1
|
Sakata Y, Nochioka K, Yasuda S, Ishida K, Shiroto T, Takahashi J, Kasahara S, Abe R, Yamanaka S, Fujihashi T, Hayashi H, Kato S, Horii K, Teramoto K, Tomita T, Miyata S, Sugimura K, Waga I, Nagasaki M, Shimokawa H. Clinical and plasma proteomic characterization of heart failure with supranormal left ventricular ejection fraction: An emerging entity of heart failure. Eur J Heart Fail 2025. [PMID: 40230291 DOI: 10.1002/ejhf.3654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
AIMS The clinical guidelines categorize heart failure (HF) based on left ventricular ejection fraction (LVEF). However, the current LVEF cutoffs, 40% and 50%, may not fully address the underlying characteristics and cardiovascular risk of HF, particularly for HF with higher LVEF. This study aimed to characterize HF with supranormal ejection fraction (HFsnEF) using different LVEF cutoffs (35%, 55%, and 70% for men, and 40%, 60%, and 75% for women). METHODS AND RESULTS This study divided 442 patients from the CHART-Omics study into four groups: HF with reduced ejection fraction (HFrEF) (n = 55, 65.5 years), HF with mildly reduced ejection fraction (HFmrEF) (n = 125, 69.3 years), HF with normal ejection fraction (HFnEF) (n = 215, 69.0 years) and HFsnEF (n = 47, 67.1 years). When clinical backgrounds were adjusted and HFnEF served as the reference, HFsnEF carried an increased hazard ratio (HR) for the composite of cardiovascular death and HF hospitalization of 2.71 (95% confidence interval [CI] 1.10-6.66, p = 0.030), while HFrEF had a HR of 3.14 (95% CI 1.36-7.23, p = 0.007). HFsnEF was characterized by an increase in relative left ventricular wall thickness and a decrease in left ventricular dimensions, whereas increased left ventricular mass and dimensions characterized HFrEF. Quantitative analysis of 4670 plasma proteins showed essential differences between HFsnEF and HFrEF, for example, 'protein synthesis' versus 'cell morphology', 'cellular assembly and organization' and 'nucleic acid metabolism' for underlying pathophysiology, and 'energy production' versus 'connective tissue disorders' and 'cell-to-cell signalling and interaction' for prognostication. CONCLUSIONS Heart failure with supranormal ejection fraction, an unnoticed but emerging entity in HF, carries a similarly increased cardiovascular risk as HFrEF but has unique structural and plasma proteomic characteristics.
Collapse
Affiliation(s)
- Yasuhiko Sakata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Kotaro Nochioka
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koichi Ishida
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Takashi Shiroto
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jun Takahashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shintaro Kasahara
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ruri Abe
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinsuke Yamanaka
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takahide Fujihashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideka Hayashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | - Kanako Teramoto
- Department of Biostatistics, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Tsutomu Tomita
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Satoshi Miyata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Teikyo University Graduate School of Public Health, Tokyo, Japan
| | - Koichiro Sugimura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- International University of Health and Welfare, Narita, Japan
| | - Iwao Waga
- NEC Solution Innovators, Ltd., Tokyo, Japan
| | - Masao Nagasaki
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Suita, Japan
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- International University of Health and Welfare, Narita, Japan
| |
Collapse
|
2
|
Benowitz NL. Predicting Cardiovascular Risk With Use of Various Tobacco Products. Circulation 2025; 151:1006-1008. [PMID: 40193542 DOI: 10.1161/circulationaha.125.073894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Affiliation(s)
- Neal L Benowitz
- Research Program in Clinical Pharmacology, Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
3
|
Lin M, Guo J, Gu Z, Tang W, Tao H, You S, Jia D, Sun Y, Jia P. Machine learning and multi-omics integration: advancing cardiovascular translational research and clinical practice. J Transl Med 2025; 23:388. [PMID: 40176068 PMCID: PMC11966820 DOI: 10.1186/s12967-025-06425-2] [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: 08/06/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
The global burden of cardiovascular diseases continues to rise, making their prevention, diagnosis and treatment increasingly critical. With advancements and breakthroughs in omics technologies such as high-throughput sequencing, multi-omics approaches can offer a closer reflection of the complex physiological and pathological changes in the body from a molecular perspective, providing new microscopic insights into cardiovascular diseases research. However, due to the vast volume and complexity of data, accurately describing, utilising, and translating these biomedical data demands substantial effort. Researchers and clinicians are actively developing artificial intelligence (AI) methods for data-driven knowledge discovery and causal inference using various omics data. These AI approaches, integrated with multi-omics research, have shown promising outcomes in cardiovascular studies. In this review, we outline the methods for integrating machine learning, one of the most successful applications of AI, with omics data and summarise representative AI models developed that leverage various omics data to facilitate the exploration of cardiovascular diseases from underlying mechanisms to clinical practice. Particular emphasis is placed on the effectiveness of using AI to extract potential molecular information to address current knowledge gaps. We discuss the challenges and opportunities of integrating omics with AI into routine diagnostic and therapeutic practices and anticipate the future development of novel AI models for wider application in the field of cardiovascular diseases.
Collapse
Affiliation(s)
- Mingzhi Lin
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Jiuqi Guo
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Zhilin Gu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Wenyi Tang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Hongqian Tao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Shilong You
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Dalin Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China.
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
4
|
Chadwick J, Hinterberg MA, Asselbergs FW, Biegel H, Boersma E, Cappola TP, Chirinos JA, Coresh J, Ganz P, Gordon DA, Kureshi N, Loupey KM, Orlenko A, Ostroff R, Sampson L, Shrestha S, Sweitzer NK, Williams SA, Zhao L, Kardys I, Lanfear DE. Harnessing the Plasma Proteome to Predict Mortality in Heart Failure Subpopulations. Circ Heart Fail 2025; 18:e011208. [PMID: 40052265 PMCID: PMC11995852 DOI: 10.1161/circheartfailure.123.011208] [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: 09/07/2023] [Revised: 01/16/2025] [Accepted: 01/29/2025] [Indexed: 03/30/2025]
Abstract
BACKGROUND We derived and validated proteomic risk scores (PRSs) for heart failure (HF) prognosis that provide absolute risk estimates for all-cause mortality within 1 year. METHODS Plasma samples from individuals with HF with reduced ejection fraction (HFrEF; ejection fraction <40%; training/validation n=1247/762) and preserved ejection fraction (HFpEF; ejection fraction ≥50%; training/validation n=725/785) from 3 independent studies were run on the SomaScan Assay measuring ≈5000 proteins. Machine learning techniques resulted in unique 17- and 14-protein models for HFrEF and HFpEF that predict 1-year mortality. Discrimination was assessed via C-index and 1-year area under the curve (AUC), and survival curves were visualized. PRSs were also compared with Meta-Analysis Global Group in Chronic HF (MAGGIC) score and NT-proBNP (N-terminal pro-B-type natriuretic peptide) measurements and further assessed for sensitivity to disease progression in longitudinal samples (HFrEF: n=396; 1107 samples; HFpEF: n=175; 350 samples). RESULTS In validation, the HFpEF PRS performed significantly better (P≤0.1) for mortality prediction (C-index, 0.79; AUC, 0.82) than MAGGIC (C-index, 0.71; AUC, 0.74) and NT-proBNP (PRS C-index, 0.76 and AUC, 0.81 versus NT-proBNP C-index, 0.72 and AUC, 0.76). The HFrEF PRS performed comparably to MAGGIC (PRS C-index, 0.76 and AUC, 0.83 versus MAGGIC C-index, 0.75 and AUC, 0.84) but had a significantly better C-Index (P=0.026) than NT-proBNP (PRS C-index, 0.75 and AUC, 0.78 versus NT-proBNP C-index, 0.73 and AUC, 0.77). PRS included known HF pathophysiology biomarkers (93%) and novel proteins (7%). Longitudinal assessment revealed that HFrEF and HFpEF PRSs were higher and increased more over time in individuals who experienced a fatal event during follow-up. CONCLUSIONS PRSs can provide valid, accurate, and dynamic prognostic estimates for patients with HF. This approach has the potential to improve longitudinal monitoring of patients and facilitate personalized care.
Collapse
Affiliation(s)
- Jessica Chadwick
- Departments of Clinical Research and Development (J. Chadwick, R.O., K.M.L., S.A.W.), SomaLogic Operating Co Inc, Boulder, CO
| | - Michael A. Hinterberg
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | - Folkert W. Asselbergs
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands (F.W.A.)
- Health Data Research UK and Institute of Health Informatics, University College London, United Kingdom (F.W.A.)
| | - Hannah Biegel
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | - Eric Boersma
- Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.B., I.K.)
| | - Thomas P. Cappola
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia (T.P.C.)
| | - Julio A. Chirinos
- University of Pennsylvania Perelman School of Medicine, Philadelphia (J.A.C.)
| | | | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco (P.G.)
| | | | - Natasha Kureshi
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | - Kelsey M. Loupey
- Departments of Clinical Research and Development (J. Chadwick, R.O., K.M.L., S.A.W.), SomaLogic Operating Co Inc, Boulder, CO
| | - Alena Orlenko
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA (A.O.)
| | - Rachel Ostroff
- Departments of Clinical Research and Development (J. Chadwick, R.O., K.M.L., S.A.W.), SomaLogic Operating Co Inc, Boulder, CO
| | - Laura Sampson
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | - Sama Shrestha
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | | | - Stephen A. Williams
- Departments of Clinical Research and Development (J. Chadwick, R.O., K.M.L., S.A.W.), SomaLogic Operating Co Inc, Boulder, CO
| | - Lei Zhao
- Bristol Myers Squibb, Princeton, NJ (D.A.G., L.Z.)
| | - Isabella Kardys
- Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.B., I.K.)
| | - David E. Lanfear
- Center for Individualized and Genomic Medicine Research, Henry Ford Hospital, Detroit, MI (D.E.L.)
| |
Collapse
|
5
|
Maddaloni E, Nguyen M, Shah SH, Holman RR. Response to Comments on Maddaloni et al. Osteoprotegerin, Osteopontin, and Osteocalcin Are Associated With Cardiovascular Events in Type 2 Diabetes: Insights From EXSCEL. Diabetes Care 2025;48:235-242. Diabetes Care 2025; 48:e61-e62. [PMID: 40117475 DOI: 10.2337/dci24-0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 01/25/2025] [Indexed: 03/23/2025]
Affiliation(s)
- Ernesto Maddaloni
- Experimental Medicine Department, Sapienza University of Rome, Rome, Italy
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, U.K
| | | | | | - Rury R Holman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, U.K
| |
Collapse
|
6
|
Mori Y, van Dijk EHC, Miyake M, Hosoda Y, den Hollander AI, Yzer S, Miki A, Chen LJ, Ahn J, Takahashi A, Morino K, Nakao SY, Hoyng CB, Ng DSC, Cen LP, Chen H, Ng TK, Pang CP, Joo K, Sato T, Sakata Y, Tajima A, Tabara Y, Park KH, Matsuda F, Yamashiro K, Honda S, Nagasaki M, Boon CJF, Tsujikawa A. Genome-wide association and multi-omics analyses provide insights into the disease mechanisms of central serous chorioretinopathy. Sci Rep 2025; 15:9158. [PMID: 40097481 PMCID: PMC11914043 DOI: 10.1038/s41598-025-92210-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Central serous chorioretinopathy (CSC) is a major cause of vision loss, especially in middle-aged men, and its chronic subtype can lead to legal blindness. Despite its clinical importance, the underlying mechanisms of CSC need further clarification. In this study, we conducted a meta-analysis of three genome-wide association studies (GWASs) for CSC consisting of 8811 Asians and Caucasians, followed by replication in an additional 4338 Asians. We identified four genome-wide hits, including a novel hit (rs12960630 at LINC01924-CDH7, Pmeta = 2.97 × 10-9). A phenome-wide association study for rs12960630 showed a positive correlation between its CSC risk allele with plasma cortisol concentration. Expression/splicing quantitative trait loci (QTL) analyses showed an association of all these hits with the expression and/or splicing of genes in genital organs, which may explain the sex differences in CSC. Protein QTL also suggested the protein-level contribution of the complement factor H pathway to CSC pathogenesis.
Collapse
Affiliation(s)
- Yuki Mori
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Elon H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Masahiro Miyake
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan.
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | | | | | - Suzanne Yzer
- Department of Ophthalmology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Akiko Miki
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeeyun Ahn
- Seoul National University, College of Medicine, Seoul, Korea
- SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Ayako Takahashi
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
| | - Kazuya Morino
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shin-Ya Nakao
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Danny S C Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ling-Ping Cen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Haoyu Chen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Kwangsic Joo
- Seoul National University, College of Medicine, Seoul, Korea
- Seoul National University Bundang Hospital, Seongnam, Korea
| | - Takehiro Sato
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasuhiko Sakata
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasuharu Tabara
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Kyu Hyung Park
- Seoul National University, College of Medicine, Seoul, Korea
- Seoul National University Hospital, Seoul, Korea
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Shigeru Honda
- Department of Ophthalmology and Visual Sciences, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masao Nagasaki
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Ophthalmology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
| |
Collapse
|
7
|
Bossuyt PM. Proteomic Prediction Models. Clin Chem 2025; 71:238-240. [PMID: 39658129 DOI: 10.1093/clinchem/hvae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Affiliation(s)
- Patrick M Bossuyt
- Professor of Clinical Epidemiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
8
|
Maretty L, Gill D, Simonsen L, Soh K, Zagkos L, Galanakis M, Sibbesen J, Iglesias MT, Secher A, Valkenborg D, Purnell JQ, Knudsen LB, Tahrani AA, Geybels M. Proteomic changes upon treatment with semaglutide in individuals with obesity. Nat Med 2025; 31:267-277. [PMID: 39753963 PMCID: PMC11750704 DOI: 10.1038/s41591-024-03355-2] [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: 03/22/2024] [Accepted: 10/14/2024] [Indexed: 01/23/2025]
Abstract
Obesity and type 2 diabetes are prevalent chronic diseases effectively managed by semaglutide. Here we studied the effects of semaglutide on the circulating proteome using baseline and end-of-treatment serum samples from two phase 3 trials in participants with overweight or obesity, with or without diabetes: STEP 1 (n = 1,311) and STEP 2 (n = 645). We identified evidence supporting broad effects of semaglutide, implicating processes related to body weight regulation, glycemic control, lipid metabolism and inflammatory pathways. Several proteins were regulated with semaglutide, after accounting for changes in body weight and HbA1c at end of trial, suggesting effects of semaglutide on the proteome beyond weight loss and glucose lowering. A comparison of semaglutide with real-world proteomic profiles revealed potential benefits on disease-specific proteomic signatures including the downregulation of specific proteins associated with cardiovascular disease risk, supporting its reported effects of lowering cardiovascular disease risk and potential drug repurposing opportunities. This study showcases the potential of proteomics data gathered from randomized trials for providing insights into disease mechanisms and drug repurposing opportunities. These data also highlight the unmet need for, and importance of, examining proteomic changes in response to weight loss pharmacotherapy in future trials.
Collapse
Affiliation(s)
- Lasse Maretty
- Data Science, Novo Nordisk A/S, Søborg, Denmark
- QIAGEN A/S, Aarhus, Denmark
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Sequoia Genetics, London, UK
| | | | - Keng Soh
- Data Science, Novo Nordisk A/S, Søborg, Denmark
| | - Loukas Zagkos
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Michael Galanakis
- Data Science, Novo Nordisk A/S, Søborg, Denmark
- Center for Statistics and Data Science Institute, Hasselt University, Hasselt, Belgium
| | | | | | - Anna Secher
- Brain and Adipose Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Dirk Valkenborg
- Center for Statistics and Data Science Institute, Hasselt University, Hasselt, Belgium
| | | | | | - Abd A Tahrani
- Medical & Science, Novo Nordisk A/S, Søborg, Denmark.
- Department of Metabolism and Systems Science, University of Birmingham, Birmingham, UK.
| | - Milan Geybels
- Data Science, Novo Nordisk A/S, Søborg, Denmark
- Genmab A/S, Valby, Denmark
| |
Collapse
|
9
|
Poudel S, Shrestha H, Pan Y, Li Q, Li K, Im C, Dixon SB, Ehrhardt MJ, Mulrooney DA, Zhou S, Tan H, High AA, Burridge PW, Bhatia S, Jefferies JL, Ness KK, Hudson MM, Robison LL, Armstrong GT, Peng J, Ky B, Yasui Y, Sapkota Y. Serum Proteins Predict Treatment-Related Cardiomyopathy Among Survivors of Childhood Cancer. JACC CardioOncol 2025; 7:56-67. [PMID: 39896123 PMCID: PMC11782007 DOI: 10.1016/j.jaccao.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/17/2024] [Accepted: 10/04/2024] [Indexed: 02/04/2025] Open
Abstract
Background Anthracyclines, a highly effective chemotherapy for many pediatric malignancies, cause cardiomyopathy, a major late effect in adult survivors. Biomarkers are needed for early detection and targeted interventions for anthracycline-associated cardiomyopathy. Objectives The aim of this study was to determine if serum proteins and/or metabolites in asymptomatic childhood cancer survivors can discriminate symptomatic cardiomyopathy. Methods Using an untargeted mass spectrometry-based approach, 867 proteins and 218 metabolites were profiled in serum samples of 75 asymptomatic survivors with subclinical cardiomyopathy and 75 individually matched survivors without cardiomyopathy from SJLIFE (St. Jude Lifetime Cohort Study). Models were developed on the basis of the most influential differentially expressed proteins and metabolites, using conditional logistic regression with a least absolute shrinkage and selection operator penalty. The best performing model was evaluated in 23 independent survivors with severe or symptomatic cardiomyopathy and 23 individually matched cardiomyopathy-free survivors. Results A 27-protein model identified using conditional logistic regression with a least absolute shrinkage and selection operator penalty discriminated symptomatic or severe cardiomyopathy requiring heart failure medications in independent survivors; 19 of 23 individually matched survivors with and without cardiomyopathy were correctly discriminated with 82.6% (95% CI: 71.4%-93.8%) accuracy. Pathway enrichment analysis revealed that the 27 proteins were enriched in various biological processes, many of which have been linked to anthracycline-related cardiomyopathy. Conclusions A risk model was developed on the basis of the differential expression of serum proteins in subclinical cardiomyopathy, which accurately discriminated the risk for severe cardiomyopathy in an independent, matched sample. Further assessment of these proteins as biomarkers of cardiomyopathy risk should be conducted in external larger cohorts and through prospective studies.
Collapse
Affiliation(s)
- Suresh Poudel
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Him Shrestha
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yue Pan
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Qian Li
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kendrick Li
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Cindy Im
- University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | - Suiping Zhou
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Haiyan Tan
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Anthony A. High
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | - Smita Bhatia
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John L. Jefferies
- University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Kirsten K. Ness
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | | | | | - Junmin Peng
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Bonnie Ky
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yutaka Yasui
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yadav Sapkota
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
10
|
Gregorich ZR. Can we use proteomics to predict cardiovascular events? Expert Rev Proteomics 2024:1-4. [PMID: 39699024 DOI: 10.1080/14789450.2024.2445248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Affiliation(s)
- Zachery R Gregorich
- Department of Animal and Dairy Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
11
|
Kiseleva OI, Arzumanian VA, Ikhalaynen YA, Kurbatov IY, Kryukova PA, Poverennaya EV. Multiomics of Aging and Aging-Related Diseases. Int J Mol Sci 2024; 25:13671. [PMID: 39769433 PMCID: PMC11677528 DOI: 10.3390/ijms252413671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Despite their astonishing biological diversity, surprisingly few shared traits connect all or nearly all living organisms. Aging, i.e., the progressive and irreversible decline in the function of multiple cells and tissues, is one of these fundamental features of all organisms, ranging from single-cell creatures to complex animals, alongside variability, adaptation, growth, healing, reproducibility, mobility, and, finally, death. Age is a key determinant for many pathologies, shaping the risks of incidence, severity, and treatment outcomes for cancer, neurodegeneration, heart failure, sarcopenia, atherosclerosis, osteoporosis, and many other diseases. In this review, we aim to systematically investigate the age-related features of the development of several diseases through the lens of multiomics: from genome instability and somatic mutations to pathway alterations and dysregulated metabolism.
Collapse
Affiliation(s)
- Olga I. Kiseleva
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| | - Viktoriia A. Arzumanian
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| | - Yuriy A. Ikhalaynen
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Ilya Y. Kurbatov
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| | - Polina A. Kryukova
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| | - Ekaterina V. Poverennaya
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10/8, 119121 Moscow, Russia; (V.A.A.); (Y.A.I.); (I.Y.K.); (P.A.K.); (E.V.P.)
| |
Collapse
|
12
|
Schuermans A, Pournamdari AB, Lee J, Bhukar R, Ganesh S, Darosa N, Small AM, Yu Z, Hornsby W, Koyama S, Kooperberg C, Reiner AP, Januzzi JL, Honigberg MC, Natarajan P. Integrative proteomic analyses across common cardiac diseases yield mechanistic insights and enhanced prediction. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1516-1530. [PMID: 39572695 PMCID: PMC11634769 DOI: 10.1038/s44161-024-00567-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Cardiac diseases represent common highly morbid conditions for which molecular mechanisms remain incompletely understood. Here we report the analysis of 1,459 protein measurements in 44,313 UK Biobank participants to characterize the circulating proteome associated with incident coronary artery disease, heart failure, atrial fibrillation and aortic stenosis. Multivariable-adjusted Cox regression identified 820 protein-disease associations-including 441 proteins-at Bonferroni-adjusted P < 8.6 × 10-6. Cis-Mendelian randomization suggested causal roles aligning with epidemiological findings for 4% of proteins identified in primary analyses, prioritizing therapeutic targets across cardiac diseases (for example, spondin-1 for atrial fibrillation and the Kunitz-type protease inhibitor 1 for coronary artery disease). Interaction analyses identified seven protein-disease associations that differed Bonferroni-significantly by sex. Models incorporating proteomic data (versus clinical risk factors alone) improved prediction for coronary artery disease, heart failure and atrial fibrillation. These results lay a foundation for future investigations to uncover disease mechanisms and assess the utility of protein-based prevention strategies for cardiac diseases.
Collapse
Affiliation(s)
- Art Schuermans
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Ashley B Pournamdari
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jiwoo Lee
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rohan Bhukar
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Shriienidhie Ganesh
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nicholas Darosa
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Aeron M Small
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Zhi Yu
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Whitney Hornsby
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Satoshi Koyama
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James L Januzzi
- Baim Institute for Clinical Research, Boston, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Michael C Honigberg
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Pradeep Natarajan
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Du S, Chen J, Kim H, Lichtenstein AH, Yu B, Appel LJ, Coresh J, Rebholz CM. Protein Biomarkers of Ultra-Processed Food Consumption and Risk of Coronary Heart Disease, Chronic Kidney Disease, and All-Cause Mortality. J Nutr 2024; 154:3235-3245. [PMID: 39299474 PMCID: PMC11600079 DOI: 10.1016/j.tjnut.2024.08.029] [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: 05/14/2024] [Revised: 07/01/2024] [Accepted: 08/01/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND There is a need to understand the underlying biological mechanisms through which ultra-processed foods negatively affect health. Proteomics offers a valuable tool with which to examine different aspects of ultra-processed foods and their impact on health. OBJECTIVES The aim of this study was to identify protein biomarkers of usual ultra-processed food consumption and assess their relation to the incidence of coronary heart disease (CHD), chronic kidney disease (CKD), and all-cause mortality risk. METHODS A total of 9361 participants from the Atherosclerosis Risk in Communities visit 3 (1993-1995) were included. Dietary intake was assessed using a 66-item food-frequency questionnaire and the processing levels were categorized on the basis of the Nova classification. Plasma proteins were detected using an aptamer-based proteomic assay. We used multivariable linear regressions to examine the association between ultra-processed food and proteins, and Cox proportional hazard models to identify associations between ultra-processed food-related proteins and health outcomes. Models extensively controlled for sociodemographic characteristics, health behaviors, and clinical factors. RESULTS Eight proteins (6 positive, 2 negative) were identified as significantly associated with ultra-processed food consumption. Over a median follow-up of 22 y, there were 1276, 3084, and 5127 cases of CHD, CKD, and death, respectively. Three, 5, and 3 ultra-processed food-related proteins were associated with each outcome, respectively. One protein (β-glucuronidase) was significantly associated with a higher risk of all 3 outcomes, and 3 proteins (receptor-type tyrosine-protein phosphatase U, C-C motif chemokine 25, and twisted gastrulation protein homolog 1) were associated with a higher risk of 2 outcomes. CONCLUSIONS We identified a panel of protein biomarkers that were significantly associated with ultra-processed food consumption. These proteins may be considered potential biomarkers for ultra-processed food intake and may elucidate the biological processes through which ultra-processed foods impact health outcomes.
Collapse
Affiliation(s)
- Shutong Du
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, United States; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Jingsha Chen
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, United States; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Hyunju Kim
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, United States
| | - Alice H Lichtenstein
- Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Lawrence J Appel
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, United States; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Josef Coresh
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States; Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Casey M Rebholz
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, United States; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
14
|
Mazidi M, Wright N, Yao P, Kartsonaki C, Millwood IY, Fry H, Said S, Pozarickij A, Pei P, Chen Y, Wang B, Avery D, Du H, Schmidt DV, Yang L, Lv J, Yu C, Sun D, Chen J, Hill M, Peto R, Collins R, Bennett DA, Walters RG, Li L, Clarke R, Chen Z. Risk prediction of ischemic heart disease using plasma proteomics, conventional risk factors and polygenic scores in Chinese and European adults. Eur J Epidemiol 2024; 39:1229-1240. [PMID: 39578299 PMCID: PMC11646273 DOI: 10.1007/s10654-024-01168-8] [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: 03/13/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024]
Abstract
Plasma proteomics could enhance risk prediction for multiple diseases beyond conventional risk factors or polygenic scores (PS). To assess utility of proteomics for risk prediction of ischemic heart disease (IHD) compared with conventional risk factors and PS in Chinese and European populations. A nested case-cohort study measured plasma levels of 2923 proteins using Olink Explore panel in ~ 4000 Chinese adults (1976 incident IHD cases and 2001 sub-cohort controls). We used conventional and machine learning (Boruta) methods to develop proteomics-based prediction models of IHD, with discrimination assessed using area under the curve (AUC), C-statistics and net reclassification index (NRI). These were compared with conventional risk factors and PS in Chinese and in 37,187 Europeans. Overall, 446 proteins were associated with IHD (false discovery rate < 0.05) in Chinese after adjustment for conventional cardiovascular disease risk factors. Proteomic risk models alone yielded higher C-statistics for IHD than conventional risk factors or PS (0.855 [95%CI 0.841-0.868] vs. 0.845 [0.829-0.860] vs 0.553 [0.528-0.578], respectively). Addition of 446 proteins to PS improved C-statistics to 0.857 (0.843-0.871) and NRI by 109.1%; and addition to conventional risk factors improved C-statistics to 0.868 (0.854-0.882) and NRI by 86.9%. Boruta analysis identified 30 proteins accounting for ~ 90% of improvement in NRI for IHD conferred by all 2923 proteins. Similar proteomic panels yielded comparable improvements in risk prediction of IHD in Europeans. Plasma proteomics improved risk prediction of IHD beyond conventional risk factors and PS and could enhance precision medicine approaches for primary prevention of IHD.
Collapse
Affiliation(s)
- Mohsen Mazidi
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Pang Yao
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Iona Y Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Hannah Fry
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Saredo Said
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Alfred Pozarickij
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Pei Pei
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Baihan Wang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Daniel Avery
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Huaidong Du
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Dan Valle Schmidt
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Ling Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major (Peking University), Ministry of Education, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major (Peking University), Ministry of Education, Beijing, China
| | - DianJianYi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major (Peking University), Ministry of Education, Beijing, China
| | - Junshi Chen
- China National Center for Food Risk Assessment, Beijing, China
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Richard Peto
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Rory Collins
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Derrick A Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Robin G Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major (Peking University), Ministry of Education, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK.
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK.
| |
Collapse
|
15
|
Kraemer S, Schneider DJ, Paterson C, Perry D, Westacott MJ, Hagar Y, Katilius E, Lynch S, Russell TM, Johnson T, Astling DP, DeLisle RK, Cleveland J, Gold L, Drolet DW, Janjic N. Crossing the Halfway Point: Aptamer-Based, Highly Multiplexed Assay for the Assessment of the Proteome. J Proteome Res 2024; 23:4771-4788. [PMID: 39038188 PMCID: PMC11536431 DOI: 10.1021/acs.jproteome.4c00411] [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: 05/10/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
Measuring responses in the proteome to various perturbations improves our understanding of biological systems. The value of information gained from such studies is directly proportional to the number of proteins measured. To overcome technical challenges associated with highly multiplexed measurements, we developed an affinity reagent-based method that uses aptamers with protein-like side chains along with an assay that takes advantage of their unique properties. As hybrid affinity reagents, modified aptamers are fully comparable to antibodies in terms of binding characteristics toward proteins, including epitope size, shape complementarity, affinity and specificity. Our assay combines these intrinsic binding properties with serial kinetic proofreading steps to allow highly effective partitioning of stable specific complexes from unstable nonspecific complexes. The use of these orthogonal methods to enhance specificity effectively overcomes the severe limitation to multiplexing inherent to the use of sandwich-based methods. Our assay currently measures half of the unique proteins encoded in the human genome with femtomolar sensitivity, broad dynamic range and exceptionally high reproducibility. Using machine learning to identify patterns of change, we have developed tests based on measurement of multiple proteins predictive of current health states and future disease risk to guide a holistic approach to precision medicine.
Collapse
Affiliation(s)
- Stephan Kraemer
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Daniel J. Schneider
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Clare Paterson
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Darryl Perry
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Matthew J. Westacott
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Yolanda Hagar
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Evaldas Katilius
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Sean Lynch
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Theresa M. Russell
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Ted Johnson
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - David P. Astling
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Robert Kirk DeLisle
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Jason Cleveland
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Larry Gold
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Daniel W. Drolet
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Nebojsa Janjic
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| |
Collapse
|
16
|
Liu M, Zhang Y, Ye Z, He P, Zhou C, Yang S, Zhang Y, Gan X, Qin X. Enhanced prediction of atrial fibrillation risk using proteomic markers: a comparative analysis with clinical and polygenic risk scores. Heart 2024; 110:1270-1276. [PMID: 39237126 DOI: 10.1136/heartjnl-2024-324274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Proteomic biomarkers have shown promise in predicting various cardiovascular conditions, but their utility in assessing the risk of atrial fibrillation (AF) remains unclear. This study aimed to develop and validate a protein-based risk score for predicting incident AF and to compare its predictive performance with traditional clinical risk factors and polygenic risk scores in a large cohort from the UK Biobank. METHODS We analysed data from 36 129 white British individuals without prior AF, assessing 2923 plasma proteins using the Olink Explore 3072 assay. The cohort was divided into a training set (70%) and a test set (30%) to develop and validate a protein risk score for AF. We compared the predictive performance of this score with the HARMS2-AF risk model and a polygenic risk score. RESULTS Over an average follow-up of 11.8 years, 2450 incident AF cases were identified. A 47-protein risk score was developed, with N-terminal prohormone of brain natriuretic peptide (NT-proBNP) being the most significant predictor. In the test set, the protein risk score (per SD increment, HR 1.94; 95% CI 1.83 to 2.05) and NT-proBNP alone (HR 1.80; 95% CI 1.70 to 1.91) demonstrated superior predictive performance (C-statistic: 0.802 and 0.785, respectively) compared with HARMS2-AF and polygenic risk scores (C-statistic: 0.751 and 0.748, respectively). CONCLUSIONS A protein-based risk score, particularly incorporating NT-proBNP, offers superior predictive value for AF risk over traditional clinical and polygenic risk scores, highlighting the potential for proteomic data in AF risk stratification.
Collapse
Affiliation(s)
- Mengyi Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| | - Yuanyuan Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| | - Ziliang Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| | - Panpan He
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| | - Chun Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| | - Sisi Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| | - Yanjun Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| | - Xiaoqin Gan
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University; National Clinical Research Center for Kidney Disease; State Key Laboratory of Organ Failure Research; Guangdong Provincial Institute of Nephrology; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Royer P, Björnson E, Adiels M, Josefson R, Hagberg E, Gummesson A, Bergström G. Large-scale plasma proteomics in the UK Biobank modestly improves prediction of major cardiovascular events in a population without previous cardiovascular disease. Eur J Prev Cardiol 2024; 31:1681-1689. [PMID: 38546334 DOI: 10.1093/eurjpc/zwae124] [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: 01/22/2024] [Revised: 03/06/2024] [Accepted: 03/24/2024] [Indexed: 10/11/2024]
Abstract
AIMS Improved identification of individuals at high risk of developing cardiovascular disease would enable targeted interventions and potentially lead to reductions in mortality and morbidity. Our aim was to determine whether use of large-scale proteomics improves prediction of cardiovascular events beyond traditional risk factors (TRFs). METHODS AND RESULTS Using proximity extension assays, 2919 plasma proteins were measured in 38 380 participants of the UK Biobank. Both data- and literature-based feature selection and trained models using extreme gradient boosting machine learning were used to predict risk of major cardiovascular events (MACEs: fatal and non-fatal myocardial infarction, stroke, and coronary artery revascularization) during a 10-year follow-up. Area under the curve (AUC) and net reclassification index (NRI) were used to evaluate the additive value of selected protein panels to MACE prediction by Systematic COronary Risk Evaluation 2 (SCORE2) or the 10 TRFs used in SCORE2. SCORE2 and SCORE2 refitted to UK Biobank data predicted MACE with AUCs of 0.740 and 0.749, respectively. Data-driven selection identified 114 proteins of greatest relevance for prediction. Prediction of MACE was not improved by using these proteins alone (AUC of 0.758) but was significantly improved by combining these proteins with SCORE2 or the 10 TRFs (AUC = 0.771, P < 001, NRI = 0.140, and AUC = 0.767, P = 0.03, NRI 0.053, respectively). Literature-based protein selection (113 proteins from five previous studies) also improved risk prediction beyond TRFs while a random selection of 114 proteins did not. CONCLUSION Large-scale plasma proteomics with data-driven and literature-based protein selection modestly improves prediction of future MACE beyond TRFs.
Collapse
Affiliation(s)
- Patrick Royer
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Critical Care, University Hospital of Martinique, Fort-de-France, Martinique, French West Indies, France
| | - Elias Björnson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
| | - Martin Adiels
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rebecca Josefson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
| | - Eva Hagberg
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Anders Gummesson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| |
Collapse
|
18
|
Greenland P, Segal MR, McNeil RB, Parker CB, Pemberton VL, Grobman WA, Silver RM, Simhan HN, Saade GR, Ganz P, Mehta P, Catov JM, Bairey Merz CN, Varagic J, Khan SS, Parry S, Reddy UM, Mercer BM, Wapner RJ, Haas DM. Large-Scale Proteomics in Early Pregnancy and Hypertensive Disorders of Pregnancy. JAMA Cardiol 2024; 9:791-799. [PMID: 38958943 PMCID: PMC11223045 DOI: 10.1001/jamacardio.2024.1621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/29/2024] [Indexed: 07/04/2024]
Abstract
Importance There is no consensus regarding the best method for prediction of hypertensive disorders of pregnancy (HDP), including gestational hypertension and preeclampsia. Objective To determine predictive ability in early pregnancy of large-scale proteomics for prediction of HDP. Design, Setting, and Participants This was a nested case-control study, conducted in 2022 to 2023, using clinical data and plasma samples collected between 2010 and 2013 during the first trimester, with follow-up until pregnancy outcome. This multicenter observational study took place at 8 academic medical centers in the US. Nulliparous individuals during first-trimester clinical visits were included. Participants with HDP were selected as cases; controls were selected from those who delivered at or after 37 weeks without any HDP, preterm birth, or small-for-gestational-age infant. Age, self-reported race and ethnicity, body mass index, diabetes, health insurance, and fetal sex were available covariates. Exposures Proteomics using an aptamer-based assay that included 6481 unique human proteins was performed on stored plasma. Covariates were used in predictive models. Main Outcomes and Measures Prediction models were developed using the elastic net, and analyses were performed on a randomly partitioned training dataset comprising 80% of study participants, with the remaining 20% used as an independent testing dataset. Primary measure of predictive performance was area under the receiver operating characteristic curve (AUC). Results This study included 753 HDP cases and 1097 controls with a mean (SD) age of 26.9 (5.5) years. Maternal race and ethnicity were 51 Asian (2.8%), 275 non-Hispanic Black (14.9%), 275 Hispanic (14.9%), 1161 non-Hispanic White (62.8% ), and 88 recorded as other (4.8%), which included those who did not identify according to these designations. The elastic net model, allowing for forced inclusion of prespecified covariates, was used to adjust protein-based models for clinical and demographic variables. Under this approach, no proteins were selected to augment the clinical and demographic covariates. The predictive performance of the resulting model was modest, with a training set AUC of 0.64 (95% CI, 0.61-0.67) and a test set AUC of 0.62 (95% CI, 0.56-0.68). Further adjustment for study site yielded only minimal changes in AUCs. Conclusions and Relevance In this case-control study with detailed clinical data and stored plasma samples available in the first trimester, an aptamer-based proteomics panel did not meaningfully add to predictive utility over and above clinical and demographic factors that are routinely available.
Collapse
Affiliation(s)
- Philip Greenland
- Departments of Medicine and Preventive Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Mark R. Segal
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | | | | | - Victoria L. Pemberton
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - William A. Grobman
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Now with Department of Obstetrics and Gynecology, The Ohio State University, Columbus
| | - Robert M. Silver
- Department of Obstetrics and Gynecology, University of Utah Health, Salt Lake City
| | - Hyagriv N. Simhan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George R. Saade
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology at UTMB Health, Galveston, Texas
- Now with Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk
| | - Peter Ganz
- Department of Medicine, Zuckerberg San Francisco General Hospital and University of California, San Francisco
| | - Priya Mehta
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Janet M. Catov
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh and Magee-Women’s Research Institute, Pittsburgh, Pennsylvania
| | - C. Noel Bairey Merz
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jasmina Varagic
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sadiya S. Khan
- Division of Cardiology, Department of Medicine and Department of Preventive Medicine, Northwestern University, Chicago, Illinois
| | - Samuel Parry
- Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Uma M. Reddy
- Maternal & Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Brian M. Mercer
- Department of Obstetrics & Gynecology, Case Western Reserve University—The MetroHealth System, Cleveland, Ohio
| | - Ronald J. Wapner
- Clinical Genetics and Genomics, Maternal & Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - David M. Haas
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis
| |
Collapse
|
19
|
Carrasco-Zanini J, Pietzner M, Davitte J, Surendran P, Croteau-Chonka DC, Robins C, Torralbo A, Tomlinson C, Grünschläger F, Fitzpatrick N, Ytsma C, Kanno T, Gade S, Freitag D, Ziebell F, Haas S, Denaxas S, Betts JC, Wareham NJ, Hemingway H, Scott RA, Langenberg C. Proteomic signatures improve risk prediction for common and rare diseases. Nat Med 2024; 30:2489-2498. [PMID: 39039249 PMCID: PMC11405273 DOI: 10.1038/s41591-024-03142-z] [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: 10/26/2023] [Accepted: 06/19/2024] [Indexed: 07/24/2024]
Abstract
For many diseases there are delays in diagnosis due to a lack of objective biomarkers for disease onset. Here, in 41,931 individuals from the United Kingdom Biobank Pharma Proteomics Project, we integrated measurements of ~3,000 plasma proteins with clinical information to derive sparse prediction models for the 10-year incidence of 218 common and rare diseases (81-6,038 cases). We then compared prediction models developed using proteomic data with models developed using either basic clinical information alone or clinical information combined with data from 37 clinical assays. The predictive performance of sparse models including as few as 5 to 20 proteins was superior to the performance of models developed using basic clinical information for 67 pathologically diverse diseases (median delta C-index = 0.07; range = 0.02-0.31). Sparse protein models further outperformed models developed using basic information combined with clinical assay data for 52 diseases, including multiple myeloma, non-Hodgkin lymphoma, motor neuron disease, pulmonary fibrosis and dilated cardiomyopathy. For multiple myeloma, single-cell RNA sequencing from bone marrow in newly diagnosed patients showed that four of the five predictor proteins were expressed specifically in plasma cells, consistent with the strong predictive power of these proteins. External replication of sparse protein models in the EPIC-Norfolk study showed good generalizability for prediction of the six diseases tested. These findings show that sparse plasma protein signatures, including both disease-specific proteins and protein predictors shared across several diseases, offer clinically useful prediction of common and rare diseases.
Collapse
Affiliation(s)
- Julia Carrasco-Zanini
- Human Genetics and Genomics, GSK Research and Development, Stevenage, UK.
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Maik Pietzner
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jonathan Davitte
- Human Genetics and Genomics, GSK Research and Development, Collegeville, PA, USA
| | - Praveen Surendran
- Human Genetics and Genomics, GSK Research and Development, Stevenage, UK
| | | | - Chloe Robins
- Human Genetics and Genomics, GSK Research and Development, Collegeville, PA, USA
| | - Ana Torralbo
- Institute of Health Informatics, University College London, London, UK
| | - Christopher Tomlinson
- Institute of Health Informatics, University College London, London, UK
- National Institute for Health Research, Biomedical Research Centre, University College London Hospitals NHS Trust, London, UK
| | - Florian Grünschläger
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | - Cai Ytsma
- Institute of Health Informatics, University College London, London, UK
| | - Tokuwa Kanno
- Human Genetics and Genomics, GSK Research and Development, Collegeville, PA, USA
| | - Stephan Gade
- Genomic Sciences, Cellzome GmbH, GSK Research and Development, Heidelberg, Germany
| | - Daniel Freitag
- Human Genetics and Genomics, GSK Research and Development, Stevenage, UK
| | - Frederik Ziebell
- Genomic Sciences, Cellzome GmbH, GSK Research and Development, Heidelberg, Germany
| | - Simon Haas
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Spiros Denaxas
- Institute of Health Informatics, University College London, London, UK
- National Institute for Health Research, Biomedical Research Centre, University College London Hospitals NHS Trust, London, UK
- Health Data Research UK, London, UK
- British Heart Foundation Data Science Centre, London, UK
| | - Joanna C Betts
- Human Genetics and Genomics, GSK Research and Development, Stevenage, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Harry Hemingway
- Institute of Health Informatics, University College London, London, UK
- National Institute for Health Research, Biomedical Research Centre, University College London Hospitals NHS Trust, London, UK
- Health Data Research UK, London, UK
| | - Robert A Scott
- Human Genetics and Genomics, GSK Research and Development, Stevenage, UK.
| | - Claudia Langenberg
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
20
|
Rajagopalan S, Dobre M, Dazard JE, Vergara-Martel A, Connelly K, Farkouh ME, Gaztanaga J, Conger H, Dever A, Razavi-Nematollahi L, Fares A, Pereira G, Edwards-Glenn J, Cameron M, Cameron C, Al-Kindi S, Brook RD, Pitt B, Weir M. Mineralocorticoid Receptor Antagonism Prevents Aortic Plaque Progression and Reduces Left Ventricular Mass and Fibrosis in Patients With Type 2 Diabetes and Chronic Kidney Disease: The MAGMA Trial. Circulation 2024; 150:663-676. [PMID: 39129649 PMCID: PMC11503525 DOI: 10.1161/circulationaha.123.067620] [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: 10/16/2023] [Accepted: 06/12/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Persistent mineralocorticoid receptor activation is a pathologic response in type 2 diabetes and chronic kidney disease. Whereas mineralocorticoid receptor antagonists are beneficial in reducing cardiovascular complications, direct mechanistic pathways for these effects in humans are lacking. METHODS The MAGMA trial (Mineralocorticoid Receptor Antagonism Clinical Evaluation in Atherosclerosis) was a randomized, double-blind, placebo-controlled trial in patients with high-risk type 2 diabetes with chronic kidney disease (not receiving dialysis) on maximum tolerated renin-angiotensin system blockade. The primary end point was change in thoracic aortic wall volume, expressed as absolute or percent value (ΔTWV or ΔPWV), using 3T magnetic resonance imaging at 12 months. Secondary end points were changes in left ventricle (LV) mass; LV fibrosis, measured as a change in myocardial native T1; and 24-hour ambulatory and central aortic blood pressures. Tertiary end points included plasma proteomic changes in 7596 plasma proteins using an aptamer-based assay. RESULTS A total of 79 patients were randomized to placebo (n=42) or 25 mg of spironolactone daily (n=37). After a modified intent-to-treat, including available baseline data of study end points, patients who completed the trial protocol were included in the final analyses. At the 12-month follow-up, the average change in PWV was 7.1±10.7% in the placebo group and 0.87±10.0% in the spironolactone group (P=0.028), and ΔTWV was 1.2±1.7 cm3 in the placebo group and 0.037±1.9 cm3 in the spironolactone group (P=0.022). Change in LV mass was 3.1±8.4 g in the placebo group and -5.8±8.4 g in the spironolactone group (P=0.001). Changes in LV T1 values were significantly different between the placebo and spironolactone groups (26.0±41.9 ms in the placebo group versus a decrease of -10.1±36.3 ms in the spironolactone group; P=6.33×10-4). Mediation analysis revealed that the spironolactone effect on thoracic aortic wall volume and myocardial mass remained significant after adjustment for ambulatory and central blood pressures. Proteomic analysis revealed a dominant effect of spironolactone on pathways involving oxidative stress, inflammation, and leukocyte activation. CONCLUSIONS Among patients with diabetes with moderate to severe chronic kidney disease at elevated cardiovascular risk, treatment with spironolactone prevented progression of aortic wall volume and resulted in regression of LV mass and favorable alterations in native T1, suggesting amelioration of left-ventricular fibrosis. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02169089.
Collapse
Affiliation(s)
- Sanjay Rajagopalan
- University Hospitals, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Mirela Dobre
- University Hospitals, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Jean-Eudes Dazard
- University Hospitals, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Armando Vergara-Martel
- University Hospitals, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Kim Connelly
- St. Michael’s Hospital, University of Toronto, Toronto, CA
| | | | - Juan Gaztanaga
- New York University Langone Health School of Medicine, Winthrop, Mineola, NY
| | | | - Ann Dever
- University Hospitals, Cleveland, OH, USA
| | | | - Anas Fares
- University Hospitals, Cleveland, OH, USA
| | | | | | - Mark Cameron
- Case Western Reserve University, Cleveland, OH, USA
| | | | - Sadeer Al-Kindi
- Debakey Heart and Vascular Center Houston Methodist Hospital, Houston TX
| | - Robert D. Brook
- University of Michigan Frankel Cardiovascular Center, Detroit, MI
| | | | - Matthew Weir
- Division of Nephrology, University of Maryland Medical Center, Baltimore, MD
| |
Collapse
|
21
|
Dubin RF, Deo R, Ren Y, Wang J, Pico AR, Mychaleckyj JC, Kozlitina J, Arthur V, Lee H, Shah A, Feldman H, Bansal N, Zelnick L, Rao P, Sukul N, Raj DS, Mehta R, Rosas SE, Bhat Z, Weir MR, He J, Chen J, Kansal M, Kimmel PL, Ramachandran VS, Waikar SS, Segal MR, Ganz P. Incident heart failure in chronic kidney disease: proteomics informs biology and risk stratification. Eur Heart J 2024; 45:2752-2767. [PMID: 38757788 PMCID: PMC11313584 DOI: 10.1093/eurheartj/ehae288] [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: 07/30/2023] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND AND AIMS Incident heart failure (HF) among individuals with chronic kidney disease (CKD) incurs hospitalizations that burden patients and health care systems. There are few preventative therapies, and the Pooled Cohort equations to Prevent Heart Failure (PCP-HF) perform poorly in the setting of CKD. New drug targets and better risk stratification are urgently needed. METHODS In this analysis of incident HF, SomaScan V4.0 (4638 proteins) was analysed in 2906 participants of the Chronic Renal Insufficiency Cohort (CRIC) with validation in the Atherosclerosis Risk in Communities (ARIC) study. The primary outcome was 14-year incident HF (390 events); secondary outcomes included 4-year HF (183 events), HF with reduced ejection fraction (137 events), and HF with preserved ejection fraction (165 events). Mendelian randomization and Gene Ontology were applied to examine causality and pathways. The performance of novel multi-protein risk models was compared to the PCP-HF risk score. RESULTS Over 200 proteins were associated with incident HF after adjustment for estimated glomerular filtration rate at P < 1 × 10-5. After adjustment for covariates including N-terminal pro-B-type natriuretic peptide, 17 proteins remained associated at P < 1 × 10-5. Mendelian randomization associations were found for six proteins, of which four are druggable targets: FCG2B, IGFBP3, CAH6, and ASGR1. For the primary outcome, the C-statistic (95% confidence interval [CI]) for the 48-protein model in CRIC was 0.790 (0.735, 0.844) vs. 0.703 (0.644, 0.762) for the PCP-HF model (P = .001). C-statistic (95% CI) for the protein model in ARIC was 0.747 (0.707, 0.787). CONCLUSIONS Large-scale proteomics reveal novel circulating protein biomarkers and potential mediators of HF in CKD. Proteomic risk models improve upon the PCP-HF risk score in this population.
Collapse
Affiliation(s)
- Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, H5.122E, Dallas, TX 75390, USA
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianqiao Wang
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Josyf C Mychaleckyj
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Julia Kozlitina
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hongzhe Lee
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amil Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harold Feldman
- Patient-Centered Outcomes Research Institute, Washington, DC, USA
| | - Nisha Bansal
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Leila Zelnick
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Panduranga Rao
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Nidhi Sukul
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Dominic S Raj
- Division of Kidney Diseases and Hypertension, George Washington University School of Medicine, Washington, DC, USA
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, USA
| | - Sylvia E Rosas
- Joslin Diabetes Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zeenat Bhat
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew R Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Jing Chen
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Mayank Kansal
- Division of Cardiology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Vasan S Ramachandran
- University of Texas School of Public Health San Antonio and the University of Texas Health Sciences Center in San Antonio, Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Peter Ganz
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
22
|
Kim T, Rhee EP. Aptamer-Based Proteomics in CKD. Am J Kidney Dis 2024; 83:825-828. [PMID: 38281681 DOI: 10.1053/j.ajkd.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 01/13/2024] [Indexed: 01/30/2024]
Affiliation(s)
- Taesoo Kim
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Eugene P Rhee
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
23
|
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
|
24
|
Ungvari Z, Tabák AG, Adany R, Purebl G, Kaposvári C, Fazekas-Pongor V, Csípő T, Szarvas Z, Horváth K, Mukli P, Balog P, Bodizs R, Ujma P, Stauder A, Belsky DW, Kovács I, Yabluchanskiy A, Maier AB, Moizs M, Östlin P, Yon Y, Varga P, Vokó Z, Papp M, Takács I, Vásárhelyi B, Torzsa P, Ferdinandy P, Csiszar A, Benyó Z, Szabó AJ, Dörnyei G, Kivimäki M, Kellermayer M, Merkely B. The Semmelweis Study: a longitudinal occupational cohort study within the framework of the Semmelweis Caring University Model Program for supporting healthy aging. GeroScience 2024; 46:191-218. [PMID: 38060158 PMCID: PMC10828351 DOI: 10.1007/s11357-023-01018-7] [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: 09/07/2023] [Accepted: 11/11/2023] [Indexed: 12/08/2023] Open
Abstract
The Semmelweis Study is a prospective occupational cohort study that seeks to enroll all employees of Semmelweis University (Budapest, Hungary) aged 25 years and older, with a population of 8866 people, 70.5% of whom are women. The study builds on the successful experiences of the Whitehall II study and aims to investigate the complex relationships between lifestyle, environmental, and occupational risk factors, and the development and progression of chronic age-associated diseases. An important goal of the Semmelweis Study is to identify groups of people who are aging unsuccessfully and therefore have an increased risk of developing age-associated diseases. To achieve this, the study takes a multidisciplinary approach, collecting economic, social, psychological, cognitive, health, and biological data. The Semmelweis Study comprises a baseline data collection with open healthcare data linkage, followed by repeated data collection waves every 5 years. Data are collected through computer-assisted self-completed questionnaires, followed by a physical health examination, physiological measurements, and the assessment of biomarkers. This article provides a comprehensive overview of the Semmelweis Study, including its origin, context, objectives, design, relevance, and expected contributions.
Collapse
Affiliation(s)
- Zoltan Ungvari
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Adam G Tabák
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- UCL Brain Sciences, University College London, London, UK
- Department of Internal Medicine and Oncology, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Roza Adany
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-UD Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Purebl
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Csilla Kaposvári
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Vince Fazekas-Pongor
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Tamás Csípő
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsófia Szarvas
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Krisztián Horváth
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Peter Mukli
- International Training Program in Geroscience/Healthy Aging Program, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Piroska Balog
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Robert Bodizs
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Peter Ujma
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Adrienne Stauder
- Institute of Behavioral Sciences, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Daniel W Belsky
- Robert N. Butler Columbia Aging Center, Columbia University, New York, NY, USA
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Illés Kovács
- Department of Ophthalmology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York City, NY, USA
- Department of Clinical Ophthalmology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System, Singapore, Singapore
- Department of Human Movement Sciences, @AgeAmsterdam, Vrije Universiteit, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Mariann Moizs
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Ministry of Interior of Hungary, Budapest, Hungary
| | | | - Yongjie Yon
- WHO Regional Office for Europe, Copenhagen, Denmark
| | - Péter Varga
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Clinical Center, Semmelweis University, Budapest, Hungary
| | - Zoltán Vokó
- Center for Health Technology Assessment, Semmelweis University, Budapest, Hungary
| | - Magor Papp
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - István Takács
- UCL Brain Sciences, University College London, London, UK
| | - Barna Vásárhelyi
- Department of Laboratory Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Torzsa
- Department of Family Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltán Benyó
- Department of Translational Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, Budapest, Hungary
| | - Attila J Szabó
- First Department of Pediatrics, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Pediatrics and Nephrology Research Group, Semmelweis University, Budapest, Hungary
| | - Gabriella Dörnyei
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Mika Kivimäki
- UCL Brain Sciences, University College London, London, UK
| | - Miklos Kellermayer
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
25
|
Dark HE, Paterson C, Daya GN, Peng Z, Duggan MR, Bilgel M, An Y, Moghekar A, Davatzikos C, Resnick SM, Loupy K, Simpson M, Candia J, Mosley T, Coresh J, Palta P, Ferrucci L, Shapiro A, Williams SA, Walker KA. Proteomic Indicators of Health Predict Alzheimer's Disease Biomarker Levels and Dementia Risk. Ann Neurol 2024; 95:260-273. [PMID: 37801487 PMCID: PMC10842994 DOI: 10.1002/ana.26817] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/06/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023]
Abstract
OBJECTIVE Few studies have comprehensively examined how health and disease risk influence Alzheimer's disease (AD) biomarkers. The present study examined the association of 14 protein-based health indicators with plasma and neuroimaging biomarkers of AD and neurodegeneration. METHODS In 706 cognitively normal adults, we examined whether 14 protein-based health indices (ie, SomaSignal® tests) were associated with concurrently measured plasma-based biomarkers of AD pathology (amyloid-β [Aβ]42/40 , tau phosphorylated at threonine-181 [pTau-181]), neuronal injury (neurofilament light chain [NfL]), and reactive astrogliosis (glial fibrillary acidic protein [GFAP]), brain volume, and cortical Aβ and tau. In a separate cohort (n = 11,285), we examined whether protein-based health indicators associated with neurodegeneration also predict 25-year dementia risk. RESULTS Greater protein-based risk for cardiovascular disease, heart failure mortality, and kidney disease was associated with lower Aβ42/40 and higher pTau-181, NfL, and GFAP levels, even in individuals without cardiovascular or kidney disease. Proteomic indicators of body fat percentage, lean body mass, and visceral fat were associated with pTau-181, NfL, and GFAP, whereas resting energy rate was negatively associated with NfL and GFAP. Together, these health indicators predicted 12, 31, 50, and 33% of plasma Aβ42/40 , pTau-181, NfL, and GFAP levels, respectively. Only protein-based measures of cardiovascular risk were associated with reduced regional brain volumes; these measures predicted 25-year dementia risk, even among those without clinically defined cardiovascular disease. INTERPRETATION Subclinical peripheral health may influence AD and neurodegenerative disease processes and relevant biomarker levels, particularly NfL. Cardiovascular health, even in the absence of clinically defined disease, plays a central role in brain aging and dementia. ANN NEUROL 2024;95:260-273.
Collapse
Affiliation(s)
- Heather E. Dark
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | | | - Gulzar N. Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Michael R. Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christos Davatzikos
- Section of Biomedical Image Analysis, Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | | | | | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Thomas Mosley
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Priya Palta
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Epidemiology, Columbia Mailman School of Public Health, New York, New York, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Allison Shapiro
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus
| | | | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
26
|
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: 1] [Impact Index Per Article: 1.0] [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
|
27
|
Lind L, Titova O, Zeng R, Zanetti D, Ingelsson M, Gustafsson S, Sundström J, Ärnlöv J, Elmståhl S, Assimes T, Michaëlsson K. Plasma Protein Profiling of Incident Cardiovascular Diseases: A Multisample Evaluation. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:e004233. [PMID: 38014560 DOI: 10.1161/circgen.123.004233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/15/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Proteomic profiling could potentially disclose new pathophysiological pathways for cardiovascular diseases (CVD) and improve prediction at the individual level. We therefore aimed to study the plasma protein profile associated with the incidence of different CVDs. METHODS Plasma levels of 245 proteins suspected to be linked to CVD or metabolism were measured in 4 Swedish prospective population-based cohorts (SIMPLER [Swedish Infrastructure for Medical Population-Based Life-Course and Environmental Research], ULSAM (Uppsala Longitudinal Study of Adult Men), EpiHealth, and POEM [Prospective Investigation of Obesity, Energy Production, and Metabolism]) comprising 11 869 individuals, free of CVD diagnoses at baseline. Our primary CVD outcome was defined by a combined end point that included either incident myocardial infarction, stroke, or heart failure. RESULTS Using a discovery/validation approach, 42 proteins were associated with our primary composite end point occurring in 1163 subjects. In separate meta-analyses for each of the 3 CVD outcomes, 49 proteins were related to myocardial infarction, 34 to ischemic stroke, and 109 to heart failure. Thirteen proteins were related to all 3 outcomes. Of those, urokinase plasminogen activator surface receptor, adrenomedullin, and KIM-1 (kidney injury molecule 1) were also related to several markers of subclinical CVD in Prospective Investigation of Obesity, Energy production and Metabolism, reflecting myocardial or arterial pathologies. In prediction analysis, a lasso selection of 11 proteins in ULSAM improved the discrimination of CVD by 3.3% (P<0.0001) in SIMPLER when added to traditional risk factors. CONCLUSIONS Protein profiling in multiple samples disclosed several new proteins to be associated with subsequent myocardial infarction, stroke, and heart failure, suggesting common pathophysiological pathways for these diseases. KIM-1, urokinase plasminogen activator surface receptor, and adrenomedullin were novel early markers of CVD. A selection of 11 proteins improved the discrimination of CVD.
Collapse
Affiliation(s)
- Lars Lind
- Department of Medical Sciences (L.L., R.Z., S.G., J.S.), Uppsala University, Sweden
| | - Olga Titova
- Department of Surgical Sciences (O.T., K.M.), Uppsala University, Sweden
| | - Rui Zeng
- Department of Medical Sciences (L.L., R.Z., S.G., J.S.), Uppsala University, Sweden
| | - Daniela Zanetti
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (T.A., D.Z.)
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences/Geriatrics (M.I.), Uppsala University, Sweden
| | - Stefan Gustafsson
- Department of Medical Sciences (L.L., R.Z., S.G., J.S.), Uppsala University, Sweden
| | - Johan Sundström
- Department of Medical Sciences (L.L., R.Z., S.G., J.S.), Uppsala University, Sweden
| | - Johan Ärnlöv
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge (J.A.)
| | - Sölve Elmståhl
- Department of Clinical Sciences in Malmö, Lund University, Sweden (S.E.)
| | - Themistocles Assimes
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (T.A., D.Z.)
- Palo Alto VA Healthcare System, CA (T.A.)
| | - Karl Michaëlsson
- Department of Surgical Sciences (O.T., K.M.), Uppsala University, Sweden
| |
Collapse
|
28
|
de Bakker M, Petersen TB, Rueten-Budde AJ, Akkerhuis KM, Umans VA, Brugts JJ, Germans T, Reinders MJT, Katsikis PD, van der Spek PJ, Ostroff R, She R, Lanfear D, Asselbergs FW, Boersma E, Rizopoulos D, Kardys I. Machine learning-based biomarker profile derived from 4210 serially measured proteins predicts clinical outcome of patients with heart failure. EUROPEAN HEART JOURNAL. DIGITAL HEALTH 2023; 4:444-454. [PMID: 38045440 PMCID: PMC10689916 DOI: 10.1093/ehjdh/ztad056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/06/2023] [Accepted: 10/03/2023] [Indexed: 12/05/2023]
Abstract
Aims Risk assessment tools are needed for timely identification of patients with heart failure (HF) with reduced ejection fraction (HFrEF) who are at high risk of adverse events. In this study, we aim to derive a small set out of 4210 repeatedly measured proteins, which, along with clinical characteristics and established biomarkers, carry optimal prognostic capacity for adverse events, in patients with HFrEF. Methods and results In 382 patients, we performed repeated blood sampling (median follow-up: 2.1 years) and applied an aptamer-based multiplex proteomic approach. We used machine learning to select the optimal set of predictors for the primary endpoint (PEP: composite of cardiovascular death, heart transplantation, left ventricular assist device implantation, and HF hospitalization). The association between repeated measures of selected proteins and PEP was investigated by multivariable joint models. Internal validation (cross-validated c-index) and external validation (Henry Ford HF PharmacoGenomic Registry cohort) were performed. Nine proteins were selected in addition to the MAGGIC risk score, N-terminal pro-hormone B-type natriuretic peptide, and troponin T: suppression of tumourigenicity 2, tryptophanyl-tRNA synthetase cytoplasmic, histone H2A Type 3, angiotensinogen, deltex-1, thrombospondin-4, ADAMTS-like protein 2, anthrax toxin receptor 1, and cathepsin D. N-terminal pro-hormone B-type natriuretic peptide and angiotensinogen showed the strongest associations [hazard ratio (95% confidence interval): 1.96 (1.17-3.40) and 0.66 (0.49-0.88), respectively]. The multivariable model yielded a c-index of 0.85 upon internal validation and c-indices up to 0.80 upon external validation. The c-index was higher than that of a model containing established risk factors (P = 0.021). Conclusion Nine serially measured proteins captured the most essential prognostic information for the occurrence of adverse events in patients with HFrEF, and provided incremental value for HF prognostication beyond established risk factors. These proteins could be used for dynamic, individual risk assessment in a prospective setting. These findings also illustrate the potential value of relatively 'novel' biomarkers for prognostication. Clinical Trial Registration https://clinicaltrials.gov/ct2/show/NCT01851538?term=nCT01851538&draw=2&rank=1 24.
Collapse
Affiliation(s)
- Marie de Bakker
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Teun B Petersen
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Anja J Rueten-Budde
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Victor A Umans
- Department of Cardiology, Northwest Clinics, Wilhelminalaan 12, 1815 JD, Alkmaar, The Netherlands
| | - Jasper J Brugts
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Northwest Clinics, Wilhelminalaan 12, 1815 JD, Alkmaar, The Netherlands
| | - Marcel J T Reinders
- Delft Bioinformatics Lab, Delft University of Technology, Van Mourik Broekmanweg 6, 2628 XE, Delft, The Netherlands
| | - Peter D Katsikis
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Peter J van der Spek
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Rachel Ostroff
- SomaLogic, Inc., 2945 Wilderness Pl., Boulder, CO 80301, USA
| | - Ruicong She
- Department of Public Health Sciences, Henry Ford Health System, 1 Ford Pl, Detroit, MI 48202, USA
| | - David Lanfear
- Center for Individualized and Genomic Medicine Research (CIGMA), Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit MI, 48202, USA
- Heart and Vascular Institute, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | - Folkert W Asselbergs
- Amsterdam University Medical Centers, Department of Cardiology, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, Gower St, London, WC1E 6BT, UK
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Dimitris Rizopoulos
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Dr. Molenwaterplein 40, 3015GD, Rotterdam, The Netherlands
| |
Collapse
|
29
|
Chen SF, Loguercio S, Chen KY, Lee SE, Park JB, Liu S, Sadaei HJ, Torkamani A. Artificial Intelligence for Risk Assessment on Primary Prevention of Coronary Artery Disease. CURRENT CARDIOVASCULAR RISK REPORTS 2023; 17:215-231. [DOI: 10.1007/s12170-023-00731-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 01/04/2025]
Abstract
Abstract
Purpose of Review
Coronary artery disease (CAD) is a common and etiologically complex disease worldwide. Current guidelines for primary prevention, or the prevention of a first acute event, include relatively simple risk assessment and leave substantial room for improvement both for risk ascertainment and selection of prevention strategies. Here, we review how advances in big data and predictive modeling foreshadow a promising future of improved risk assessment and precision medicine for CAD.
Recent Findings
Artificial intelligence (AI) has improved the utility of high dimensional data, providing an opportunity to better understand the interplay between numerous CAD risk factors. Beyond applications of AI in cardiac imaging, the vanguard application of AI in healthcare, recent translational research is also revealing a promising path for AI in multi-modal risk prediction using standard biomarkers, genetic and other omics technologies, a variety of biosensors, and unstructured data from electronic health records (EHRs). However, gaps remain in clinical validation of AI models, most notably in the actionability of complex risk prediction for more precise therapeutic interventions.
Summary
The recent availability of nation-scale biobank datasets has provided a tremendous opportunity to richly characterize longitudinal health trajectories using health data collected at home, at laboratories, and through clinic visits. The ever-growing availability of deep genotype-phenotype data is poised to drive a transition from simple risk prediction algorithms to complex, “data-hungry,” AI models in clinical decision-making. While AI models provide the means to incorporate essentially all risk factors into comprehensive risk prediction frameworks, there remains a need to wrap these predictions in interpretable frameworks that map to our understanding of underlying biological mechanisms and associated personalized intervention. This review explores recent advances in the role of machine learning and AI in CAD primary prevention and highlights current strengths as well as limitations mediating potential future applications.
Collapse
|
30
|
Mahoney SA, Dey AK, Basisty N, Herman AB. Identification and functional analysis of senescent cells in the cardiovascular system using omics approaches. Am J Physiol Heart Circ Physiol 2023; 325:H1039-H1058. [PMID: 37656130 PMCID: PMC10908411 DOI: 10.1152/ajpheart.00352.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide, and senescent cells have emerged as key contributors to its pathogenesis. Senescent cells exhibit cell cycle arrest and secrete a range of proinflammatory factors, termed the senescence-associated secretory phenotype (SASP), which promotes tissue dysfunction and exacerbates CVD progression. Omics technologies, specifically transcriptomics and proteomics, offer powerful tools to uncover and define the molecular signatures of senescent cells in cardiovascular tissue. By analyzing the comprehensive molecular profiles of senescent cells, omics approaches can identify specific genetic alterations, gene expression patterns, protein abundances, and metabolite levels associated with senescence in CVD. These omics-based discoveries provide insights into the mechanisms underlying senescence-induced cardiovascular damage, facilitating the development of novel diagnostic biomarkers and therapeutic targets. Furthermore, integration of multiple omics data sets enables a systems-level understanding of senescence in CVD, paving the way for precision medicine approaches to prevent or treat cardiovascular aging and its associated complications.
Collapse
Affiliation(s)
- Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, Colorado, United States
| | - Amit K Dey
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| | - Nathan Basisty
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| | - Allison B Herman
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| |
Collapse
|
31
|
Sattar N, Taheri S, Astling DP, Chadwick J, Hinterberg MA, Holmes MV, Troth EV, Welsh P, Zaghloul H, Chagoury O, Lean M, Taylor R, Williams S. Prediction of Cardiometabolic Health Through Changes in Plasma Proteins With Intentional Weight Loss in the DiRECT and DIADEM-I Randomized Clinical Trials of Type 2 Diabetes Remission. Diabetes Care 2023; 46:1949-1957. [PMID: 37756566 PMCID: PMC10628468 DOI: 10.2337/dc23-0602] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/04/2023] [Indexed: 09/29/2023]
Abstract
OBJECTIVE To determine the extent to which changes in plasma proteins, previously predictive of cardiometabolic outcomes, predict changes in two diabetes remission trials. RESEARCH DESIGN AND METHODS We applied SomaSignal predictive tests (each derived from ∼5,000 plasma protein measurements using aptamer-based proteomics assay) to baseline and 1-year samples of trial intervention (Diabetes Remission Clinical Trial [DiRECT], n = 118, and Diabetes Intervention Accentuating Diet and Enhancing Metabolism [DIADEM-I], n = 66) and control (DiRECT, n = 144, DIADEM-I, n = 76) group participants. RESULTS Mean (SD) weight loss in DiRECT (U.K.) and DIADEM-I (Qatar) was 10.2 (7.4) kg and 12.1 (9.5) kg, respectively, vs. 1.0 (3.7) kg and 4.0 (5.4) kg in control groups. Cardiometabolic SomaSignal test results showed significant improvement (Bonferroni-adjusted P < 0.05) in DiRECT and DIADEM-I (expressed as relative difference, intervention minus control) as follows, respectively: liver fat (-26.4%, -37.3%), glucose tolerance (-36.6%, -37.4%), body fat percentage (-8.6%, -8.7%), resting energy rate (-8.0%, -5.1%), visceral fat (-34.3%, -26.1%), and cardiorespiratory fitness (9.5%, 10.3%). Cardiovascular risk (measured with SomaSignal tests) also improved in intervention groups relative to control, but this was significant only in DiRECT (DiRECT, -44.2%, and DIADEM-I, -9.2%). However, weight loss >10 kg predicted significant reductions in cardiovascular risk, -19.1% (95% CI -33.4 to -4.91) in DiRECT and -33.4% (95% CI -57.3, -9.6) in DIADEM-I. DIADEM-I also demonstrated rapid emergence of metabolic improvements at 3 months. CONCLUSIONS Intentional weight loss in recent-onset type 2 diabetes rapidly induces changes in protein-based risk models consistent with widespread cardiometabolic improvements, including cardiorespiratory fitness. Protein changes with greater (>10 kg) weight loss also predicted lower cardiovascular risk, providing a positive outlook for relevant ongoing trials.
Collapse
Affiliation(s)
- Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Shahrad Taheri
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
- Weill Cornell Medicine-Qatar, Doha, Qatar
- Weill Cornell Medicine, New York, NY
| | | | | | | | - Michael V. Holmes
- Medical Research Council, Integrative Epidemiology Unit, University of Bristol, Bristol, U.K
| | | | - Paul Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Hadeel Zaghloul
- Weill Cornell Medicine-Qatar, Doha, Qatar
- Weill Cornell Medicine, New York, NY
| | - Odette Chagoury
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
- Weill Cornell Medicine-Qatar, Doha, Qatar
- Weill Cornell Medicine, New York, NY
| | - Mike Lean
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, U.K
| | | |
Collapse
|
32
|
Dubin RF, Deo R, Ren Y, Wang J, Zheng Z, Shou H, Go AS, Parsa A, Lash JP, Rahman M, Hsu CY, Weir MR, Chen J, Anderson A, Grams ME, Surapaneni A, Coresh J, Li H, Kimmel PL, Vasan RS, Feldman H, Segal MR, Ganz P. Proteomics of CKD progression in the chronic renal insufficiency cohort. Nat Commun 2023; 14:6340. [PMID: 37816758 PMCID: PMC10564759 DOI: 10.1038/s41467-023-41642-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
Progression of chronic kidney disease (CKD) portends myriad complications, including kidney failure. In this study, we analyze associations of 4638 plasma proteins among 3235 participants of the Chronic Renal Insufficiency Cohort Study with the primary outcome of 50% decline in estimated glomerular filtration rate or kidney failure over 10 years. We validate key findings in the Atherosclerosis Risk in the Communities study. We identify 100 circulating proteins that are associated with the primary outcome after multivariable adjustment, using a Bonferroni statistical threshold of significance. Individual protein associations and biological pathway analyses highlight the roles of bone morphogenetic proteins, ephrin signaling, and prothrombin activation. A 65-protein risk model for the primary outcome has excellent discrimination (C-statistic[95%CI] 0.862 [0.835, 0.889]), and 14/65 proteins are druggable targets. Potentially causal associations for five proteins, to our knowledge not previously reported, are supported by Mendelian randomization: EGFL9, LRP-11, MXRA7, IL-1 sRII and ILT-2. Modifiable protein risk markers can guide therapeutic drug development aimed at slowing CKD progression.
Collapse
Affiliation(s)
- Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianqiao Wang
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alan S Go
- Division of Research, Kaiser Permanente Northern California, Oakland, the Department of Health Systems Science, Oakland, CA, USA
| | - Afshin Parsa
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - James P Lash
- Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Mahboob Rahman
- Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Chi-Yuan Hsu
- Division of Research, Kaiser Permanente Northern California, Oakland, the Department of Health Systems Science, Oakland, CA, USA
- Division of Nephrology, University of California San Francisco, San Francisco, CA, USA
| | - Matthew R Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jing Chen
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Amanda Anderson
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Morgan E Grams
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Aditya Surapaneni
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Josef Coresh
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hongzhe Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ramachandran S Vasan
- University of Texas School of Public Health San Antonio and the University of Texas Health Sciences Center in San Antonio. Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Harold Feldman
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Peter Ganz
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
33
|
Helgason H, Eiriksdottir T, Ulfarsson MO, Choudhary A, Lund SH, Ivarsdottir EV, Hjorleifsson Eldjarn G, Einarsson G, Ferkingstad E, Moore KHS, Honarpour N, Liu T, Wang H, Hucko T, Sabatine MS, Morrow DA, Giugliano RP, Ostrowski SR, Pedersen OB, Bundgaard H, Erikstrup C, Arnar DO, Thorgeirsson G, Masson G, Magnusson OT, Saemundsdottir J, Gretarsdottir S, Steinthorsdottir V, Thorleifsson G, Helgadottir A, Sulem P, Thorsteinsdottir U, Holm H, Gudbjartsson D, Stefansson K. Evaluation of Large-Scale Proteomics for Prediction of Cardiovascular Events. JAMA 2023; 330:725-735. [PMID: 37606673 PMCID: PMC10445198 DOI: 10.1001/jama.2023.13258] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 06/29/2023] [Indexed: 08/23/2023]
Abstract
Importance Whether protein risk scores derived from a single plasma sample could be useful for risk assessment for atherosclerotic cardiovascular disease (ASCVD), in conjunction with clinical risk factors and polygenic risk scores, is uncertain. Objective To develop protein risk scores for ASCVD risk prediction and compare them to clinical risk factors and polygenic risk scores in primary and secondary event populations. Design, Setting, and Participants The primary analysis was a retrospective study of primary events among 13 540 individuals in Iceland (aged 40-75 years) with proteomics data and no history of major ASCVD events at recruitment (study duration, August 23, 2000 until October 26, 2006; follow-up through 2018). We also analyzed a secondary event population from a randomized, double-blind lipid-lowering clinical trial (2013-2016), consisting of individuals with stable ASCVD receiving statin therapy and for whom proteomic data were available for 6791 individuals. Exposures Protein risk scores (based on 4963 plasma protein levels and developed in a training set in the primary event population); polygenic risk scores for coronary artery disease and stroke; and clinical risk factors that included age, sex, statin use, hypertension treatment, type 2 diabetes, body mass index, and smoking status at the time of plasma sampling. Main Outcomes and Measures Outcomes were composites of myocardial infarction, stroke, and coronary heart disease death or cardiovascular death. Performance was evaluated using Cox survival models and measures of discrimination and reclassification that accounted for the competing risk of non-ASCVD death. Results In the primary event population test set (4018 individuals [59.0% women]; 465 events; median follow-up, 15.8 years), the protein risk score had a hazard ratio (HR) of 1.93 per SD (95% CI, 1.75 to 2.13). Addition of protein risk score and polygenic risk scores significantly increased the C index when added to a clinical risk factor model (C index change, 0.022 [95% CI, 0.007 to 0.038]). Addition of the protein risk score alone to a clinical risk factor model also led to a significantly increased C index (difference, 0.014 [95% CI, 0.002 to 0.028]). Among White individuals in the secondary event population (6307 participants; 432 events; median follow-up, 2.2 years), the protein risk score had an HR of 1.62 per SD (95% CI, 1.48 to 1.79) and significantly increased C index when added to a clinical risk factor model (C index change, 0.026 [95% CI, 0.011 to 0.042]). The protein risk score was significantly associated with major adverse cardiovascular events among individuals of African and Asian ancestries in the secondary event population. Conclusions and Relevance A protein risk score was significantly associated with ASCVD events in primary and secondary event populations. When added to clinical risk factors, the protein risk score and polygenic risk score both provided statistically significant but modest improvement in discrimination.
Collapse
Affiliation(s)
- Hannes Helgason
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- University of Iceland, Reykjavik, Iceland
| | | | - Magnus O. Ulfarsson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- University of Iceland, Reykjavik, Iceland
| | | | | | | | | | | | | | | | | | | | - Huei Wang
- Amgen, Inc, Thousand Oaks, California
| | | | - Marc S. Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - David A. Morrow
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Robert P. Giugliano
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole Birger Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Henning Bundgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, The Heart Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David O. Arnar
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- University of Iceland, Reykjavik, Iceland
- Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- University of Iceland, Reykjavik, Iceland
- Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland
| | | | | | | | | | | | | | | | | | | | - Hilma Holm
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
| | - Daniel Gudbjartsson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- University of Iceland, Reykjavik, Iceland
| |
Collapse
|
34
|
Sasamoto N, Ngo L, Vitonis AF, Dillon ST, Sieberg CB, Missmer SA, Libermann TA, Terry KL. Plasma proteomic profiles of pain subtypes in adolescents and young adults with endometriosis. Hum Reprod 2023; 38:1509-1519. [PMID: 37196326 PMCID: PMC10391309 DOI: 10.1093/humrep/dead099] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/12/2023] [Indexed: 05/19/2023] Open
Abstract
STUDY QUESTION What are the similarities and differences in the systemic proteomic profiles by endometriosis-associated pain subtypes among adolescents and young adults with endometriosis? SUMMARY ANSWER Endometriosis-associated pain subtypes exhibited distinct plasma proteomic profiles. WHAT IS KNOWN ALREADY Endometriosis patients, especially those diagnosed in adolescents and young adults, are often plagued by various pain symptoms. However, it is not clear what biological processes underlie this heterogeneity. STUDY DESIGN, SIZE, DURATION We conducted a cross-sectional analysis using data and plasma samples from 142 adolescent or young adult participants of the Women's Health Study: From Adolescence to Adulthood cohort with laparoscopically confirmed endometriosis. PARTICIPANTS/MATERIALS, SETTING, METHODS We measured 1305 plasma protein levels by SomaScan. We classified self-reported endometriosis-associated pain into subtypes of dysmenorrhea, acyclic pelvic pain, life impacting pelvic pain, bladder pain, bowel pain, and widespread pain phenotype. We used logistic regression to calculate the odds ratios and 95% confidence intervals for differentially expressed proteins, adjusting for age, BMI, fasting status, and hormone use at blood draw. Ingenuity Pathway Analysis identified enriched biological pathways. MAIN RESULTS AND THE ROLE OF CHANCE Our study population consisted mainly of adolescents and young adults (mean age at blood draw = 18 years), with nearly all (97%) scored as rASRM stage I/II at laparoscopic diagnosis of endometriosis, which is a common clinical presentation of endometriosis diagnosed at a younger age. Pain subtypes exhibited distinct plasma proteomic profiles. Multiple cell movement pathways were downregulated in cases with severe dysmenorrhea and life impacting pelvic pain compared to those without (P < 7.5×10-15). Endometriosis cases with acyclic pelvic pain had upregulation of immune cell adhesion pathways (P < 9.0×10-9), while those with bladder pain had upregulation of immune cell migration (P < 3.7×10-8) and those with bowel pain had downregulation (P < 6.5×10-7) of the immune cell migration pathways compared to those without. Having a wide-spread pain phenotype involved downregulation of multiple immune pathways (P < 8.0×10-10). LIMITATIONS, REASONS FOR CAUTION Our study was limited by the lack of an independent validation cohort. We were also only able to explore any presence of a pain subtype and could not evaluate multiple combinations by pain subtypes. Further mechanistic studies are warranted to elucidate the differences in pathophysiology by endometriosis-pain subtype. WIDER IMPLICATIONS OF THE FINDINGS The observed variation in plasma protein profiles by pain subtypes suggests different underlying molecular mechanisms, highlighting the need for potential consideration of pain subtypes for effectively treating endometriosis patients presenting with various pain symptoms. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the Department of Defense W81XWH1910318 and the 2017 Boston Center for Endometriosis Trainee Award. Financial support for establishment of and data collection within the A2A cohort were provided by the J. Willard and Alice S. Marriott Foundation. N.S., A.F.V., S.A.M., and K.L.T. have received funding from the Marriott Family Foundation. C.B.S. is funded by an R35 MIRA Award from NIGMS (5R35GM142676). S.A.M. and K.L.T. are supported by NICHD R01HD094842. S.A.M. reports serving as an advisory board member for AbbVie and Roche, Field Chief Editor for Frontiers in Reproductive Health, personal fees from Abbott for roundtable participation; none of these are related to this study. Other authors report no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Naoko Sasamoto
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
| | - Long Ngo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Allison F Vitonis
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
| | - Simon T Dillon
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Christine B Sieberg
- Biobehavioral Pain Innovations Lab, Department of Psychiatry & Behavioral Sciences, Boston Children’s Hospital, Boston, MA, USA
- Pain & Affective Neuroscience Center, Department of Anesthesiology, Critical Care, & Pain Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Stacey A Missmer
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Towia A Libermann
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kathryn L Terry
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
35
|
De Belen E, Ganesan D, Paculdo D, Gill R, Peabody JW. Clinical Variation in the Treatment Practices for Patients With Type 2 Diabetes: A Cross-Sectional Patient Simulation Study Among Primary Care Physicians and Cardiologists. J Am Heart Assoc 2023; 12:e028634. [PMID: 37382120 PMCID: PMC10356086 DOI: 10.1161/jaha.122.028634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/07/2023] [Indexed: 06/30/2023]
Abstract
Background Cardiovascular disease risk stratification is necessary and critically important in patients with type 2 diabetes. Despite its known benefits to guide treatment and prevention, we hypothesized that providers do not routinely incorporate this into their diagnostic and treatment decisions. Methods and Results The QuiCER DM (QURE CVD Evaluation of Risk in Diabetes Mellitus) study enrolled 161 primary care physicians and 80 cardiologists. Between March 2022 and June 2022, we measured the care variation in risk determination among these providers caring for simulated patients with type 2 diabetes. We found a wide variation in the overall assessment of cardiovascular disease in patients with type 2 diabetes. Participants performed half of the necessary care items with quality-of-care scores, ranging between 13% and 84%, averaging 49.4±12.6%. Participants did not assess cardiovascular risk in 18.3% of cases and incorrectly stratified risk in 42.8% of cases. Only 38.9% of participants arrived at the correct cardiovascular risk stratification. Those who correctly identified a cardiovascular risk score were significantly more likely to order nonpharmacologic treatments, advising on their patients' nutrition (38.8% versus 29.9%, P=0.013) and the correct glycated hemoglobin target (37.7% versus 15.6%, P<0.001). Pharmacologic treatments, however, did not vary between those who correctly specified risk and those who did not. Conclusions Physician participants struggled to determine the correct cardiovascular disease risk and specify the appropriate pharmacologic interventions in simulated patients with type 2 diabetes. Additionally, there was a wide variation in the quality of care regardless of risk level, indicating opportunities to improve risk stratification.
Collapse
Affiliation(s)
| | | | | | | | - John W. Peabody
- QURE HealthcareSan FranciscoCAUSA
- University of CaliforniaSan FranciscoCAUSA
- University of CaliforniaLos AngelesCAUSA
| |
Collapse
|
36
|
Kivimäki M, Livingston G, Singh-Manoux A, Mars N, Lindbohm JV, Pentti J, Nyberg ST, Pirinen M, Anderson EL, Hingorani AD, Sipilä PN. Estimating Dementia Risk Using Multifactorial Prediction Models. JAMA Netw Open 2023; 6:e2318132. [PMID: 37310738 PMCID: PMC10265307 DOI: 10.1001/jamanetworkopen.2023.18132] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/27/2023] [Indexed: 06/14/2023] Open
Abstract
Importance The clinical value of current multifactorial algorithms for individualized assessment of dementia risk remains unclear. Objective To evaluate the clinical value associated with 4 widely used dementia risk scores in estimating 10-year dementia risk. Design, Setting, and Participants This prospective population-based UK Biobank cohort study assessed 4 dementia risk scores at baseline (2006-2010) and ascertained incident dementia during the following 10 years. Replication with a 20-year follow-up was based on the British Whitehall II study. For both analyses, participants who had no dementia at baseline, had complete data on at least 1 dementia risk score, and were linked to electronic health records from hospitalizations or mortality were included. Data analysis was conducted from July 5, 2022, to April 20, 2023. Exposures Four existing dementia risk scores: the Cardiovascular Risk Factors, Aging and Dementia (CAIDE)-Clinical score, the CAIDE-APOE-supplemented score, the Brief Dementia Screening Indicator (BDSI), and the Australian National University Alzheimer Disease Risk Index (ANU-ADRI). Main Outcomes and Measures Dementia was ascertained from linked electronic health records. To evaluate how well each score predicted the 10-year risk of dementia, concordance (C) statistics, detection rate, false-positive rate, and the ratio of true to false positives were calculated for each risk score and for a model including age alone. Results Of 465 929 UK Biobank participants without dementia at baseline (mean [SD] age, 56.5 [8.1] years; range, 38-73 years; 252 778 [54.3%] female participants), 3421 were diagnosed with dementia at follow-up (7.5 per 10 000 person-years). If the threshold for a positive test result was calibrated to achieve a 5% false-positive rate, all 4 risk scores detected 9% to 16% of incident dementia and therefore missed 84% to 91% (failure rate). The corresponding failure rate was 84% for a model that included age only. For a positive test result calibrated to detect at least half of future incident dementia, the ratio of true to false positives ranged between 1 to 66 (for CAIDE-APOE-supplemented) and 1 to 116 (for ANU-ADRI). For age alone, the ratio was 1 to 43. The C statistic was 0.66 (95% CI, 0.65-0.67) for the CAIDE clinical version, 0.73 (95% CI, 0.72-0.73) for the CAIDE-APOE-supplemented, 0.68 (95% CI, 0.67-0.69) for BDSI, 0.59 (95% CI, 0.58-0.60) for ANU-ADRI, and 0.79 (95% CI, 0.79-0.80) for age alone. Similar C statistics were seen for 20-year dementia risk in the Whitehall II study cohort, which included 4865 participants (mean [SD] age, 54.9 [5.9] years; 1342 [27.6%] female participants). In a subgroup analysis of same-aged participants aged 65 (±1) years, discriminatory capacity of risk scores was low (C statistics between 0.52 and 0.60). Conclusions and Relevance In these cohort studies, individualized assessments of dementia risk using existing risk prediction scores had high error rates. These findings suggest that the scores were of limited value in targeting people for dementia prevention. Further research is needed to develop more accurate algorithms for estimation of dementia risk.
Collapse
Affiliation(s)
- Mika Kivimäki
- Department of Mental Health of Older People, UCL Brain Sciences, University College London, London, United Kingdom
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Gill Livingston
- Department of Mental Health of Older People, UCL Brain Sciences, University College London, London, United Kingdom
| | - Archana Singh-Manoux
- Department of Mental Health of Older People, UCL Brain Sciences, University College London, London, United Kingdom
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Nina Mars
- Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Joni V. Lindbohm
- Department of Mental Health of Older People, UCL Brain Sciences, University College London, London, United Kingdom
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Jaana Pentti
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Solja T. Nyberg
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matti Pirinen
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| | - Emma L. Anderson
- Department of Mental Health of Older People, UCL Brain Sciences, University College London, London, United Kingdom
- MRC Integrative Epidemiology Unit and Population Health Sciences, University of Bristol Medical School, Bristol, United Kingdom
| | - Aroon D. Hingorani
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom
| | - Pyry N. Sipilä
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
37
|
Nurmohamed NS, Kraaijenhof JM, Mayr M, Nicholls SJ, Koenig W, Catapano AL, Stroes ESG. Proteomics and lipidomics in atherosclerotic cardiovascular disease risk prediction. Eur Heart J 2023; 44:1594-1607. [PMID: 36988179 PMCID: PMC10163980 DOI: 10.1093/eurheartj/ehad161] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/04/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Given the limited accuracy of clinically used risk scores such as the Systematic COronary Risk Evaluation 2 system and the Second Manifestations of ARTerial disease 2 risk scores, novel risk algorithms determining an individual's susceptibility of future incident or recurrent atherosclerotic cardiovascular disease (ASCVD) risk are urgently needed. Due to major improvements in assay techniques, multimarker proteomic and lipidomic panels hold the promise to be reliably assessed in a high-throughput routine. Novel machine learning-based approaches have facilitated the use of this high-dimensional data resulting from these analyses for ASCVD risk prediction. More than a dozen of large-scale retrospective studies using different sets of biomarkers and different statistical methods have consistently demonstrated the additive prognostic value of these panels over traditionally used clinical risk scores. Prospective studies are needed to determine the clinical utility of a biomarker panel in clinical ASCVD risk stratification. When combined with the genetic predisposition captured with polygenic risk scores and the actual ASCVD phenotype observed with coronary artery imaging, proteomics and lipidomics can advance understanding of the complex multifactorial causes underlying an individual's ASCVD risk.
Collapse
Affiliation(s)
- Nick S Nurmohamed
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Jordan M Kraaijenhof
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Manuel Mayr
- School of Cardiovascular and Metabolic Medicine & Science, King’s College London, Strand, London WC2R 2LS, UK
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Währinger Gürtel, 18-201090 Vienna, Austria
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, VIC 3168, Australia
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Lazarettstraße 36, 80636 München, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Pettenkoferstr. 8a & 9, 80336 Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Helmholtzstr. 22, 89081 Ulm, Germany
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
- IRCCS Multimedica, Via Milanese, 300, 20099 Sesto San Giovanni (MI), Italy
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
38
|
Sun W, Lin Y, Huang Y, Chan J, Terrillon S, Rosenbaum AI, Contrepois K. Robust and High-Throughput Analytical Flow Proteomics Analysis of Cynomolgus Monkey and Human Matrices with Zeno SWATH Data Independent Acquisition. Mol Cell Proteomics 2023:100562. [PMID: 37142056 DOI: 10.1016/j.mcpro.2023.100562] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/17/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023] Open
Abstract
Modern mass spectrometers routinely allow deep proteome coverage in a single experiment. These methods are typically operated at nano and micro flow regimes, but they often lack throughput and chromatographic robustness, which is critical for large-scale studies. In this context, we have developed, optimized and benchmarked LC-MS methods combining the robustness and throughput of analytical flow chromatography with the added sensitivity provided by the Zeno trap across a wide range of cynomolgus monkey and human matrices of interest for toxicological studies and clinical biomarker discovery. SWATH data independent acquisition (DIA) experiments with Zeno trap activated (Zeno SWATH DIA) provided a clear advantage over conventional SWATH DIA in all sample types tested with improved sensitivity, quantitative robustness and signal linearity as well as increased protein coverage by up to 9-fold. Using a 10-min gradient chromatography, up to 3,300 proteins were identified in tissues at 2 μg peptide load. Importantly, the performance gains with Zeno SWATH translated into better biological pathway representation and improved the ability to identify dysregulated proteins and pathways associated with two metabolic diseases in human plasma. Finally, we demonstrate that this method is highly stable over time with the acquisition of reliable data over the injection of 1,000+ samples (14.2 days of uninterrupted acquisition) without the need for human intervention or normalization. Altogether, Zeno SWATH DIA methodology allows fast, sensitive and robust proteomic workflows using analytical flow and is amenable to large-scale studies. This work provides detailed method performance assessment on a variety of relevant biological matrices and serves as a valuable resource for the proteomics community.
Collapse
Affiliation(s)
- Weiwen Sun
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA
| | - Yuan Lin
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA
| | - Yue Huang
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA
| | - Josolyn Chan
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA
| | - Sonia Terrillon
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA
| | - Anton I Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA.
| | - Kévin Contrepois
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA.
| |
Collapse
|
39
|
Chahine Y, Magoon MJ, Maidu B, del Álamo JC, Boyle PM, Akoum N. Machine Learning and the Conundrum of Stroke Risk Prediction. Arrhythm Electrophysiol Rev 2023; 12:e07. [PMID: 37427297 PMCID: PMC10326666 DOI: 10.15420/aer.2022.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/07/2023] [Indexed: 07/11/2023] Open
Abstract
Stroke is a leading cause of death worldwide. With escalating healthcare costs, early non-invasive stroke risk stratification is vital. The current paradigm of stroke risk assessment and mitigation is focused on clinical risk factors and comorbidities. Standard algorithms predict risk using regression-based statistical associations, which, while useful and easy to use, have moderate predictive accuracy. This review summarises recent efforts to deploy machine learning (ML) to predict stroke risk and enrich the understanding of the mechanisms underlying stroke. The surveyed body of literature includes studies comparing ML algorithms with conventional statistical models for predicting cardiovascular disease and, in particular, different stroke subtypes. Another avenue of research explored is ML as a means of enriching multiscale computational modelling, which holds great promise for revealing thrombogenesis mechanisms. Overall, ML offers a new approach to stroke risk stratification that accounts for subtle physiologic variants between patients, potentially leading to more reliable and personalised predictions than standard regression-based statistical associations.
Collapse
Affiliation(s)
- Yaacoub Chahine
- Division of Cardiology, University of Washington, Seattle, WA, US
| | - Matthew J Magoon
- Department of Bioengineering, University of Washington, Seattle, WA, US
| | - Bahetihazi Maidu
- Department of Mechanical Engineering, University of Washington, Seattle, WA, US
| | - Juan C del Álamo
- Department of Mechanical Engineering, University of Washington, Seattle, WA, US
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, US
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, US
| | - Patrick M Boyle
- Department of Bioengineering, University of Washington, Seattle, WA, US
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, US
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, US
| | - Nazem Akoum
- Division of Cardiology, University of Washington, Seattle, WA, US
- Department of Bioengineering, University of Washington, Seattle, WA, US
| |
Collapse
|
40
|
Gomez-Lopez N, Romero R, Escobar MF, Carvajal JA, Echavarria MP, Albornoz LL, Nasner D, Miller D, Gallo DM, Galaz J, Arenas-Hernandez M, Bhatti G, Done B, Zambrano MA, Ramos I, Fernandez PA, Posada L, Chaiworapongsa T, Jung E, Garcia-Flores V, Suksai M, Gotsch F, Bosco M, Than NG, Tarca AL. Pregnancy-specific responses to COVID-19 revealed by high-throughput proteomics of human plasma. COMMUNICATIONS MEDICINE 2023; 3:48. [PMID: 37016066 PMCID: PMC10071476 DOI: 10.1038/s43856-023-00268-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 03/03/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Pregnant women are at greater risk of adverse outcomes, including mortality, as well as obstetrical complications resulting from COVID-19. However, pregnancy-specific changes that underlie such worsened outcomes remain unclear. METHODS Plasma samples were collected from pregnant women and non-pregnant individuals (male and female) with (n = 72 pregnant, 52 non-pregnant) and without (n = 29 pregnant, 41 non-pregnant) COVID-19. COVID-19 patients were grouped as asymptomatic, mild, moderate, severe, or critically ill according to NIH classifications. Proteomic profiling of 7,288 analytes corresponding to 6,596 unique protein targets was performed using the SOMAmer platform. RESULTS Herein, we profile the plasma proteome of pregnant and non-pregnant COVID-19 patients and controls and show alterations that display a dose-response relationship with disease severity; yet, such proteomic perturbations are dampened during pregnancy. In both pregnant and non-pregnant state, the proteome response induced by COVID-19 shows enrichment of mediators implicated in cytokine storm, endothelial dysfunction, and angiogenesis. Shared and pregnancy-specific proteomic changes are identified: pregnant women display a tailored response that may protect the conceptus from heightened inflammation, while non-pregnant individuals display a stronger response to repel infection. Furthermore, the plasma proteome can accurately identify COVID-19 patients, even when asymptomatic or with mild symptoms. CONCLUSION This study represents the most comprehensive characterization of the plasma proteome of pregnant and non-pregnant COVID-19 patients. Our findings emphasize the distinct immune modulation between the non-pregnant and pregnant states, providing insight into the pathogenesis of COVID-19 as well as a potential explanation for the more severe outcomes observed in pregnant women.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.
- Detroit Medical Center, Detroit, MI, USA.
| | - María Fernanda Escobar
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Javier Andres Carvajal
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Maria Paula Echavarria
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Ludwig L Albornoz
- Departamento de Laboratorio Clínico y Patología, Fundación Valle del Lili, Cali, Colombia
- Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Daniela Nasner
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, Cali, Colombia
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dahiana M Gallo
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gaurav Bhatti
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Done
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maria Andrea Zambrano
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Isabella Ramos
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Paula Andrea Fernandez
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Leandro Posada
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Tinnakorn Chaiworapongsa
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Francesca Gotsch
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariachiara Bosco
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nandor Gabor Than
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
- Genesis Theranostix Group, Budapest, Hungary
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA.
| |
Collapse
|
41
|
Blatt S, Kämmerer PW, Krüger M, Surabattula R, Thiem DGE, Dillon ST, Al-Nawas B, Libermann TA, Schuppan D. High-Multiplex Aptamer-Based Serum Proteomics to Identify Candidate Serum Biomarkers of Oral Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15072071. [PMID: 37046731 PMCID: PMC10093013 DOI: 10.3390/cancers15072071] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Improved serological biomarkers are needed for the early detection, risk stratification and treatment surveillance of patients with oral squamous cell carcinoma (OSCC). We performed an exploratory study using advanced, highly specific, DNA-aptamer-based serum proteomics (SOMAscan, 1305-plex) to identify distinct proteomic changes in patients with OSCC pre- vs. post-resection and compared to healthy controls. A total of 63 significantly differentially expressed serum proteins (each p < 0.05) were found that could discriminate between OSCC and healthy controls with 100% accuracy. Furthermore, 121 proteins were detected that were significantly altered between pre- and post-resection sera, and 12 OSCC-associated proteins reversed to levels equivalent to healthy controls after resection. Of these, 6 were increased and 6 were decreased relative to healthy controls, highlighting the potential relevance of these proteins as OSCC tumor markers. Pathway analyses revealed potential pathophysiological mechanisms associated with OSCC. Hence, quantitative proteome analysis using SOMAscan technology is promising and may aid in the development of defined serum marker assays to predict tumor occurrence, progression and recurrence in OSCC, and to guide personalized therapies.
Collapse
|
42
|
Vangeti S, Falck-Jones S, Yu M, Österberg B, Liu S, Asghar M, Sondén K, Paterson C, Whitley P, Albert J, Johansson N, Färnert A, Smed-Sörensen A. Human influenza virus infection elicits distinct patterns of monocyte and dendritic cell mobilization in blood and the nasopharynx. eLife 2023; 12:77345. [PMID: 36752598 PMCID: PMC9977282 DOI: 10.7554/elife.77345] [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: 01/25/2022] [Accepted: 02/07/2023] [Indexed: 02/09/2023] Open
Abstract
During respiratory viral infections, the precise roles of monocytes and dendritic cells (DCs) in the nasopharynx in limiting infection and influencing disease severity are incompletely described. We studied circulating and nasopharyngeal monocytes and DCs in healthy controls (HCs) and in patients with mild to moderate infections (primarily influenza A virus [IAV]). As compared to HCs, patients with acute IAV infection displayed reduced DC but increased intermediate monocytes frequencies in blood, and an accumulation of most monocyte and DC subsets in the nasopharynx. IAV patients had more mature monocytes and DCs in the nasopharynx, and higher levels of TNFα, IL-6, and IFNα in plasma and the nasopharynx than HCs. In blood, monocytes were the most frequent cellular source of TNFα during IAV infection and remained responsive to additional stimulation with TLR7/8L. Immune responses in older patients skewed towards increased monocyte frequencies rather than DCs, suggesting a contributory role for monocytes in disease severity. In patients with other respiratory virus infections, we observed changes in monocyte and DC frequencies in the nasopharynx distinct from IAV patients, while differences in blood were more similar across infection groups. Using SomaScan, a high-throughput aptamer-based assay to study proteomic changes between patients and HCs, we found differential expression of innate immunity-related proteins in plasma and nasopharyngeal secretions of IAV and SARS-CoV-2 patients. Together, our findings demonstrate tissue-specific and pathogen-specific patterns of monocyte and DC function during human respiratory viral infections and highlight the importance of comparative investigations in blood and the nasopharynx.
Collapse
Affiliation(s)
- Sindhu Vangeti
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Sara Falck-Jones
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Meng Yu
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Björn Österberg
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Sang Liu
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Muhammad Asghar
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska InstitutetStockholmSweden
- Department of Infectious Diseases, Karolinska University HospitalStockholmSweden
| | - Klara Sondén
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska InstitutetStockholmSweden
- Department of Infectious Diseases, Karolinska University HospitalStockholmSweden
| | | | | | - Jan Albert
- Department of Microbiology, Tumor and Cell Biology, Karolinska InstitutetStockholmSweden
- Department of Clinical Microbiology, Karolinska University HospitalStockholmSweden
| | - Niclas Johansson
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska InstitutetStockholmSweden
- Department of Infectious Diseases, Karolinska University HospitalStockholmSweden
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska InstitutetStockholmSweden
- Department of Infectious Diseases, Karolinska University HospitalStockholmSweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University HospitalStockholmSweden
| |
Collapse
|
43
|
Mohammadi-Shemirani P, Sood T, Paré G. From 'Omics to Multi-omics Technologies: the Discovery of Novel Causal Mediators. Curr Atheroscler Rep 2023; 25:55-65. [PMID: 36595202 PMCID: PMC9807989 DOI: 10.1007/s11883-022-01078-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW 'Omics studies provide a comprehensive characterisation of a biological entity, such as the genome, epigenome, transcriptome, proteome, metabolome, or microbiome. This review covers the unique properties of these types of 'omics and their roles as causal mediators in cardiovascular disease. Moreover, applications and challenges of integrating multiple types of 'omics data to increase predictive power, improve causal inference, and elucidate biological mechanisms are discussed. RECENT FINDINGS Multi-omics approaches are growing in adoption as they provide orthogonal evidence and overcome the limitations of individual types of 'omics data. Studies with multiple types of 'omics data have improved the diagnosis and prediction of disease states and afforded a deeper understanding of underlying pathophysiological mechanisms, beyond any single type of 'omics data. For instance, disease-associated loci in the genome can be supplemented with other 'omics to prioritise causal genes and understand the function of non-coding variants. Alternatively, techniques, such as Mendelian randomisation, can leverage genetics to provide evidence supporting a causal role for disease-associated molecules, and elucidate their role in disease pathogenesis. As technologies improve, costs for 'omics studies will continue to fall and datasets will become increasingly accessible to researchers. The intrinsically unbiased nature of 'omics data is well-suited to exploratory analyses that discover causal mediators of disease, and multi-omics is an emerging discipline that leverages the strengths of each type of 'omics data to provide insights greater than the sum of its parts.
Collapse
Affiliation(s)
- Pedrum Mohammadi-Shemirani
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON Canada
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON Canada
| | - Tushar Sood
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON Canada
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON Canada
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON Canada
| |
Collapse
|
44
|
Kivimäki M, Hingorani AD, Lindbohm JV. Comment on "A proteomic surrogate for cardiovascular outcomes that is sensitive to multiple mechanisms of change in risk". Sci Transl Med 2022; 14:eabq4810. [PMID: 36197964 DOI: 10.1126/scitranslmed.abq4810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
A 27-protein signature has been proposed to predict cardiovascular disease, but its applicability in clinical decision-making remains unclear.
Collapse
Affiliation(s)
- Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, London WC1E 6BT, UK.,Clinicum, University of Helsinki, Helsinki FIN-00014, Finland
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK.,UCL British Heart Foundation Research Accelerator, London WC1E 6BT, UK.,UCL National Institute of Health Research Biomedical Research Centre, London W1T 7DN, UK.,Health Data Research UK, London NW1 2BE, UK
| | - Joni V Lindbohm
- Department of Epidemiology and Public Health, University College London, London WC1E 6BT, UK.,Clinicum, University of Helsinki, Helsinki FIN-00014, Finland.,Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| |
Collapse
|
45
|
Williams SA, Ganz P. Response to comment on "A proteomic surrogate for cardiovascular outcomes that is sensitive to multiple mechanisms of change in risk". Sci Transl Med 2022; 14:eadd1355. [PMID: 36197965 DOI: 10.1126/scitranslmed.add1355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
A 27-protein signature has been proposed to predict cardiovascular disease, and its applicability in some clinical decision-making situations is discussed.
Collapse
Affiliation(s)
| | - Peter Ganz
- Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
46
|
Stătescu C, Anghel L, Tudurachi BS, Leonte A, Benchea LC, Sascău RA. From Classic to Modern Prognostic Biomarkers in Patients with Acute Myocardial Infarction. Int J Mol Sci 2022; 23:9168. [PMID: 36012430 PMCID: PMC9409468 DOI: 10.3390/ijms23169168] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Despite all the important advances in its diagnosis and treatment, acute myocardial infarction (AMI) is still one of the most prominent causes of morbidity and mortality worldwide. Early identification of patients at high risk of poor outcomes through the measurement of various biomarker concentrations might contribute to more accurate risk stratification and help to guide more individualized therapeutic strategies, thus improving prognoses. The aim of this article is to provide an overview of the role and applications of cardiac biomarkers in risk stratification and prognostic assessment for patients with myocardial infarction. Although there is no ideal biomarker that can provide prognostic information for risk assessment in patients with AMI, the results obtained in recent years are promising. Several novel biomarkers related to the pathophysiological processes found in patients with myocardial infarction, such as inflammation, neurohormonal activation, myocardial stress, myocardial necrosis, cardiac remodeling and vasoactive processes, have been identified; they may bring additional value for AMI prognosis when included in multi-biomarker strategies. Furthermore, the use of artificial intelligence algorithms for risk stratification and prognostic assessment in these patients may have an extremely important role in improving outcomes.
Collapse
Affiliation(s)
- Cristian Stătescu
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iași, Romania
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iași, Romania
| | - Larisa Anghel
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iași, Romania
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iași, Romania
| | - Bogdan-Sorin Tudurachi
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iași, Romania
| | - Andreea Leonte
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iași, Romania
| | - Laura-Cătălina Benchea
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iași, Romania
| | - Radu-Andy Sascău
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iași, Romania
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iași, Romania
| |
Collapse
|
47
|
Rhee EP. Proteomic discovery in diabetic kidney disease-to what end? Kidney Int 2022; 102:236-238. [PMID: 35870813 DOI: 10.1016/j.kint.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 11/25/2022]
Abstract
Emerging approaches that enable high-throughput analysis of the plasma proteome have been increasingly deployed in nephrology research. Kobayashi et al. provide a valuable addition to this literature, describing an untargeted proteomic analysis of diabetic kidney disease progression to end-stage kidney disease. This commentary places the study's findings in the context of the broader literature and outlines potential avenues toward biological insight and clinical utility.
Collapse
Affiliation(s)
- Eugene P Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
48
|
Fernández-Ruiz I. A proteomic model shows potential as a surrogate end point for CVD risk. Nat Rev Cardiol 2022; 19:352. [PMID: 35444299 DOI: 10.1038/s41569-022-00716-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|