai I. Novel proteomic signatures may indicate MRI-assessed intrahepatic fat state and changes: The DIRECT PLUS clinical trial. Hepatology 2024:01515467-990000000-00821. [PMID: 38537153 DOI: 10.1097/hep.0000000000000867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/03/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND AND AIMS We demonstrated in the randomized 18-month DIRECT PLUS trial (n = 294) that a Mediterranean (MED) diet, supplemented with polyphenol-rich Mankai duckweed, green tea, and walnuts and restricted in red/processed meat, caused substantial intrahepatic fat (IHF%) loss compared with 2 other healthy diets, reducing NAFLD by half, regardless of similar weight loss. Here, we investigated the baseline proteomic profile associated with IHF% and the changes in proteomics associated with IHF% changes induced by lifestyle intervention. APPROACH AND RESULTS We calculated IHF% by proton magnetic resonance spectroscopy (normal IHF% <5% and abnormal IHF% ≥5%). We assayed baseline and 18-month samples for 95 proteomic biomarkers.Participants (age = 51.3 ± 10.8 y; 89% men; and body mass index = 31.3 ± 3.9 kg/m 2 ) had an 89.8% 18-month retention rate; 83% had eligible follow-up proteomics measurements, and 78% had follow-up proton magnetic resonance spectroscopy. At baseline, 39 candidate proteins were significantly associated with IHF% (false discovery rate <0.05), mostly related to immune function pathways (eg, hydroxyacid oxidase 1). An IHF% prediction based on the DIRECT PLUS by combined model ( R2 = 0.47, root mean square error = 1.05) successfully predicted IHF% ( R2 = 0.53) during testing and was stronger than separately inputting proteins/traditional markers ( R2 = 0.43/0.44). The 18-month lifestyle intervention induced changes in 18 of the 39 candidate proteins, which were significantly associated with IHF% change, with proteins related to metabolism, extracellular matrix remodeling, and immune function pathways. Thrombospondin-2 protein change was higher in the green-MED compared to the MED group, beyond weight and IHF% loss ( p = 0.01). Protein principal component analysis revealed differences in the third principal component time distinct interactions across abnormal/normal IHF% trajectory combinations; p < 0.05 for all). CONCLUSIONS Our findings suggest novel proteomic signatures that may indicate MRI-assessed IHF state and changes during lifestyle intervention. Specifically, carbonic anhydrase 5A, hydroxyacid oxidase 1, and thrombospondin-2 protein changes are independently associated with IHF% change, and thrombospondin-2 protein change is greater in the green-MED/high polyphenols diet.
Collapse
Affiliation(s)
- Dana T Goldberg
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anat Yaskolka Meir
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gal Tsaban
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ehud Rinott
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Kaplan
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hila Zelicha
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nora Klöting
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Berend Iserman
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Ilan Shelef
- Department of Diagnostic Imaging, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Philip Rosen
- Department of Diagnostic Imaging, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Ohad Etzion
- Department of Gastroenterology and Liver Diseases, Soroka University Medical Center, Beersheba, Israel
| | - Meir J Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Iris Shai
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
59
|
Li J, Wang F, Li Z, Feng J, Men Y, Han J, Xia J, Zhang C, Han Y, Chen T, Zhao Y, Zhou S, Da Y, Chai G, Hao J. Integrative multi-omics analysis identifies genetically supported druggable targets and immune cell specificity for myasthenia gravis. J Transl Med 2024; 22:302. [PMID: 38521921 PMCID: PMC10960998 DOI: 10.1186/s12967-024-04994-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/12/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Myasthenia gravis (MG) is a chronic autoimmune disorder characterized by fluctuating muscle weakness. Despite the availability of established therapies, the management of MG symptoms remains suboptimal, partially attributed to lack of efficacy or intolerable side-effects. Therefore, new effective drugs are warranted for treatment of MG. METHODS By employing an analytical framework that combines Mendelian randomization (MR) and colocalization analysis, we estimate the causal effects of blood druggable expression quantitative trait loci (eQTLs) and protein quantitative trait loci (pQTLs) on the susceptibility of MG. We subsequently investigated whether potential genetic effects exhibit cell-type specificity by utilizing genetic colocalization analysis to assess the interplay between immune-cell-specific eQTLs and MG risk. RESULTS We identified significant MR results for four genes (CDC42BPB, CD226, PRSS36, and TNFSF12) using cis-eQTL genetic instruments and three proteins (CTSH, PRSS8, and CPN2) using cis-pQTL genetic instruments. Six of these loci demonstrated evidence of colocalization with MG susceptibility (posterior probability > 0.80). We next undertook genetic colocalization to investigate cell-type-specific effects at these loci. Notably, we identified robust evidence of colocalization, with a posterior probability of 0.854, linking CTSH expression in TH2 cells and MG risk. CONCLUSIONS This study provides crucial insights into the genetic and molecular factors associated with MG susceptibility, singling out CTSH as a potential candidate for in-depth investigation and clinical consideration. It additionally sheds light on the immune-cell regulatory mechanisms related to the disease. However, further research is imperative to validate these targets and evaluate their feasibility for drug development.
Collapse
Affiliation(s)
- Jiao Li
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
- Beijing Municipal Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Fei Wang
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Zhen Li
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Jingjing Feng
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Yi Men
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Jinming Han
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Jiangwei Xia
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Chen Zhang
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Yilai Han
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Teng Chen
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Yinan Zhao
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Sirui Zhou
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Guoliang Chai
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China.
- Beijing Municipal Geriatric Medical Research Center, Beijing, China.
| | - Junwei Hao
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China.
- Beijing Municipal Geriatric Medical Research Center, Beijing, China.
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China.
| |
Collapse
|
60
|
Austin TR, Fink HA, Jalal DI, Törnqvist AE, Buzkova P, Barzilay JI, Lu T, Carbone L, Gabrielsen ME, Grahnemo L, Hveem K, Jonasson C, Kizer JR, Langhammer A, Mukamal KJ, Gerszten RE, Nethander M, Psaty BM, Robbins JA, Sun YV, Skogholt AH, Åsvold BO, Valderrabano RJ, Zheng J, Richards JB, Coward E, Ohlsson C. Large-scale circulating proteome association study (CPAS) meta-analysis identifies circulating proteins and pathways predicting incident hip fractures. J Bone Miner Res 2024; 39:139-149. [PMID: 38477735 PMCID: PMC11070286 DOI: 10.1093/jbmr/zjad011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 03/14/2024]
Abstract
Hip fractures are associated with significant disability, high cost, and mortality. However, the exact biological mechanisms underlying susceptibility to hip fractures remain incompletely understood. In an exploratory search of the underlying biology as reflected through the circulating proteome, we performed a comprehensive Circulating Proteome Association Study (CPAS) meta-analysis for incident hip fractures. Analyses included 6430 subjects from two prospective cohort studies (Cardiovascular Health Study and Trøndelag Health Study) with circulating proteomics data (aptamer-based 5 K SomaScan version 4.0 assay; 4979 aptamers). Associations between circulating protein levels and incident hip fractures were estimated for each cohort using age and sex-adjusted Cox regression models. Participants experienced 643 incident hip fractures. Compared with the individual studies, inverse-variance weighted meta-analyses yielded more statistically significant associations, identifying 23 aptamers associated with incident hip fractures (conservative Bonferroni correction 0.05/4979, P < 1.0 × 10-5). The aptamers most strongly associated with hip fracture risk corresponded to two proteins of the growth hormone/insulin growth factor system (GHR and IGFBP2), as well as GDF15 and EGFR. High levels of several inflammation-related proteins (CD14, CXCL12, MMP12, ITIH3) were also associated with increased hip fracture risk. Ingenuity pathway analysis identified reduced LXR/RXR activation and increased acute phase response signaling to be overrepresented among those proteins associated with increased hip fracture risk. These analyses identified several circulating proteins and pathways consistently associated with incident hip fractures. These findings underscore the usefulness of the meta-analytic approach for comprehensive CPAS in a similar manner as has previously been observed for large-scale human genetic studies. Future studies should investigate the underlying biology of these potential novel drug targets.
Collapse
Affiliation(s)
- Thomas R Austin
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, 98195, United States
| | - Howard A Fink
- Geriatric Research Education and Clinical Center, VA Health Care System, Minneapolis, MN, 56401, United States
| | - Diana I Jalal
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, Iowa City, IA, 52242, United States
- Iowa City VA Medical Center, Iowa City, IA, 52246, United States
| | - Anna E Törnqvist
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Petra Buzkova
- Department of Biostatistics, University of Washington, Seattle, WA, 98115, United States
| | - Joshua I Barzilay
- Division of Endocrinology, Kaiser Permanente of Georgia, Atlanta, GA, 30339, United States
| | - Tianyuan Lu
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
- Quantitative Life Sciences Program, McGill University, Montreal, Quebec, H3G 0B1, Canada
- 5 Prime Sciences Inc, Montreal, Quebec, H3Y 2W4, Canada
| | - Laura Carbone
- Charlie Norwood VAMC, Augusta, GA, 30901, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Maiken E Gabrielsen
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Louise Grahnemo
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Kristian Hveem
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
- HUNT Research Centre, NTNU, 7600, Levanger, Norway
| | - Christian Jonasson
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Jorge R Kizer
- Cardiology Section, San Francisco VA Health Care System, San Francisco, CA, 94121, United States
- Department of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94158, United States
| | - Arnulf Langhammer
- HUNT Research Centre, NTNU, 7600, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, 7600, Levanger, Norway
| | - Kenneth J Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Center, Brookline, MA, 2446, United States
| | - Robert E Gerszten
- Department of Medicine, Beth Israel Deaconess Medical Center, Brookline, MA, 2446, United States
| | - Maria Nethander
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
- Bioinformatics and Data Center, Sahlgrenska Academy, University of Gothenburg, 413 90, Gothenburg, Sweden
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, 98195, United States
- Departments of Medicine, Epidemiology, and Health Systems and Population Health, University of Washington, Seattle, WA, 98195, United States
| | - John A Robbins
- Department of Medicine, University of California, Davis, CA, 95817, United States
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, United States
| | - Anne Heidi Skogholt
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Bjørn Olav Åsvold
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, 7491, Trondheim, Norway
| | - Rodrigo J Valderrabano
- Research Program in Men’s Health, Aging and Metabolism, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, 2130, United States
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Shanghai, 200025, China
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai Digital Medicine Innovation Center, Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Shanghai, 200025, China
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, United Kingdom
| | - J Brent Richards
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
- 5 Prime Sciences Inc, Montreal, Quebec, H3Y 2W4, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
- Department of Twin Research, King’s College London, London, SE1 7EH, United Kingdom
| | - Eivind Coward
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
- Department of Drug Treatment, Region Västra Götaland, Sahlgrenska University Hospital, 413 45, Gothenburg, Sweden
| |
Collapse
|
61
|
Suhre K. Genetic associations with ratios between protein levels detect new pQTLs and reveal protein-protein interactions. CELL GENOMICS 2024; 4:100506. [PMID: 38412862 PMCID: PMC10943581 DOI: 10.1016/j.xgen.2024.100506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/25/2023] [Accepted: 01/26/2024] [Indexed: 02/29/2024]
Abstract
Protein quantitative trait loci (pQTLs) are an invaluable source of information for drug target development because they provide genetic evidence to support protein function, suggest relationships between cis- and trans-associated proteins, and link proteins to disease endpoints. Using Olink proteomics data for 1,463 proteins measured in over 54,000 samples of the UK Biobank, we identified 4,248 associations with 2,821 ratios between protein levels (rQTLs). rQTLs were 7.6-fold enriched in known protein-protein interactions, suggesting that their ratios reflect biological links between the implicated proteins. Conducting a GWAS on ratios increased the number of discovered genetic signals by 24.7%. The approach can identify novel loci of clinical relevance, support causal gene identification, and reveal complex networks of interacting proteins. Taken together, our study adds significant value to the genetic insights that can be derived from the UKB proteomics data and motivates the wider use of ratios in large-scale GWAS.
Collapse
Affiliation(s)
- Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha 24144, Qatar; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
62
|
Wittich H, Ardlie K, Taylor KD, Durda P, Liu Y, Mikhaylova A, Gignoux CR, Cho MH, Rich SS, Rotter JI, Manichaikul A, Im HK, Wheeler HE. Transcriptome-wide association study of the plasma proteome reveals cis and trans regulatory mechanisms underlying complex traits. Am J Hum Genet 2024; 111:445-455. [PMID: 38320554 PMCID: PMC10940016 DOI: 10.1016/j.ajhg.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Regulation of transcription and translation are mechanisms through which genetic variants affect complex traits. Expression quantitative trait locus (eQTL) studies have been more successful at identifying cis-eQTL (within 1 Mb of the transcription start site) than trans-eQTL. Here, we tested the cis component of gene expression for association with observed plasma protein levels to identify cis- and trans-acting genes that regulate protein levels. We used transcriptome prediction models from 49 Genotype-Tissue Expression (GTEx) Project tissues to predict the cis component of gene expression and tested the predicted expression of every gene in every tissue for association with the observed abundance of 3,622 plasma proteins measured in 3,301 individuals from the INTERVAL study. We tested significant results for replication in 971 individuals from the Trans-omics for Precision Medicine (TOPMed) Multi-Ethnic Study of Atherosclerosis (MESA). We found 1,168 and 1,210 cis- and trans-acting associations that replicated in TOPMed (FDR < 0.05) with a median expected true positive rate (π1) across tissues of 0.806 and 0.390, respectively. The target proteins of trans-acting genes were enriched for transcription factor binding sites and autoimmune diseases in the GWAS catalog. Furthermore, we found a higher correlation between predicted expression and protein levels of the same underlying gene (R = 0.17) than observed expression (R = 0.10, p = 7.50 × 10-11). This indicates the cis-acting genetically regulated (heritable) component of gene expression is more consistent across tissues than total observed expression (genetics + environment) and is useful in uncovering the function of SNPs associated with complex traits.
Collapse
Affiliation(s)
- Henry Wittich
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL 60660, USA
| | - Kristin Ardlie
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Peter Durda
- Laboratory for Clinical Biochemistry Research, University of Vermont, Colchester, VT 05446, USA
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anna Mikhaylova
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Chris R Gignoux
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Stephen S Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA 22908, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA 22908, USA
| | - Hae Kyung Im
- Section of Genetic Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Heather E Wheeler
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL 60660, USA; Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA.
| |
Collapse
|
63
|
Benson M, Smelik M, Li X, Loscalzo J, Sysoev O, Mahmud F, Aly DM, Zhao Y. An interactive atlas of genomic, proteomic, and metabolomic biomarkers promotes the potential of proteins to predict complex diseases. RESEARCH SQUARE 2024:rs.3.rs-3921099. [PMID: 38496611 PMCID: PMC10942575 DOI: 10.21203/rs.3.rs-3921099/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Multiomics analyses have identified multiple potential biomarkers of the incidence and prevalence of complex diseases. However, it is not known which type of biomarker is optimal for clinical purposes. Here, we make a systematic comparison of 90 million genetic variants, 1,453 proteins, and 325 metabolites from 500,000 individuals with complex diseases from the UK Biobank. A machine learning pipeline consisting of data cleaning, data imputation, feature selection, and model training using cross-validation and comparison of the results on holdout test sets showed that proteins were most predictive, followed by metabolites, and genetic variants. Only five proteins per disease resulted in median (min-max) areas under the receiver operating characteristic curves for incidence of 0.79 (0.65-0.86) and 0.84 (0.70-0.91) for prevalence. In summary, our work suggests the potential of predicting complex diseases based on a limited number of proteins. We provide an interactive atlas (macd.shinyapps.io/ShinyApp/) to find genomic, proteomic, or metabolomic biomarkers for different complex diseases.
Collapse
|
64
|
Xu J, Cai X, Miao Z, Yan Y, Chen D, Yang Z, Yue L, Hu W, Zhuo L, Wang J, Xue Z, Fu Y, Xu Y, Zheng J, Guo T, Chen Y. Proteome-wide profiling reveals dysregulated molecular features and accelerated aging in osteoporosis: A 9.8-year prospective study. Aging Cell 2024; 23:e14035. [PMID: 37970652 PMCID: PMC10861190 DOI: 10.1111/acel.14035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/17/2023] Open
Abstract
The role of circulatory proteomics in osteoporosis is unclear. Proteome-wide profiling holds the potential to offer mechanistic insights into osteoporosis. Serum proteome with 413 proteins was profiled by liquid chromatography-tandem mass spectrometry (LC-MS/MS) at baseline, and the 2nd, and 3rd follow-ups (7704 person-tests) in the prospective Chinese cohorts with 9.8 follow-up years: discovery cohort (n = 1785) and internal validation cohort (n = 1630). Bone mineral density (BMD) was measured using dual-energy X-ray absorptiometry (DXA) at follow-ups 1 through 3 at lumbar spine (LS) and femoral neck (FN). We used the Light Gradient Boosting Machine (LightGBM) to identify the osteoporosis (OP)-related proteomic features. The relationships between serum proteins and BMD in the two cohorts were estimated by linear mixed-effects model (LMM). Meta-analysis was then performed to explore the combined associations. We identified 53 proteins associated with osteoporosis using LightGBM, and a meta-analysis showed that 22 of these proteins illuminated a significant correlation with BMD (p < 0.05). The most common proteins among them were PHLD, SAMP, PEDF, HPTR, APOA1, SHBG, CO6, A2MG, CBPN, RAIN APOD, and THBG. The identified proteins were used to generate the biological age (BA) of bone. Each 1 SD-year increase in KDM-Proage was associated with higher risk of LS-OP (hazard ratio [HR], 1.25; 95% CI, 1.14-1.36, p = 4.96 × 10-06 ), and FN-OP (HR, 1.13; 95% CI, 1.02-1.23, p = 9.71 × 10-03 ). The findings uncovered that the apolipoproteins, zymoproteins, complements, and binding proteins presented new mechanistic insights into osteoporosis. Serum proteomics could be a crucial indicator for evaluating bone aging.
Collapse
Affiliation(s)
- Jinjian Xu
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Xue Cai
- School of Life SciencesWestlake UniversityHangzhouChina
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
| | - Zelei Miao
- School of Life SciencesWestlake UniversityHangzhouChina
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
| | - Yan Yan
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Danyu Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Zhen‐xiao Yang
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Liang Yue
- School of Life SciencesWestlake UniversityHangzhouChina
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
| | - Wei Hu
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Laibao Zhuo
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Jia‐ting Wang
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Zhangzhi Xue
- School of Life SciencesWestlake UniversityHangzhouChina
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
| | - Yuanqing Fu
- School of Life SciencesWestlake UniversityHangzhouChina
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
| | - Ying Xu
- Shenzhen Bao'an Center for Chronic Diseases ControlShenzhenChina
| | - Ju‐Sheng Zheng
- School of Life SciencesWestlake UniversityHangzhouChina
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
| | - Tiannan Guo
- School of Life SciencesWestlake UniversityHangzhouChina
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
| | - Yu‐ming Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
65
|
Zilinskas R, Li C, Shen X, Pan W, Yang T. Inferring a directed acyclic graph of phenotypes from GWAS summary statistics. Biometrics 2024; 80:ujad039. [PMID: 38470257 PMCID: PMC10928990 DOI: 10.1093/biomtc/ujad039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 11/24/2023] [Accepted: 01/04/2024] [Indexed: 03/13/2024]
Abstract
Estimating phenotype networks is a growing field in computational biology. It deepens the understanding of disease etiology and is useful in many applications. In this study, we present a method that constructs a phenotype network by assuming a Gaussian linear structure model embedding a directed acyclic graph (DAG). We utilize genetic variants as instrumental variables and show how our method only requires access to summary statistics from a genome-wide association study (GWAS) and a reference panel of genotype data. Besides estimation, a distinct feature of the method is its summary statistics-based likelihood ratio test on directed edges. We applied our method to estimate a causal network of 29 cardiovascular-related proteins and linked the estimated network to Alzheimer's disease (AD). A simulation study was conducted to demonstrate the effectiveness of this method. An R package sumdag implementing the proposed method, all relevant code, and a Shiny application are available.
Collapse
Affiliation(s)
| | - Chunlin Li
- Department of Statistics, Iowa State University, Ames, IA 50011, United States
| | - Xiaotong Shen
- School of Statistics, University of Minnesota, Minneapolis, MN 55455, United States
| | - Wei Pan
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN 55455, United States
| | - Tianzhong Yang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN 55455, United States
| |
Collapse
|
66
|
Kuku KO, Oyetoro R, Hashemian M, Livinski AA, Shearer JJ, Joo J, Psaty BM, Levy D, Ganz P, Roger VL. Proteomics for heart failure risk stratification: a systematic review. BMC Med 2024; 22:34. [PMID: 38273315 PMCID: PMC10809595 DOI: 10.1186/s12916-024-03249-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/05/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a complex clinical syndrome with persistently high mortality. High-throughput proteomic technologies offer new opportunities to improve HF risk stratification, but their contribution remains to be clearly defined. We aimed to systematically review prognostic studies using high-throughput proteomics to identify protein signatures associated with HF mortality. METHODS We searched four databases and two clinical trial registries for articles published from 2012 to 2023. HF proteomics studies measuring high numbers of proteins using aptamer or antibody-based affinity platforms on human plasma or serum with outcomes of all-cause or cardiovascular death were included. Two reviewers independently screened articles, extracted data, and assessed the risk of bias. A third reviewer resolved conflicts. We assessed the risk of bias using the Risk Of Bias In Non-randomized Studies-of Exposure tool. RESULTS Out of 5131 unique articles identified, nine articles were included in the review. The nine studies were observational; three used the aptamer platform, and six used the antibody platform. We found considerable heterogeneity across studies in measurement panels, HF definitions, ejection fraction categorization, follow-up duration, and outcome definitions, and a lack of risk estimates for most protein associations. Hence, we proceeded with a systematic review rather than a meta-analysis. In two comparable aptamer studies in patients with HF with reduced ejection fraction, 21 proteins were identified in common for the association with all-cause death. Among these, one protein, WAP four-disulfide core domain protein 2 was also reported in an antibody study on HFrEF and for the association with CV death. We proposed standardized reporting criteria to facilitate the interpretation of future studies. CONCLUSIONS In this systematic review of nine studies evaluating the association of proteomics with mortality in HF, we identified a limited number of proteins common across several studies. Heterogeneity across studies compromised drawing broad inferences, underscoring the importance of standardized approaches to reporting.
Collapse
Affiliation(s)
- Kayode O Kuku
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca Oyetoro
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maryam Hashemian
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alicia A Livinski
- Office of Research Services, Office of the Director, National Institutes of Health Library, National Institutes of Health, Bethesda, MD, USA
| | - Joseph J Shearer
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jungnam Joo
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Daniel Levy
- Laboratory for Cardiovascular Epidemiology and Genomics, Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter Ganz
- Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, USA
| | - Véronique L Roger
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
67
|
Duijvelaar E, Gisby J, Peters JE, Bogaard HJ, Aman J. Longitudinal plasma proteomics reveals biomarkers of alveolar-capillary barrier disruption in critically ill COVID-19 patients. Nat Commun 2024; 15:744. [PMID: 38272877 PMCID: PMC10811341 DOI: 10.1038/s41467-024-44986-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
The pathobiology of respiratory failure in COVID-19 consists of a complex interplay between viral cytopathic effects and a dysregulated host immune response. In critically ill patients, imatinib treatment demonstrated potential for reducing invasive ventilation duration and mortality. Here, we perform longitudinal profiling of 6385 plasma proteins in 318 hospitalised patients to investigate the biological processes involved in critical COVID-19, and assess the effects of imatinib treatment. Nine proteins measured at hospital admission accurately predict critical illness development. Next to dysregulation of inflammation, critical illness is characterised by pathways involving cellular adhesion, extracellular matrix turnover and tissue remodelling. Imatinib treatment attenuates protein perturbations associated with inflammation and extracellular matrix turnover. These proteomic alterations are contextualised using external pulmonary RNA-sequencing data of deceased COVID-19 patients and imatinib-treated Syrian hamsters. Together, we show that alveolar capillary barrier disruption in critical COVID-19 is reflected in the plasma proteome, and is attenuated with imatinib treatment. This study comprises a secondary analysis of both clinical data and plasma samples derived from a clinical trial that was registered with the EU Clinical Trials Register (EudraCT 2020-001236-10, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001236-10/NL ) and Netherlands Trial Register (NL8491, https://www.trialregister.nl/trial/8491 ).
Collapse
Affiliation(s)
- Erik Duijvelaar
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| | - Jack Gisby
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - James E Peters
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
68
|
Ben Yellin, Lahav C, Sela I, Yahalom G, Shoval SR, Elon Y, Fuller J, Harel M. Analytical validation of the PROphet test for treatment decision-making guidance in metastatic non-small cell lung cancer. J Pharm Biomed Anal 2024; 238:115803. [PMID: 37871417 DOI: 10.1016/j.jpba.2023.115803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/22/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
The blood proteome, consisting of thousands of proteins engaged in various biological processes, acts as a valuable source of potential biomarkers for various medical applications. PROphet is a plasma proteomics-based test that serves as a decision-support tool for non-small cell lung cancer (NSCLC) patients, combining proteomic profiling using SomaScan technology and subsequent computational algorithm. PROphet was implemented as a laboratory developed test (LDT). Under the Clinical Laboratory Improvement Amendments (CLIA) and Commission on Office Laboratory Accreditation (COLA) regulations, prior to releasing patient test results, a clinical laboratory located in the United States employing an LDT must examine its performance characteristics with regard to analytical validity. This study describes the experimental and computational analytical validity of the PROphet test, as required by CLIA/COLA regulations. Experimental precision analysis displayed a median coefficient of variation (CV) of 3.9 % and 4.7 % for intra-plate and inter-plate examination, respectively, and the median accuracy rate between sites was 88 %. Computational precision exhibited a high accuracy rate, with 93 % of samples displaying complete concordance in results. A cross-platform comparison between SomaScan and other proteomics platforms yielded a median Spearman's rank correlation coefficient of 0.51, affirming the consistency and reliability of the SomaScan platform as used under the PROphet test. Our study presents a robust framework for evaluating the analytical validity of a platform that combines an experimental assay with subsequent computational algorithms. When applied to the PROphet test, strong analytical performance of the test was demonstrated.
Collapse
Affiliation(s)
- Ben Yellin
- OncoHost LTD, Hamelacha 17 Binyamina, 3057324, Israel
| | - Coren Lahav
- OncoHost LTD, Hamelacha 17 Binyamina, 3057324, Israel
| | - Itamar Sela
- OncoHost LTD, Hamelacha 17 Binyamina, 3057324, Israel
| | - Galit Yahalom
- OncoHost LTD, Hamelacha 17 Binyamina, 3057324, Israel
| | | | | | - James Fuller
- OncoHost Inc., 1110 SE Cary Parkway, Suite 205, Cary, NC 27518, USA
| | - Michal Harel
- OncoHost LTD, Hamelacha 17 Binyamina, 3057324, Israel.
| |
Collapse
|
69
|
Axelsson GT, Jonmundsson T, Woo Y, Frick EA, Aspelund T, Loureiro JJ, Orth AP, Jennings LL, Gudmundsson G, Emilsson V, Gudmundsdottir V, Gudnason V. Proteomic associations with forced expiratory volume: a Mendelian randomisation study. Respir Res 2024; 25:44. [PMID: 38238732 PMCID: PMC10797790 DOI: 10.1186/s12931-023-02587-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/30/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND A decline in forced expiratory volume (FEV1) is a hallmark of respiratory diseases that are an important cause of morbidity among the elderly. While some data exist on biomarkers that are related to FEV1, we sought to do a systematic analysis of causal relations of biomarkers with FEV1. METHODS Data from the population-based AGES-Reykjavik study were used. Serum proteomic measurements were done using 4782 DNA aptamers (SOMAmers). Data from 1479 participants with spirometric data were used to assess the association of SOMAmer measurements with FEV1 using linear regression. Bi-directional two-sample Mendelian randomisation (MR) analyses were done to assess causal relations of observationally associated SOMAmers with FEV1, using genotype and SOMAmer data from 5368 AGES-Reykjavik participants and genetic associations with FEV1 from a publicly available GWAS (n = 400,102). RESULTS In observational analyses, 530 SOMAmers were associated with FEV1 after multiple testing adjustment (FDR < 0.05). The most significant were Retinoic Acid Receptor Responder 2 (RARRES2), R-Spondin 4 (RSPO4) and Alkaline Phosphatase, Placental Like 2 (ALPPL2). Of the 257 SOMAmers with genetic instruments available, eight were associated with FEV1 in MR analyses. Three were directionally consistent with the observational estimate, Thrombospondin 2 (THBS2), Endoplasmic Reticulum Oxidoreductase 1 Beta (ERO1B) and Apolipoprotein M (APOM). THBS2 was further supported by a colocalization analysis. Analyses in the reverse direction, testing whether changes in SOMAmer levels were caused by changes in FEV1, were performed but no significant associations were found after multiple testing adjustments. CONCLUSIONS In summary, this large scale proteogenomic analyses of FEV1 reveals circulating protein markers of FEV1, as well as several proteins with potential causality to lung function.
Collapse
Affiliation(s)
- Gisli Thor Axelsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Department of Internal Medicine, Landspitali University Hospital, 101, Reykjavik, Iceland
| | - Thorarinn Jonmundsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Youngjae Woo
- Novartis Biomedical Research, Cambridge, MA, 02139, USA
| | | | - Thor Aspelund
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | | | - Anthony P Orth
- Novartis Institutes for Biomedical Research, San Diego, CA, 92121, USA
| | | | - Gunnar Gudmundsson
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
- Department of Respiratory Medicine and Sleep, Landspitali University Hospital, 108, Reykjavik, Iceland
| | - Valur Emilsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| | - Vilmundur Gudnason
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| |
Collapse
|
70
|
Shah AM, Myhre PL, Arthur V, Dorbala P, Rasheed H, Buckley LF, Claggett B, Liu G, Ma J, Nguyen NQ, Matsushita K, Ndumele C, Tin A, Hveem K, Jonasson C, Dalen H, Boerwinkle E, Hoogeveen RC, Ballantyne C, Coresh J, Omland T, Yu B. Large scale plasma proteomics identifies novel proteins and protein networks associated with heart failure development. Nat Commun 2024; 15:528. [PMID: 38225249 PMCID: PMC10789789 DOI: 10.1038/s41467-023-44680-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Heart failure (HF) causes substantial morbidity and mortality but its pathobiology is incompletely understood. The proteome is a promising intermediate phenotype for discovery of novel mechanisms. We measured 4877 plasma proteins in 13,900 HF-free individuals across three analysis sets with diverse age, geography, and HF ascertainment to identify circulating proteins and protein networks associated with HF development. Parallel analyses in Atherosclerosis Risk in Communities study participants in mid-life and late-life and in Trøndelag Health Study participants identified 37 proteins consistently associated with incident HF independent of traditional risk factors. Mendelian randomization supported causal effects of 10 on HF, HF risk factors, or left ventricular size and function, including matricellular (e.g. SPON1, MFAP4), senescence-associated (FSTL3, IGFBP7), and inflammatory (SVEP1, CCL15, ITIH3) proteins. Protein co-regulation network analyses identified 5 modules associated with HF risk, two of which were influenced by genetic variants that implicated trans hotspots within the VTN and CFH genes.
Collapse
Affiliation(s)
- Amil M Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Peder L Myhre
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pranav Dorbala
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Humaira Rasheed
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Leo F Buckley
- Department of Pharmacy, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian Claggett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Guning Liu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Jianzhong Ma
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ngoc Quynh Nguyen
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Chiadi Ndumele
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Adrienne Tin
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristian Hveem
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Christian Jonasson
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Håvard Dalen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Cardiology, St Olavs University Hospital, Trondheim, Norway
- Department of Internal Medicine, Levanger Hospital, Levanger, Norway
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ron C Hoogeveen
- Division of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Josef Coresh
- Departments of Medicine and Population Health, NYU Langone Health, New York, NY, USA
| | - Torbjørn Omland
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| |
Collapse
|
71
|
Gudmundsdottir V, Frick E, Emilsson V, Jonmundsson T, Steindorsdottir A, Johnson ECB, Puerta R, Dammer E, Shantaraman A, Cano A, Boada M, Valero S, Garcia-Gonzalez P, Gudmundsson E, Gudjonsson A, Pitts R, Qiu X, Finkel N, Loureiro J, Orth A, Seyfried N, Levey A, Ruiz A, Aspelund T, Jennings L, Launer L, Gudnason V. Serum proteomics reveals APOE dependent and independent protein signatures in Alzheimer's disease. RESEARCH SQUARE 2024:rs.3.rs-3706206. [PMID: 38260284 PMCID: PMC10802738 DOI: 10.21203/rs.3.rs-3706206/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The current demand for early intervention, prevention, and treatment of late onset Alzheimer's disease (LOAD) warrants deeper understanding of the underlying molecular processes which could contribute to biomarker and drug target discovery. Utilizing high-throughput proteomic measurements in serum from a prospective population-based cohort of older adults (n = 5,294), we identified 303 unique proteins associated with incident LOAD (median follow-up 12.8 years). Over 40% of these proteins were associated with LOAD independently of APOE-ε4 carrier status. These proteins were implicated in neuronal processes and overlapped with protein signatures of LOAD in brain and cerebrospinal fluid. We found 17 proteins which LOAD-association was strongly dependent on APOE-ε4 carrier status. Most of them showed consistent associations with LOAD in cerebrospinal fluid and a third had brain-specific gene expression. Remarkably, four proteins in this group (TBCA, ARL2, S100A13 and IRF6) were downregulated by APOE-ε4 yet upregulated as a consequence of LOAD as determined in a bi-directional Mendelian randomization analysis, reflecting a potential response to the disease onset. Accordingly, the direct association of these proteins to LOAD was reversed upon APOE-ε4 genotype adjustment, a finding which we replicate in an external cohort (n = 719). Our findings provide an insight into the dysregulated pathways that may lead to the development and early detection of LOAD, including those both independent and dependent on APOE-ε4. Importantly, many of the LOAD-associated proteins we find in the circulation have been found to be expressed - and have a direct link with AD - in brain tissue. Thus, the proteins identified here, and their upstream modulating pathways, provide a new source of circulating biomarker and therapeutic target candidates for LOAD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Merce Boada
- Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades-UIC, Barcelona
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lenore Launer
- National Institute on Aging, National Institutes of Health
| | | |
Collapse
|
72
|
Uribe-Carretero E, Rey V, Fuentes JM, Tamargo-Gómez I. Lysosomal Dysfunction: Connecting the Dots in the Landscape of Human Diseases. BIOLOGY 2024; 13:34. [PMID: 38248465 PMCID: PMC10813815 DOI: 10.3390/biology13010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Lysosomes are the main organelles responsible for the degradation of macromolecules in eukaryotic cells. Beyond their fundamental role in degradation, lysosomes are involved in different physiological processes such as autophagy, nutrient sensing, and intracellular signaling. In some circumstances, lysosomal abnormalities underlie several human pathologies with different etiologies known as known as lysosomal storage disorders (LSDs). These disorders can result from deficiencies in primary lysosomal enzymes, dysfunction of lysosomal enzyme activators, alterations in modifiers that impact lysosomal function, or changes in membrane-associated proteins, among other factors. The clinical phenotype observed in affected patients hinges on the type and location of the accumulating substrate, influenced by genetic mutations and residual enzyme activity. In this context, the scientific community is dedicated to exploring potential therapeutic approaches, striving not only to extend lifespan but also to enhance the overall quality of life for individuals afflicted with LSDs. This review provides insights into lysosomal dysfunction from a molecular perspective, particularly in the context of human diseases, and highlights recent advancements and breakthroughs in this field.
Collapse
Affiliation(s)
- Elisabet Uribe-Carretero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (E.U.-C.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Verónica Rey
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Jose Manuel Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (E.U.-C.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Isaac Tamargo-Gómez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
73
|
Yazdanpanah N, Jumentier B, Yazdanpanah M, Ong KK, Perry JRB, Manousaki D. Mendelian randomization identifies circulating proteins as biomarkers for age at menarche and age at natural menopause. Commun Biol 2024; 7:47. [PMID: 38184718 PMCID: PMC10771430 DOI: 10.1038/s42003-023-05737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Age at menarche (AAM) and age at natural menopause (ANM) are highly heritable traits and have been linked to various health outcomes. We aimed to identify circulating proteins associated with altered ANM and AAM using an unbiased two-sample Mendelian randomization (MR) and colocalization approach. By testing causal effects of 1,271 proteins on AAM, we identified 22 proteins causally associated with AAM in MR, among which 13 proteins (GCKR, FOXO3, SEMA3G, PATE4, AZGP1, NEGR1, LHB, DLK1, ANXA2, YWHAB, DNAJB12, RMDN1 and HPGDS) colocalized. Among 1,349 proteins tested for causal association with ANM using MR, we identified 19 causal proteins among which 7 proteins (CPNE1, TYMP, DNER, ADAMTS13, LCT, ARL and PLXNA1) colocalized. Follow-up pathway and gene enrichment analyses demonstrated links between AAM-related proteins and obesity and diabetes, and between AAM and ANM-related proteins and various types of cancer. In conclusion, we identified proteomic signatures of reproductive ageing in women, highlighting biological processes at both ends of the reproductive lifespan.
Collapse
Affiliation(s)
- Nahid Yazdanpanah
- Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Basile Jumentier
- Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Mojgan Yazdanpanah
- Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Ken K Ong
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - John R B Perry
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Despoina Manousaki
- Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada.
- Departments of Pediatrics, Biochemistry and Molecular Medicine, University of Montreal, Montreal, Canada.
| |
Collapse
|
74
|
He Q, Wang W, Xiong Y, Tao C, Ma L, You C. Potential Biomarkers in Cerebrospinal Fluid and Plasma for Dementia. J Alzheimers Dis 2024; 100:603-611. [PMID: 38875042 DOI: 10.3233/jad-240260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Background The identification of biomarkers for different dementias in plasma and cerebrospinal fluid (CSF) has made substantial progress. However, they are observational studies, and there remains a lack of research on dementias with low incidence rates. Objective We performed a comprehensive Mendelian randomization to identify potential biomarkers for different dementia type. Methods The summary-level datasets encompassed 734 plasma and 154 cerebrospinal fluid proteins sourced from recently published genome-wide association studies (GWAS). Summary statistics for different dementias, including any dementia (refering to any type of dementia symptoms, 218,792 samples), Alzheimer's disease (AD, 63,926 samples), vascular dementia (212,389 samples), frontotemporal dementia (3,024 samples), dementia with Lewy bodies (DLB, 6,618 samples), and dementia in Parkinson's disease (216,895 samples), were collected from large GWAS. The primary method is inverse variance weighting, with additional sensitivity analyses conducted to ensure the robustness of the findings. Results The molecules released into CSF, namely APOE2 for any dementia, APOE2 and Siglec-3 for AD, APOE2 for vascular dementia, and APOE2 for DLB, might be potential biomarkers. CD33 for AD and SNCA for DLB in plasma could be promising biomarkers. Conclusions This is the first study to integrate plasma and CSF proteins to identify potential biomarkers for different dementias.
Collapse
Affiliation(s)
- Qiang He
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenjing Wang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Xiong
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Chuanyuan Tao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
75
|
Emilsson V, Jonsson BG, Austin TR, Gudmundsdottir V, Axelsson GT, Frick EA, Jonmundsson T, Steindorsdottir AE, Loureiro J, Brody JA, Aspelund T, Launer LJ, Thorgeirsson G, Kortekaas KA, Lindeman JH, Orth AP, Lamb JR, Psaty BM, Kizer JR, Jennings LL, Gudnason V. Proteomic prediction of incident heart failure and its main subtypes. Eur J Heart Fail 2024; 26:87-102. [PMID: 37936531 DOI: 10.1002/ejhf.3086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/17/2023] [Accepted: 11/04/2023] [Indexed: 11/09/2023] Open
Abstract
AIM To examine the ability of serum proteins in predicting future heart failure (HF) events, including HF with reduced or preserved ejection fraction (HFrEF or HFpEF), in relation to event time, and with or without considering established HF-associated clinical variables. METHODS AND RESULTS In the prospective population-based Age, Gene/Environment Susceptibility Reykjavik Study (AGES-RS), 440 individuals developed HF after their first visit with a median follow-up of 5.45 years. Among them, 167 were diagnosed with HFrEF and 188 with HFpEF. A least absolute shrinkage and selection operator regression model with non-parametric bootstrap were used to select predictors from an analysis of 4782 serum proteins, and several pre-established clinical parameters linked to HF. A subset of 8-10 distinct or overlapping serum proteins predicted different future HF outcomes, and C-statistics were used to assess discrimination, revealing proteins combined with a C-index of 0.80 for all incident HF, 0.78 and 0.80 for incident HFpEF or HFrEF, respectively. In the AGES-RS, protein panels alone encompassed the risk contained in the clinical information and improved the performance characteristics of prediction models based on N-terminal pro-B-type natriuretic peptide and clinical risk factors. Finally, the protein predictors performed particularly well close to the time of an HF event, an outcome that was replicated in the Cardiovascular Health Study. CONCLUSION A small number of circulating proteins accurately predicted future HF in the AGES-RS cohort of older adults, and they alone encompass the risk information found in a collection of clinical data. Incident HF events were predicted up to 8 years, with predictor performance significantly improving for events occurring less than 1 year ahead, a finding replicated in an external cohort study.
Collapse
Affiliation(s)
- Valur Emilsson
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Thomas R Austin
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | | | | | | | - Joseph Loureiro
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, MD, USA
| | - Gudmundur Thorgeirsson
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Kirsten A Kortekaas
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan H Lindeman
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Anthony P Orth
- Novartis Institutes for Biomedical Research, San Diego, CA, USA
| | | | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Jorge R Kizer
- Division of Cardiology, San Francisco Veterans Affairs Health Care System, and Departments of Medicine, Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Lori L Jennings
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
76
|
Kizer JR, Patel S, Ganz P, Newman AB, Bhasin S, Lee SJ, Cawthon PM, LeBrasseur NK, Shah SJ, Psaty BM, Tracy RP, Cummings SR. Circulating Growth Differentiation Factors 11 and 8, Their Antagonists Follistatin and Follistatin-Like-3, and Risk of Heart Failure in Elders. J Gerontol A Biol Sci Med Sci 2024; 79:glad206. [PMID: 37624693 PMCID: PMC10733168 DOI: 10.1093/gerona/glad206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Heterochronic parabiosis has identified growth differentiation factor (GDF)-11 as a potential means of cardiac rejuvenation, but findings have been inconsistent. A major barrier has been lack of assay specificity for GDF-11 and its homolog GDF-8. METHODS We tested the hypothesis that GDF-11 and GDF-8, and their major antagonists follistatin and follistatin-like (FSTL)-3, are associated with incident heart failure (HF) and its subtypes in elders. Based on validation experiments, we used liquid chromatography-tandem mass spectrometry to measure total serum GDF-11 and GDF-8, along with follistatin and FSTL-3 by immunoassay, in 2 longitudinal cohorts of older adults. RESULTS In 2 599 participants (age 75.2 ± 4.3) followed for 10.8 ± 5.6 years, 721 HF events occurred. After adjustment, neither GDF-11 (HR per doubling: 0.93 [0.67, 1.30]) nor GDF-8 (HR: 1.02 per doubling [0.83, 1.27]) was associated with incident HF or its subtypes. Positive associations with HF were detected for follistatin (HR: 1.15 [1.00, 1.32]) and FLST-3 (HR: 1.38 [1.03, 1.85]), and with HF with preserved ejection fraction for FSTL-3 (HR: 1.77 [1.03, 3.02]). (All HRs per doubling of biomarker.) FSTL-3 associations with HF appeared stronger at higher follistatin levels and vice versa, and also for men, Blacks, and lower kidney function. CONCLUSIONS Among older adults, serum follistatin and FSTL-3, but not GDF-11 or GDF-8, were associated with incident HF. These findings do not support the concept that low serum levels of total GDF-11 or GDF-8 contribute to HF late in life, but do implicate transforming growth factor-β superfamily pathways as potential therapeutic targets.
Collapse
Affiliation(s)
- Jorge R Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Sheena Patel
- Research Institute, California Pacific Medical Center, San Francisco, California, USA
| | - Peter Ganz
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Cardiology Division, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Anne B Newman
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Se-Jin Lee
- The Jackson Laboratory and University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Peggy M Cawthon
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
- Research Institute, California Pacific Medical Center, San Francisco, California, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, and Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population Health, University of Washington, Seattle, Washington, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - Steven R Cummings
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
- Research Institute, California Pacific Medical Center, San Francisco, California, USA
| |
Collapse
|
77
|
Pessuti CL, Medley QG, Li N, Huang CL, Loureiro J, Banks A, Zhang Q, Costa DF, Ribeiro KS, Nascimento H, Muccioli C, Commodaro AG, Huang Q, Belfort R. Differential Proteins Expression Distinguished Between Patients With Infectious and Noninfectious Uveitis. Ocul Immunol Inflamm 2024; 32:40-47. [PMID: 36637883 DOI: 10.1080/09273948.2022.2150224] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/15/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE We investigated the aqueous humor proteome and associated plasma proteome in patients with infectious or noninfectious uveitis. METHODS AH and plasma were obtained from 28 patients with infectious uveitis (IU), 29 patients with noninfectious uveitis (NIU) and 35 healthy controls undergoing cataract surgery. The proteins profile was analyzed by SomaScan technology. RESULTS We found 1844 and 2484 proteins up-regulated and 124 and 161 proteins down-regulated in the AH from IU and NIU groups, respectively. In the plasma, three proteins were up-regulated in NIU patients, and one and five proteins were down-regulated in the IU and NIU patients, respectively. The results of pathway enrichment analysis for both IU and NIU groups were related mostly to inflammatory and regulatory processes. CONCLUSION SomaScan was able to detect novel AH and plasma protein biomarkers in IU and NIU patients. Also, the unique proteins found in both AH and plasma suggest a protein signature that could distinguish between infectious and noninfectious uveitis.
Collapse
Affiliation(s)
- Carmen L Pessuti
- Department of Ophthalmology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Quintus G Medley
- Ophthalmology, Novartis Institutes for Biomedical, Cambridge, Massachusetts, USA
| | - Ning Li
- Ophthalmology, Novartis Institutes for Biomedical, Cambridge, Massachusetts, USA
| | - Chia-Ling Huang
- Ophthalmology, Novartis Institutes for Biomedical, Cambridge, Massachusetts, USA
| | - Joseph Loureiro
- Ophthalmology, Novartis Institutes for Biomedical, Cambridge, Massachusetts, USA
| | - Angela Banks
- Ophthalmology, Novartis Institutes for Biomedical, Cambridge, Massachusetts, USA
| | - Qin Zhang
- Ophthalmology, Novartis Institutes for Biomedical, Cambridge, Massachusetts, USA
| | - Deise F Costa
- Department of Ophthalmology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Kleber S Ribeiro
- Department of Ophthalmology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Heloisa Nascimento
- Department of Ophthalmology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Cristina Muccioli
- Department of Ophthalmology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | | - Qian Huang
- Ophthalmology, Novartis Institutes for Biomedical, Cambridge, Massachusetts, USA
| | - Rubens Belfort
- Department of Ophthalmology, Federal University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
78
|
Chen YH, van Zon S, Adams A, Schmidt-Arras D, Laurence ADJ, Uhlig HH. The Human GP130 Cytokine Receptor and Its Expression-an Atlas and Functional Taxonomy of Genetic Variants. J Clin Immunol 2023; 44:30. [PMID: 38133879 PMCID: PMC10746620 DOI: 10.1007/s10875-023-01603-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 10/30/2023] [Indexed: 12/23/2023]
Abstract
Genetic variants in IL6ST encoding the shared cytokine receptor for the IL-6 cytokine family GP130 have been associated with a diverse number of clinical phenotypes and disorders. We provide a molecular classification for 59 reported rare IL6ST pathogenic or likely pathogenic variants and additional polymorphisms. Based on loss- or gain-of-function, cytokine selectivity, mono- and biallelic associations, and variable cellular mosaicism, we grade six classes of IL6ST variants and explore the potential for additional variants. We classify variants according to the American College of Medical Genetics and Genomics criteria. Loss-of-function variants with (i) biallelic complete loss of GP130 function that presents with extended Stüve-Wiedemann Syndrome; (ii) autosomal recessive hyper-IgE syndrome (HIES) caused by biallelic; and (iii) autosomal dominant HIES caused by monoallelic IL6ST variants both causing selective IL-6 and IL-11 cytokine loss-of-function defects; (iv) a biallelic cytokine-specific variant that exclusively impairs IL-11 signaling, associated with craniosynostosis and tooth abnormalities; (v) somatic monoallelic mosaic constitutively active gain-of-function variants in hepatocytes that present with inflammatory hepatocellular adenoma; and (vi) mosaic constitutively active gain-of-function variants in hematopoietic and non-hematopoietic cells that are associated with an immune dysregulation syndrome. In addition to Mendelian IL6ST coding variants, there are common non-coding cis-acting variants that modify gene expression, which are associated with an increased risk of complex immune-mediated disorders and trans-acting variants that affect GP130 protein function. Our taxonomy highlights IL6ST as a gene with particularly strong functional and phenotypic diversity due to the combinatorial biology of the IL-6 cytokine family and predicts additional genotype-phenotype associations.
Collapse
Affiliation(s)
- Yin-Huai Chen
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Sarah van Zon
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Alex Adams
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Dirk Schmidt-Arras
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | | | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
- Biomedical Research Centre, University of Oxford, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
| |
Collapse
|
79
|
Wu J, Fan Q, He Q, Zhong Q, Zhu X, Cai H, He X, Xu Y, Huang Y, Di X. Potential drug targets for myocardial infarction identified through Mendelian randomization analysis and Genetic colocalization. Medicine (Baltimore) 2023; 102:e36284. [PMID: 38065874 PMCID: PMC10713171 DOI: 10.1097/md.0000000000036284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Myocardial infarction (MI) is a major cause of death and disability worldwide, but current treatments are limited by their invasiveness, side effects, and lack of efficacy. Novel drug targets for MI prevention are urgently needed. In this study, we used Mendelian randomization to identify potential therapeutic targets for MI using plasma protein quantitative trait loci as exposure variables and MI as the outcome variable. We further validated our findings using reverse causation analysis, Bayesian co-localization analysis, and external datasets. We also constructed a protein-protein interaction network to explore the relationships between the identified proteins and known MI targets. Our analysis revealed 2 proteins, LPA and APOA5, as potential drug targets for MI, with causal effects on MI risk confirmed by multiple lines of evidence. LPA and APOA5 are involved in lipid metabolism and interact with target proteins of current MI medications. We also found 4 other proteins, IL1RN, FN1, NT5C, and SEMA3C, that may have potential as drug targets but require further confirmation. Our study demonstrates the utility of Mendelian randomization and protein quantitative trait loci in discovering novel drug targets for complex diseases such as MI. It provides insights into the underlying mechanisms of MI pathology and treatment.
Collapse
Affiliation(s)
- Jiayu Wu
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiaoming Fan
- Clifford Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi He
- The Eighth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Zhong
- The First Affiliated Hospital of Jinzhou Medical University, China
| | - Xianqiong Zhu
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, China
| | - Huilian Cai
- Clifford Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaolin He
- Clifford Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Xu
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuxuan Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xingwei Di
- The First Affiliated Hospital of Jinzhou Medical University, China
| |
Collapse
|
80
|
Schmitz D, Li Z, Lo Faro V, Rask-Andersen M, Ameur A, Rafati N, Johansson Å. Copy number variations and their effect on the plasma proteome. Genetics 2023; 225:iyad179. [PMID: 37793096 PMCID: PMC10697815 DOI: 10.1093/genetics/iyad179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 08/25/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023] Open
Abstract
Structural variations, including copy number variations (CNVs), affect around 20 million bases in the human genome and are common causes of rare conditions. CNVs are rarely investigated in complex disease research because most CNVs are not targeted on the genotyping arrays or the reference panels for genetic imputation. In this study, we characterize CNVs in a Swedish cohort (N = 1,021) using short-read whole-genome sequencing (WGS) and use long-read WGS for validation in a subcohort (N = 15), and explore their effect on 438 plasma proteins. We detected 184,182 polymorphic CNVs and identified 15 CNVs to be associated with 16 proteins (P < 8.22×10-10). Of these, 5 CNVs could be perfectly validated using long-read sequencing, including a CNV which was associated with measurements of the osteoclast-associated immunoglobulin-like receptor (OSCAR) and located upstream of OSCAR, a gene important for bone health. Two other CNVs were identified to be clusters of many short repetitive elements and another represented a complex rearrangement including an inversion. Our findings provide insights into the structure of common CNVs and their effects on the plasma proteome, and highlights the importance of investigating common CNVs, also in relation to complex diseases.
Collapse
Affiliation(s)
- Daniel Schmitz
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Box 815, 751 08 Uppsala, Sweden
| | - Zhiwei Li
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Box 815, 751 08 Uppsala, Sweden
| | - Valeria Lo Faro
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Box 815, 751 08 Uppsala, Sweden
| | - Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Box 815, 751 08 Uppsala, Sweden
| | - Adam Ameur
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Box 815, 751 08 Uppsala, Sweden
| | - Nima Rafati
- Department of Medical Biochemistry and Microbiology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Box 582, 751 23 Uppsala, Sweden
| | - Åsa Johansson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Box 815, 751 08 Uppsala, Sweden
| |
Collapse
|
81
|
Gilly A, Park YC, Tsafantakis E, Karaleftheri M, Dedoussis G, Zeggini E. Genome-wide meta-analysis of 92 cardiometabolic protein serum levels. Mol Metab 2023; 78:101810. [PMID: 37778719 PMCID: PMC10582065 DOI: 10.1016/j.molmet.2023.101810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVES Global cardiometabolic disease prevalence has grown rapidly over the years, making it the leading cause of death worldwide. Proteins are crucial components in biological pathways dysregulated in disease states. Identifying genetic components that influence circulating protein levels may lead to the discovery of biomarkers for early stages of disease or offer opportunities as therapeutic targets. METHODS Here, we carry out a genome-wide association study (GWAS) utilising whole genome sequencing data in 3,005 individuals from the HELIC founder populations cohort, across 92 proteins of cardiometabolic relevance. RESULTS We report 322 protein quantitative trait loci (pQTL) signals across 92 proteins, of which 76 are located in or near the coding gene (cis-pQTL). We link those association signals with changes in protein expression and cardiometabolic disease risk using colocalisation and Mendelian randomisation (MR) analyses. CONCLUSIONS The majority of previously unknown signals we describe point to proteins or protein interactions involved in inflammation and immune response, providing genetic evidence for the contributing role of inflammation in cardiometabolic disease processes.
Collapse
Affiliation(s)
- Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens, Greece
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany.
| |
Collapse
|
82
|
Debbs J, Hannawi B, Peterson E, Gui H, Zeld N, Luzum JA, Sabbah HN, Snider J, Pinto YM, Williams LK, Lanfear DE. Evaluation of a New Aptamer-Based Array for Soluble Suppressor of Tumorgenicity (ST2) and N-terminal Pro-B-Type Natriuretic Peptide (NTproBNP) in Heart Failure Patients. J Cardiovasc Transl Res 2023; 16:1343-1348. [PMID: 37191882 PMCID: PMC10651796 DOI: 10.1007/s12265-023-10397-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND Recent advances in multi-marker platforms offer faster data generation, but the fidelity of these methods compared to the ELISA is not established. We tested the correlation and predictive performance of SOMAscan vs. ELISA methods for NTproBNP and ST2. METHODS Patients ≥ 18 years with heart failure and ejection fraction < 50% were enrolled. We tested the correlation between SOMA and ELISA for each biomarker and their association with outcomes. RESULTS There was good correlation of SOMA vs. ELISA for ST2 (ρ = 0.71) and excellent correlation for NTproBNP (ρ = 0.94). The two versions of both markers were not significantly different regarding survival association. The two ST2 assays and NTproBNP assays were similarly associated with all-cause mortality and cardiovascular mortality. These associations remained statistically significant when adjusted for MAGGIC risk score (all p < 0.05). CONCLUSION SOMAscan quantifications of ST2 and NTproBNP correlate to ELISA versions and carry similar prognosis.
Collapse
Affiliation(s)
- Joseph Debbs
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Bashar Hannawi
- Heart and Vascular Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Edward Peterson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Hongsheng Gui
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Nicole Zeld
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Jasmine A Luzum
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Hani N Sabbah
- Heart and Vascular Institute, Henry Ford Hospital, Detroit, MI, USA
| | | | - Yigal M Pinto
- Department of Cardiology, University of Amsterdam, Amsterdam, the Netherlands
| | - L Keoki Williams
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - David E Lanfear
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA.
- Heart and Vascular Institute, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
83
|
Lecluze E, Lettre G. Association Analyses of Predicted Loss-of-Function Variants Prioritized 15 Genes as Blood Pressure Regulators. Can J Cardiol 2023; 39:1888-1897. [PMID: 37451613 DOI: 10.1016/j.cjca.2023.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/26/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Hypertension, clinically defined by elevated blood pressure (BP), is an important cause of mortality and morbidity worldwide. Many risk factors for hypertension are known, including a positive family history, which suggests that genetics contribute to interindividual BP variation. Genome-wide association studies (GWAS) have identified > 1000 loci associated with BP, yet the identity of the genes responsible for these associations remains largely unknown. METHODS To pinpoint genes that causally affect variation of BP in humans, we analyzed predicted loss-of-function (pLoF) variants in the UK Biobank whole-exome sequencing dataset (n = 454,709 participants, 6% non-European ancestry). We analyzed genetic associations between systolic or diastolic BP (SBP/DBP) and single pLoF variants (additive and recessive genetic models) as well as with the burden of very rare pLoF variants (minor allele frequency [MAF] < 0.01%). RESULTS Single pLoF variants in 10 genes were associated with BP (ANKDD1B, ENPEP, PNCK, BTN3A2, C1orf145 [OBSCN-AS1], CASP9, DBH, KIAA1161 [MYORG], OR4X1, and TMC3). We also found a burden of rare pLoF variants in 5 additional genes associated with BP (TTN, NOS3, FES, SMAD6, COL21A1). Except for PNCK, which is located on the X-chromosome, these genes map near variants previously associated with BP by GWAS, validating the study of pLoF variants to prioritize causal genes at GWAS loci. CONCLUSIONS Our study highlights 15 genes that likely modulate BP in humans, including 5 genes that harbour pLoF variants associated with lower BP.
Collapse
Affiliation(s)
- Estelle Lecluze
- Montreal Heart Institute, Montréal, Québec, Canada; Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute, Montréal, Québec, Canada; Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
84
|
Li J, Yao YX, Yao PS. Circulating Biomarkers and Risk of Hypertension: A Two-Sample Mendelian Randomisation Study. Heart Lung Circ 2023; 32:1434-1442. [PMID: 38042639 DOI: 10.1016/j.hlc.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/27/2023] [Accepted: 09/02/2023] [Indexed: 12/04/2023]
Abstract
OBJECTIVE This study systematically assessed circulating proteins to identify new serum biomarkers and risk of hypertension using Mendelian randomisation. METHODS The associations between 4,782 human circulating proteins and the risk of hypertension were evaluated using two-sample Mendelian randomisation. The FinnGen study demonstrated a link between genetic predisposition and hypertension in 85,438 cases and 223,663 controls. RESULTS Inverse variance weighted and sensitivity analysis revealed nine proteins in circulation that have a causative effect on hypertension. SMOC1 and TIE1 were determined to be causative factors in the decreased likelihood of developing hypertension, with odds ratios of 0.86 (95% CI 0.81-0.91; p=1.06e-06) and 0.96 (95% CI 0.94-0.98; p=9.39e-05), respectively. NDUFB4, ETHE1, POFUT2, TRIL, ADAM23, GXYLT1, OXT, TPST2, and TMCC3 showed a possible connection to hypertension. CONCLUSIONS This two-sample Mendelian randomisation study found that SMOC1 and TIE1 are causally linked to hypertension, making them a promising target for therapy.
Collapse
Affiliation(s)
- Jin Li
- Department of Cardiovascular Medicine, Fujian Provincial Geriatric Hospital, Fuzhou, China
| | - Yue-Xian Yao
- Department of Cardiovascular Medicine, Fujian Provincial Geriatric Hospital, Fuzhou, China
| | - Pei-Sen Yao
- Department of Neurosurgery, Neurosurgical Research Institute, First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
85
|
You J, Guo Y, Zhang Y, Kang JJ, Wang LB, Feng JF, Cheng W, Yu JT. Plasma proteomic profiles predict individual future health risk. Nat Commun 2023; 14:7817. [PMID: 38016990 PMCID: PMC10684756 DOI: 10.1038/s41467-023-43575-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Developing a single-domain assay to identify individuals at high risk of future events is a priority for multi-disease and mortality prevention. By training a neural network, we developed a disease/mortality-specific proteomic risk score (ProRS) based on 1461 Olink plasma proteins measured in 52,006 UK Biobank participants. This integrative score markedly stratified the risk for 45 common conditions, including infectious, hematological, endocrine, psychiatric, neurological, sensory, circulatory, respiratory, digestive, cutaneous, musculoskeletal, and genitourinary diseases, cancers, and mortality. The discriminations witnessed high accuracies achieved by ProRS for 10 endpoints (e.g., cancer, dementia, and death), with C-indexes exceeding 0.80. Notably, ProRS produced much better or equivalent predictive performance than established clinical indicators for almost all endpoints. Incorporating clinical predictors with ProRS enhanced predictive power for most endpoints, but this combination only exhibited limited improvement when compared to ProRS alone. Some proteins, e.g., GDF15, exhibited important discriminative values for various diseases. We also showed that the good discriminative performance observed could be largely translated into practical clinical utility. Taken together, proteomic profiles may serve as a replacement for complex laboratory tests or clinical measures to refine the comprehensive risk assessments of multiple diseases and mortalities simultaneously. Our models were internally validated in the UK Biobank; thus, further independent external validations are necessary to confirm our findings before application in clinical settings.
Collapse
Affiliation(s)
- Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Lin-Bo Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
- School of Data Science, Fudan University, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer Center, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
86
|
Zilinskas R, Li C, Shen X, Pan W, Yang T. Inferring a directed acyclic graph of phenotypes from GWAS summary statistics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528092. [PMID: 38045347 PMCID: PMC10690198 DOI: 10.1101/2023.02.10.528092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Estimating phenotype networks is a growing field in computational biology. It deepens the understanding of disease etiology and is useful in many applications. In this study, we present a method that constructs a phenotype network by assuming a Gaussian linear structure model embedding a directed acyclic graph (DAG). We utilize genetic variants as instrumental variables and show how our method only requires access to summary statistics from a genome-wide association study (GWAS) and a reference panel of genotype data. Besides estimation, a distinct feature of the method is its summary statistics-based likelihood ratio test on directed edges. We applied our method to estimate a causal network of 29 cardiovascular-related proteins and linked the estimated network to Alzheimer's disease (AD). A simulation study was conducted to demonstrate the effectiveness of this method. An R package sumdag implementing the proposed method, all relevant code, and a Shiny application are available at https://github.com/chunlinli/sumdag.
Collapse
Affiliation(s)
| | - Chunlin Li
- Department of Statistics, Iowa State University, Ames, Iowa 50011, U.S.A
| | - Xiaotong Shen
- School of Statistics, University of Minnesota, Minneapolis, Minnesota 55455, U.S.A
| | - Wei Pan
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota 55455, U.S.A
| | - Tianzhong Yang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota 55455, U.S.A
| |
Collapse
|
87
|
Frick EA, Emilsson V, Jonmundsson T, Steindorsdottir AE, Johnson ECB, Puerta R, Dammer EB, Shantaraman A, Cano A, Boada M, Valero S, García-González P, Gudmundsson EF, Gudjonsson A, Loureiro JJ, Orth AP, Seyfried NT, Levey AI, Ruiz A, Aspelund T, Jennings LL, Launer LJ, Gudmundsdottir V, Gudnason V. Serum proteomics reveals APOE dependent and independent protein signatures in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.08.23298251. [PMID: 37986771 PMCID: PMC10659486 DOI: 10.1101/2023.11.08.23298251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The current demand for early intervention, prevention, and treatment of late onset Alzheimer's disease (LOAD) warrants deeper understanding of the underlying molecular processes which could contribute to biomarker and drug target discovery. Utilizing high-throughput proteomic measurements in serum from a prospective population-based cohort of older adults (n=5,294), we identified 303 unique proteins associated with incident LOAD (median follow-up 12.8 years). Over 40% of these proteins were associated with LOAD independently of APOE-ε4 carrier status. These proteins were implicated in neuronal processes and overlapped with protein signatures of LOAD in brain and cerebrospinal fluid. We found 17 proteins which LOAD-association was strongly dependent on APOE-ε4 carrier status. Most of them showed consistent associations with LOAD in cerebrospinal fluid and a third had brain-specific gene expression. Remarkably, four proteins in this group (TBCA, ARL2, S100A13 and IRF6) were downregulated by APOE-ε4 yet upregulated as a consequence of LOAD as determined in a bi-directional Mendelian randomization analysis, reflecting a potential response to the disease onset. Accordingly, the direct association of these proteins to LOAD was reversed upon APOE-ε4 genotype adjustment, a finding which we replicate in an external cohort (n=719). Our findings provide an insight into the dysregulated pathways that may lead to the development and early detection of LOAD, including those both independent and dependent on APOE-ε4. Importantly, many of the LOAD-associated proteins we find in the circulation have been found to be expressed - and have a direct link with AD - in brain tissue. Thus, the proteins identified here, and their upstream modulating pathways, provide a new source of circulating biomarker and therapeutic target candidates for LOAD.
Collapse
Affiliation(s)
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, 200, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | | | - Erik C B Johnson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Raquel Puerta
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
| | - Eric B Dammer
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Anantharaman Shantaraman
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Amanda Cano
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | - Mercè Boada
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | - Sergi Valero
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | - Pablo García-González
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | | | | | | | | | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Agustin Ruiz
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | - Thor Aspelund
- Icelandic Heart Association, Kopavogur, 200, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, 20892, MD, USA
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Kopavogur, 200, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, 200, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| |
Collapse
|
88
|
Li H, Wang Y, Sonestedt E, Borné Y. Associations of ultra-processed food consumption, circulating protein biomarkers, and risk of cardiovascular disease. BMC Med 2023; 21:415. [PMID: 37919714 PMCID: PMC10623817 DOI: 10.1186/s12916-023-03111-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND We aim to examine the association between ultra-processed foods (UPF) consumption and cardiovascular disease (CVD) risk and to identify plasma proteins associated with UPF. METHODS This prospective cohort study included 26,369 participants from the Swedish Malmö Diet and Cancer Study, established in 1991-1996. Dietary intake was assessed using a modified diet history method, and UPF consumption was estimated using the NOVA classification system. A total of 88 selected CVD-related proteins were measured among 4475 subjects. Incident CVD (coronary heart disease and ischemic stroke) was defined as a hospital admission or death through registers. Cox proportional hazards regression models were performed to analyze the associations of UPF intake with risks of CVD. Linear regression models were used to identify the plasma proteins associated with UPF intake. RESULTS During 24.6 years of median follow-up, 6236 participants developed CVD, of whom 3566 developed coronary heart disease and 3272 developed ischemic stroke. The adjusted hazard ratio (95% confidence interval) in the 4th versus 1st quartile of UPF was 1.18 (1.08, 1.29) for CVD, 1.20 (1.07, 1.35) for coronary heart disease, and 1.17 (1.03, 1.32) for ischemic stroke. Plasma proteins interleukin 18, tumor necrosis factor receptor 2, macrophage colony-stimulating factor 1, thrombomodulin, tumor necrosis factor receptor 1, hepatocyte growth factor, stem cell factor, resistin, C-C motif chemokine 3, and endothelial cell-specific molecule 1 were positively associated with UPF after correcting for multiple testing. CONCLUSIONS Our study showed that high UPF intake increased the risk of CVD and was associated with several protein biomarkers. Future studies are warranted to validate these findings and assess the potential pathways between UPF intake and CVD.
Collapse
Affiliation(s)
- Huiping Li
- School of Medicine, Northwest University, Taibai North Road, Beilin District, Xi'an, 710069, China.
- School of Public Health, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China.
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms Gata 35, 21428, Malmö, Sweden.
| | - Yaogang Wang
- School of Public Health, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Emily Sonestedt
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms Gata 35, 21428, Malmö, Sweden
| | - Yan Borné
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms Gata 35, 21428, Malmö, Sweden.
| |
Collapse
|
89
|
Jonmundsson T, Steindorsdottir AE, Austin TR, Frick EA, Axelsson GT, Launer L, Psaty BM, Loureiro J, Orth AP, Aspelund T, Emilsson V, Floyd JS, Jennings L, Gudnason V, Gudmundsdottir V. A proteomic analysis of atrial fibrillation in a prospective longitudinal cohort (AGES-Reykjavik study). Europace 2023; 25:euad320. [PMID: 37967346 PMCID: PMC10685397 DOI: 10.1093/europace/euad320] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/01/2023] [Accepted: 10/06/2023] [Indexed: 11/17/2023] Open
Abstract
AIMS Atrial fibrillation (AF) is associated with high risk of comorbidities and mortality. Our aim was to examine causal and predictive relationships between 4137 serum proteins and incident AF in the prospective population-based Age, Gene/Environment Susceptibility-Reykjavik (AGES-Reykjavik) study. METHODS AND RESULTS The study included 4765 participants, of whom 1172 developed AF. Cox proportional hazards regression models were fitted for 4137 baseline protein measurements adjusting for known risk factors. Protein associations were tested for replication in the Cardiovascular Health Study (CHS). Causal relationships were examined in a bidirectional, two-sample Mendelian randomization analysis. The time-dependent area under the receiver operating characteristic curve (AUC)-statistic was examined as protein levels and an AF-polygenic risk score (PRS) were added to clinical risk models. The proteomic signature of incident AF consisted of 76 proteins, of which 63 (83%) were novel and 29 (38%) were replicated in CHS. The signature included both N-terminal prohormone of brain natriuretic peptide (NT-proBNP)-dependent (e.g. CHST15, ATP1B1, and SVEP1) and independent components (e.g. ASPN, AKR1B, and LAMA1/LAMB1/LAMC1). Nine causal candidates were identified (TAGLN, WARS, CHST15, CHMP3, COL15A1, DUSP13, MANBA, QSOX2, and SRL). The reverse causal analysis suggested that most AF-associated proteins were affected by the genetic liability to AF. N-terminal prohormone of brain natriuretic peptide improved the prediction of incident AF events close to baseline with further improvements gained by the AF-PRS at all time points. CONCLUSION The AF proteomic signature includes biologically relevant proteins, some of which may be causal. It mainly reflects an NT-proBNP-dependent consequence of the genetic liability to AF. N-terminal prohormone of brain natriuretic peptide is a promising marker for incident AF in the short term, but risk assessment incorporating a PRS may improve long-term risk assessment.
Collapse
Affiliation(s)
- Thorarinn Jonmundsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | | | - Thomas R Austin
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Elisabet A Frick
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Gisli T Axelsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | | | | | - Thor Aspelund
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Valur Emilsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| |
Collapse
|
90
|
Perry AS, Zhao S, Gajjar P, Murthy VL, Lehallier B, Miller P, Nair S, Neill C, Carr JJ, Fearon W, Kapadia S, Kumbhani D, Gillam L, Lindenfeld J, Farrell L, Marron MM, Tian Q, Newman AB, Murabito J, Gerszten RE, Nayor M, Elmariah S, Lindman BR, Shah R. Proteomic architecture of frailty across the spectrum of cardiovascular disease. Aging Cell 2023; 22:e13978. [PMID: 37731195 PMCID: PMC10652351 DOI: 10.1111/acel.13978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023] Open
Abstract
While frailty is a prominent risk factor in an aging population, the underlying biology of frailty is incompletely described. Here, we integrate 979 circulating proteins across a wide range of physiologies with 12 measures of frailty in a prospective discovery cohort of 809 individuals with severe aortic stenosis (AS) undergoing transcatheter aortic valve implantation. Our aim was to characterize the proteomic architecture of frailty in a highly susceptible population and study its relation to clinical outcome and systems-wide phenotypes to define potential novel, clinically relevant frailty biology. Proteomic signatures (specifically of physical function) were related to post-intervention outcome in AS, specifying pathways of innate immunity, cell growth/senescence, fibrosis/metabolism, and a host of proteins not widely described in human aging. In published cohorts, the "frailty proteome" displayed heterogeneous trajectories across age (20-100 years, age only explaining a small fraction of variance) and were associated with cardiac and non-cardiac phenotypes and outcomes across two broad validation cohorts (N > 35,000) over ≈2-3 decades. These findings suggest the importance of precision biomarkers of underlying multi-organ health status in age-related morbidity and frailty.
Collapse
Affiliation(s)
- Andrew S. Perry
- Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Shilin Zhao
- Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Priya Gajjar
- Cardiovascular Medicine Section, Department of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | | | | | - Patricia Miller
- Department of Medicine, and Department of BiostatisticsBoston University School of MedicineBostonMassachusettsUSA
| | - Sangeeta Nair
- Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Colin Neill
- Department of Medicine, Division of Cardiovascular MedicineUniversity of Wisconsin Hospital and ClinicsMadisonWisconsinUSA
| | - J. Jeffrey Carr
- Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - William Fearon
- Department of Medicine, Division of CardiologyStanford Medical CenterPalo AltoCaliforniaUSA
| | - Samir Kapadia
- Department of Medicine, Division of CardiologyCleveland Clinic FoundationClevelandOhioUSA
| | - Dharam Kumbhani
- Department of Medicine, Division of CardiologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Linda Gillam
- Department of Cardiovascular MedicineMorristown Medical CenterMorristownNew JerseyUSA
| | - JoAnn Lindenfeld
- Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Laurie Farrell
- Broad Institute of Harvard and MITCambridgeMassachusettsUSA
| | - Megan M. Marron
- Department of Epidemiology, Graduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Qu Tian
- National Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Anne B. Newman
- Department of Epidemiology, Graduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Departments of Medicine and Clinical and Translational ScienceUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Joanne Murabito
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Robert E. Gerszten
- Broad Institute of Harvard and MITCambridgeMassachusettsUSA
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonMassachusettsUSA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Sammy Elmariah
- Department of Medicine, Division of CardiologyThe University of CaliforniaSan FranciscoCaliforniaUSA
| | - Brian R. Lindman
- Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Ravi Shah
- Vanderbilt Translational and Clinical Cardiovascular Research CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| |
Collapse
|
91
|
Dammer EB, Shantaraman A, Ping L, Duong DM, Gerasimov ES, Ravindran SP, Gudmundsdottir V, Frick EA, Gomez GT, Walker KA, Emilsson V, Jennings LL, Gudnason V, Western D, Cruchaga C, Lah JJ, Wingo TS, Wingo AP, Seyfried NT, Levey AI, Johnson EC. Proteomic Network Analysis of Alzheimer's Disease Cerebrospinal Fluid Reveals Alterations Associated with APOE ε4 Genotype and Atomoxetine Treatment. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.29.23297651. [PMID: 37961720 PMCID: PMC10635242 DOI: 10.1101/2023.10.29.23297651] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Alzheimer's disease (AD) is currently defined at the research level by the aggregation of amyloid-β (Aβ) and tau proteins in brain. While biofluid biomarkers are available to measure Aβ and tau pathology, few biomarkers are available to measure the complex pathophysiology that is associated with these two cardinal neuropathologies. Here we describe the proteomic landscape of cerebrospinal fluid (CSF) changes associated with Aβ and tau pathology in 300 individuals as assessed by two different proteomic technologies-tandem mass tag (TMT) mass spectrometry and SomaScan. Harmonization and integration of both data types allowed for generation of a robust protein co-expression network consisting of 34 modules derived from 5242 protein measurements, including disease-relevant modules associated with autophagy, ubiquitination, endocytosis, and glycolysis. Three modules strongly associated with the apolipoprotein E ε4 (APOE ε4) AD risk genotype mapped to oxidant detoxification, mitogen associated protein kinase (MAPK) signaling, neddylation, and mitochondrial biology, and overlapped with a previously described lipoprotein module in serum. Neddylation and oxidant detoxification/MAPK signaling modules had a negative association with APOE ε4 whereas the mitochondrion module had a positive association with APOE ε4. The directions of association were consistent between CSF and blood in two independent longitudinal cohorts, and altered levels of all three modules in blood were associated with dementia over 20 years prior to diagnosis. Dual-proteomic platform analysis of CSF samples from an AD phase 2 clinical trial of atomoxetine (ATX) demonstrated that abnormal elevations in the glycolysis CSF module-the network module most strongly correlated to cognitive function-were reduced by ATX treatment. Individuals who had more severe glycolytic changes at baseline responded better to ATX. Clustering of individuals based on their CSF proteomic network profiles revealed ten groups that did not cleanly stratify by Aβ and tau status, underscoring the heterogeneity of pathological changes not fully reflected by Aβ and tau. AD biofluid proteomics holds promise for the development of biomarkers that reflect diverse pathologies for use in clinical trials and precision medicine.
Collapse
Affiliation(s)
- Eric B. Dammer
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Anantharaman Shantaraman
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lingyan Ping
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M. Duong
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Valborg Gudmundsdottir
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Gabriela T. Gomez
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Daniel Western
- Department of Psychiatry, Washington University, St. Louis, MO, USA
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO, USA
- NeuroGenomics and Informatics, Washington University, St. Louis, MO, USA
| | - James J. Lah
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas S. Wingo
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Aliza P. Wingo
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
- Division of Mental Health, Atlanta VA Medical Center, GA, USA
| | - Nicholas T. Seyfried
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I. Levey
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Erik C.B. Johnson
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
92
|
McBride N, Fernández-Sanlés A, Arab MA, Bond TA, Zheng J, Magnus MC, Corfield EC, Clayton GL, Hwang LD, Beaumont RN, Evans DM, Freathy RM, Gaunt TR, Lawlor DA, Borges MC. Effects of the maternal and fetal proteome on birth weight: a Mendelian randomization analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.20.23297135. [PMID: 37904919 PMCID: PMC10615012 DOI: 10.1101/2023.10.20.23297135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Fetal growth is an indicator of fetal survival, regulated by maternal and fetal factors, but little is known about the underlying molecular mechanisms. We used Mendelian randomization to explore the effects of maternal and fetal genetically-instrumented plasma proteins on birth weight using genome-wide association summary data (n=406,063 with maternal and/or fetal genotype), with independent replication (n=74,932 mothers and n=62,108 offspring), and colocalisation. Higher genetically-predicted maternal levels of PCSK1 increased birthweight (mean-difference: 9g (95% CI: 5g, 13g) per 1 standard deviation protein level). Higher maternal levels of LGALS4 decreased birthweight (-54g (-29g, -80g)), as did VCAM1, RAD51D and GP1BA. In the offspring, higher genetically-predicted fetal levels of LGALS4 (46g (23g, 70g)) increased birthweight, alongside FCGR2B. Higher offspring levels of PCSK1 decreased birth weight (-9g (-16g, 4g), alongside LEPR. Results support maternal and fetal protein effects on birth weight, implicating roles for glucose metabolism, energy homeostasis, endothelial function and adipocyte differentiation.
Collapse
|
93
|
Szrok-Jurga S, Czumaj A, Turyn J, Hebanowska A, Swierczynski J, Sledzinski T, Stelmanska E. The Physiological and Pathological Role of Acyl-CoA Oxidation. Int J Mol Sci 2023; 24:14857. [PMID: 37834305 PMCID: PMC10573383 DOI: 10.3390/ijms241914857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Fatty acid metabolism, including β-oxidation (βOX), plays an important role in human physiology and pathology. βOX is an essential process in the energy metabolism of most human cells. Moreover, βOX is also the source of acetyl-CoA, the substrate for (a) ketone bodies synthesis, (b) cholesterol synthesis, (c) phase II detoxication, (d) protein acetylation, and (d) the synthesis of many other compounds, including N-acetylglutamate-an important regulator of urea synthesis. This review describes the current knowledge on the importance of the mitochondrial and peroxisomal βOX in various organs, including the liver, heart, kidney, lung, gastrointestinal tract, peripheral white blood cells, and other cells. In addition, the diseases associated with a disturbance of fatty acid oxidation (FAO) in the liver, heart, kidney, lung, alimentary tract, and other organs or cells are presented. Special attention was paid to abnormalities of FAO in cancer cells and the diseases caused by mutations in gene-encoding enzymes involved in FAO. Finally, issues related to α- and ω- fatty acid oxidation are discussed.
Collapse
Affiliation(s)
- Sylwia Szrok-Jurga
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Jacek Turyn
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Areta Hebanowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Julian Swierczynski
- Institue of Nursing and Medical Rescue, State University of Applied Sciences in Koszalin, 75-582 Koszalin, Poland;
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Ewa Stelmanska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| |
Collapse
|
94
|
Grover SP, Mackman N, Bendapudi PK. Heat shock protein 47 and venous thrombosis: letting sleeping bears lie. J Thromb Haemost 2023; 21:2648-2652. [PMID: 37473845 DOI: 10.1016/j.jtha.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/22/2023]
Affiliation(s)
- Steven P Grover
- University of North Carolina Blood Research Center, The University of North Carolina at Chapel Hill, North Carolina, USA; Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, North Carolina, USA.
| | - Nigel Mackman
- University of North Carolina Blood Research Center, The University of North Carolina at Chapel Hill, North Carolina, USA; Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, North Carolina, USA
| | - Pavan K Bendapudi
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Center for the Development of Therapeutics, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
95
|
Grover SP, Sundler Björkman L, Brækkan SK, Zöller B, Hansen JB, Mackman N. "C1-inhibitor levels and Venous Thromboembolism: Results from a Mendelian Randomization Study": comment from Grover et al. J Thromb Haemost 2023; 21:2985-2987. [PMID: 37739595 DOI: 10.1016/j.jtha.2023.06.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 09/24/2023]
Affiliation(s)
- Steven P Grover
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| | - Linda Sundler Björkman
- Respiratory Medicine, Allergology, and Palliative Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Skåne University Hospital, Lund, Sweden
| | - Sigrid K Brækkan
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Bengt Zöller
- Center for Primary Health Care Research, Lund University and Region Skåne, Malmö, Sweden
| | - John-Bjarne Hansen
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
96
|
Cupido AJ, Petersen RS, Schmidt AF, Levi M, Cohn DM, Fijen LM. "C1-inhibitor levels and venous thromboembolism: results from a Mendelian randomization study": reply. J Thromb Haemost 2023; 21:2988-2990. [PMID: 37739596 DOI: 10.1016/j.jtha.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 09/24/2023]
Affiliation(s)
- Arjen J Cupido
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Remy S Petersen
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Amand F Schmidt
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, the Netherlands; Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK; Health Data Research UK and Institute of Health Informatics, University College London, London, UK; UCL British Heart Foundation Research Accelerator, London, UK
| | - Marcel Levi
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Danny M Cohn
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Lauré M Fijen
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
97
|
Sun BB, Chiou J, Traylor M, Benner C, Hsu YH, Richardson TG, Surendran P, Mahajan A, Robins C, Vasquez-Grinnell SG, Hou L, Kvikstad EM, Burren OS, Davitte J, Ferber KL, Gillies CE, Hedman ÅK, Hu S, Lin T, Mikkilineni R, Pendergrass RK, Pickering C, Prins B, Baird D, Chen CY, Ward LD, Deaton AM, Welsh S, Willis CM, Lehner N, Arnold M, Wörheide MA, Suhre K, Kastenmüller G, Sethi A, Cule M, Raj A, Burkitt-Gray L, Melamud E, Black MH, Fauman EB, Howson JMM, Kang HM, McCarthy MI, Nioi P, Petrovski S, Scott RA, Smith EN, Szalma S, Waterworth DM, Mitnaul LJ, Szustakowski JD, Gibson BW, Miller MR, Whelan CD. Plasma proteomic associations with genetics and health in the UK Biobank. Nature 2023; 622:329-338. [PMID: 37794186 PMCID: PMC10567551 DOI: 10.1038/s41586-023-06592-6] [Citation(s) in RCA: 130] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/31/2023] [Indexed: 10/06/2023]
Abstract
The Pharma Proteomics Project is a precompetitive biopharmaceutical consortium characterizing the plasma proteomic profiles of 54,219 UK Biobank participants. Here we provide a detailed summary of this initiative, including technical and biological validations, insights into proteomic disease signatures, and prediction modelling for various demographic and health indicators. We present comprehensive protein quantitative trait locus (pQTL) mapping of 2,923 proteins that identifies 14,287 primary genetic associations, of which 81% are previously undescribed, alongside ancestry-specific pQTL mapping in non-European individuals. The study provides an updated characterization of the genetic architecture of the plasma proteome, contextualized with projected pQTL discovery rates as sample sizes and proteomic assay coverages increase over time. We offer extensive insights into trans pQTLs across multiple biological domains, highlight genetic influences on ligand-receptor interactions and pathway perturbations across a diverse collection of cytokines and complement networks, and illustrate long-range epistatic effects of ABO blood group and FUT2 secretor status on proteins with gastrointestinal tissue-enriched expression. We demonstrate the utility of these data for drug discovery by extending the genetic proxied effects of protein targets, such as PCSK9, on additional endpoints, and disentangle specific genes and proteins perturbed at loci associated with COVID-19 susceptibility. This public-private partnership provides the scientific community with an open-access proteomics resource of considerable breadth and depth to help to elucidate the biological mechanisms underlying proteo-genomic discoveries and accelerate the development of biomarkers, predictive models and therapeutics1.
Collapse
Affiliation(s)
- Benjamin B Sun
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA.
| | - Joshua Chiou
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Matthew Traylor
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | | | | | - Tom G Richardson
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
- Genomic Sciences, GlaxoSmithKline, Stevenage, UK
| | | | | | - Chloe Robins
- Genomic Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | | | - Liping Hou
- Population Analytics, Janssen Research & Development, Spring House, PA, USA
| | | | - Oliver S Burren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Kyle L Ferber
- Biostatistics, Research and Development, Biogen, Cambridge, MA, USA
| | | | - Åsa K Hedman
- External Science and Innovation Target Sciences, Worldwide Research, Development and Medical, Pfizer, Stockholm, Sweden
| | - Sile Hu
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Tinchi Lin
- Analytics and Data Sciences, Biogen, Cambridge, MA, USA
| | - Rajesh Mikkilineni
- Data Science Institute, Takeda Development Center Americas, Cambridge, MA, USA
| | | | | | - Bram Prins
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Denis Baird
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA
| | - Chia-Yen Chen
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA
| | - Lucas D Ward
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Aimee M Deaton
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | - Carissa M Willis
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Nick Lehner
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Maria A Wörheide
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - Anil Raj
- Calico Life Sciences, San Francisco, CA, USA
| | | | | | - Mary Helen Black
- Population Analytics, Janssen Research & Development, Spring House, PA, USA
| | - Eric B Fauman
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Joanna M M Howson
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | | | | | - Paul Nioi
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Department of Medicine, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | | | - Erin N Smith
- Takeda Development Center Americas, San Diego, CA, USA
| | - Sándor Szalma
- Takeda Development Center Americas, San Diego, CA, USA
| | | | | | | | | | - Melissa R Miller
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Christopher D Whelan
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA.
- Neuroscience Data Science, Janssen Research & Development, Cambridge, MA, USA.
| |
Collapse
|
98
|
Ren F, Jin Q, Liu T, Ren X, Zhan Y. Proteome-wide mendelian randomization study implicates therapeutic targets in common cancers. J Transl Med 2023; 21:646. [PMID: 37735436 PMCID: PMC10512580 DOI: 10.1186/s12967-023-04525-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND The interest in targeted cancer therapies has been growing rapidly. While numerous cancer biomarkers and targeted treatment strategies have been developed and employed, there are still significant limitations and challenges in the early diagnosis and targeted treatment of cancers. Accordingly, there is an urgent need to identify novel targets and develop new targeted drugs. METHODS The study was conducted using combined cis-Mendelian randomization (cis-MR) and colocalization analysis. We analyzed data from 732 plasma proteins to identify potential drug targets associated with eight site-specific cancers. These findings were further validated using the UK Biobank dataset. Then, a protein-protein interaction network was also constructed to examine the interplay between the identified proteins and the targets of existing cancer medications. RESULTS This MR analysis revealed associations between five plasma proteins and prostate cancer, five with breast cancer, and three with lung cancer. Subsequently, these proteins were classified into four distinct target groups, with a focus on tier 1 and 2 targets due to their higher potential to become drug targets. Our study indicatied that genetically predicted KDELC2 (OR: 0.89, 95% CI 0.86-0.93) and TNFRSF10B (OR: 0.74, 95% CI 0.65-0.83) are inversely associated with prostate cancer. Furthermore, we observed an inverse association between CPNE1 (OR: 0.96, 95% CI 0.94-0.98) and breast cancer, while PDIA3 (OR: 1.19, 95% CI 1.10-1.30) were found to be associated with the risk of breast cancer. In addition, we also propose that SPINT2 (OR: 1.05, 95% CI 1.03-1.06), GSTP1 (OR: 0.82, 95% CI 0.74-0.90), and CTSS (OR: 0.91, 95% CI 0.88-0.95) may serve as potential therapeutic targets in prostate cancer. Similarly, GDI2 (OR: 0.85, 95% CI 0.80-0.91), ISLR2 (OR: 0.87, 95% CI 0.82-0.93), and CTSF (OR: 1.14, 95% CI 1.08-1.21) could potentially be targets for breast cancer. Additionally, we identified SFTPB (OR: 0.93, 95% CI 0.91-0.95), ICAM5 (OR: 0.95, 95% CI 0.93-0.97), and FLRT3 (OR: 1.10, 95% CI 1.05-1.15) as potential targets for lung cancer. Notably, TNFRSF10B, GSTP1, and PDIA3 were found to interact with the target proteins of current medications used in prostate or breast cancer treatment. CONCLUSIONS This comprehensive analysis has highlighted thirteen plasma proteins with potential roles in three site-specific cancers. Continued research in this area may reveal their therapeutic potential, particularly KDELC2, TNFRSF10B, CPNE1, and PDIA3, paving the way for more effective cancer treatments.
Collapse
Affiliation(s)
- Feihong Ren
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qiubai Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xuelei Ren
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
99
|
Wang Y, Wang J, Yan Z, Liu S, Xu W. Potential drug targets for asthma identified in the plasma and brain through Mendelian randomization analysis. Front Immunol 2023; 14:1240517. [PMID: 37809092 PMCID: PMC10551444 DOI: 10.3389/fimmu.2023.1240517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
Background Asthma is a heterogeneous disease, and the involvement of neurogenic inflammation is crucial in its development. The standardized treatments focus on alleviating symptoms. Despite the availability of medications for asthma, they have proven to be inadequate in controlling relapses and halting the progression of the disease. Therefore, there is a need for novel drug targets to prevent asthma. Methods We utilized Mendelian randomization to investigate potential drug targets for asthma. We analyzed summary statistics from the UK Biobank and then replicated our findings in GWAS data by Demenais et al. and the FinnGen cohort. We obtained genetic instruments for 734 plasma and 73 brain proteins from recently reported GWAS. Next, we utilized reverse causal relationship analysis, Bayesian co-localization, and phenotype scanning as part of our sensitivity analysis. Furthermore, we performed a comparison and protein-protein interaction analysis to identify causal proteins. We also analyzed the possible consequences of our discoveries by the given existing asthma drugs and their targets. Results Using Mendelian randomization analysis, we identified five protein-asthma pairs that were significant at the Bonferroni level (P < 6.35 × 10-5). Specifically, in plasma, we found that an increase of one standard deviation in IL1R1 and ECM1 was associated with an increased risk of asthma, while an increase in ADAM19 was found to be protective. The corresponding odds ratios were 1.03 (95% CI, 1.02-1.04), 1.00 (95% CI, 1.00-1.01), and 0.99 (95% CI, 0.98-0.99), respectively. In the brain, per 10-fold increase in ECM1 (OR, 1.05; 95% CI, 1.03-1.08) and PDLIM4 (OR, 1.05; 95% CI, 1.04-1.07) increased the risk of asthma. Bayesian co-localization found that ECM1 in the plasma (coloc.abf-PPH4 = 0.965) and in the brain (coloc.abf-PPH4 = 0.931) shared the same mutation with asthma. The target proteins of current asthma medications were found to interact with IL1R1. IL1R1 and PDLIM4 were validated in two replication cohorts. Conclusion Our integrative analysis revealed that asthma risk is causally affected by the levels of IL1R1, ECM1, and PDLIM4. The results suggest that these three proteins have the potential to be used as drug targets for asthma, and further investigation through clinical trials is needed.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Otorhinolaryngology, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jiaxi Wang
- Department of Otorhinolaryngology, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhanfeng Yan
- Department of Otorhinolaryngology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Siming Liu
- Department of Otorhinolaryngology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Wenlong Xu
- Department of Otorhinolaryngology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
100
|
Li X, Shen A, Zhao Y, Xia J. Mendelian Randomization Using the Druggable Genome Reveals Genetically Supported Drug Targets for Psychiatric Disorders. Schizophr Bull 2023; 49:1305-1315. [PMID: 37418754 PMCID: PMC10483453 DOI: 10.1093/schbul/sbad100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
BACKGROUND AND HYPOTHESIS Psychiatric disorders impose a huge health and economic burden on modern society. However, there is currently no proven completely effective treatment available, partly owing to the inefficiency of drug target identification and validation. We aim to identify therapeutic targets relevant to psychiatric disorders by conducting Mendelian randomization (MR) analysis. STUDY DESIGN We performed genome-wide MR analysis by integrating expression quantitative trait loci (eQTL) of 4479 actionable genes that encode druggable proteins and genetic summary statistics from genome-wide association studies of psychiatric disorders. After conducting colocalization analysis on the brain MR findings, we employed protein quantitative trait loci (pQTL) data as genetic proposed instruments for intersecting the colocalized genes to provide further genetic evidence. STUDY RESULTS By performing MR and colocalization analysis with eQTL genetic instruments, we obtained 31 promising drug targets for psychiatric disorders, including 21 significant genes for schizophrenia, 7 for bipolar disorder, 2 for depression, 1 for attention deficit and hyperactivity (ADHD) and none for autism spectrum disorder. Combining MR results using pQTL genetic instruments, we finally proposed 8 drug-targeting genes supported by the strongest MR evidence, including gene ACE, BTN3A3, HAPLN4, MAPK3 and NEK4 for schizophrenia, gene NEK4 and HAPLN4 for bipolar disorder, and gene TIE1 for ADHD. CONCLUSIONS Our findings with genetic support were more likely to be to succeed in clinical trials. In addition, our study prioritizes approved drug targets for the development of new therapies and provides critical drug reuse opportunities for psychiatric disorders.
Collapse
Affiliation(s)
- Xiaoyan Li
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Aotian Shen
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Yiran Zhao
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Junfeng Xia
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|