1
|
Blomdahl J, Åberg M, Fridén M, Ahlström H, Hockings P, Hulthe J, Eriksson N, Gabrysch K, Nasr P, Risérus U, Kechagias S, Rorsman F, Ekstedt M, Vessby J. Proteomic signatures for fibrosis in MASLD: a biopsy-proven dual-cohort study. Scand J Gastroenterol 2025; 60:597-605. [PMID: 40237197 DOI: 10.1080/00365521.2025.2490996] [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: 01/14/2025] [Revised: 03/30/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
OBJECTIVES Predicting disease progression in metabolic dysfunction-associated steatotic liver disease (MASLD) is challenging, and current non-invasive tests (NITs) lack the precision to replace liver biopsy. This study aimed to identify plasma biomarkers for different stages of fibrosis using affinity-based proteomics in two biopsy-proven cohorts. The primary objective was to identify biomarkers capable of distinguishing between low-to-no fibrosis (F0-1) and significant fibrosis (F2-4) in MASLD. MATERIALS AND METHODS Participants in the discovery cohort were recruited from Uppsala University Hospital and Swedish CArdioPulmonary bioImage Study (SCAPIS), while the validation cohort was included from Linköping University Hospital. All participants diagnosed with MASLD underwent liver biopsy and were categorized by fibrosis stage (F0-1 or F2-4). A total of 276 plasma proteins were analyzed using Olink® panels, with biomarkers identified through ordinal logistic regression, random forest (RF) analysis and the Boruta algorithm. RESULTS The discovery cohort included 60 participants, with 60% having fibrosis stage F0-1 and 40% having F2-4. The validation cohort had 59 participants, of whom 35 had fibrosis stage F0-1 (59.3%) and 24 had stage F2-4 (40.7%). Five biomarkers were significantly associated with fibrosis stage in the discovery cohort, with four confirmed in the validation cohort. A model combining angiotensin converting enzyme-2 (ACE2), hepatocyte growth factor (HGF) and insulin-like growth factor-binding protein-7 (IGFBP-7) demonstrated strong predictive performance for significant fibrosis (c-statistics 0.82-0.83), outperforming fibrosis-4 (FIB-4) (c-statistics 0.61-0.72). CONCLUSIONS A biomarker model including ACE2, HGF and IGFBP7 shows promise in distinguishing between low-stage and significant fibrosis.
Collapse
Affiliation(s)
- Julia Blomdahl
- Department of Medical Sciences, Gastroenterology Research Group, Uppsala University, Uppsala, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Michael Fridén
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Håkan Ahlström
- Department of Surgical Sciences, Section of Radiology, Uppsala University, Uppsala, Sweden
- Antaros Medical, Mölndal, Sweden
| | | | | | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Katja Gabrysch
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Patrik Nasr
- Division of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
- Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Stergios Kechagias
- Division of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Fredrik Rorsman
- Department of Medical Sciences, Gastroenterology Research Group, Uppsala University, Uppsala, Sweden
| | - Mattias Ekstedt
- Division of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Johan Vessby
- Department of Medical Sciences, Gastroenterology Research Group, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Birrenkott DA, Kabrhel C. The Plasma Proteome and Risk of Future Venous Thromboembolism-Results from the HUNT Study in Thrombosis and Haemostasis. Thromb Haemost 2025; 125:585-588. [PMID: 40280185 DOI: 10.1055/a-2575-3388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Affiliation(s)
- Drew A Birrenkott
- Department of Emergency Medicine, Center for Vascular Emergencies, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Christopher Kabrhel
- Department of Emergency Medicine, Center for Vascular Emergencies, Massachusetts General Hospital, Boston, Massachusetts, United States
| |
Collapse
|
3
|
Assi IZ, Landzberg MJ, Becker KC, Renaud D, Reyes FB, Leone DM, Benson M, Michel M, Gerszten RE, Opotowsky AR. Correlation between Olink and SomaScan proteomics platforms in adults with a Fontan circulation. INTERNATIONAL JOURNAL OF CARDIOLOGY CONGENITAL HEART DISEASE 2025; 20:100584. [PMID: 40330320 PMCID: PMC12053979 DOI: 10.1016/j.ijcchd.2025.100584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Background High-throughput proteomics platforms using aptamers (SomaScan) or proximity extension assay (Olink) provide novel opportunities for improving diagnostic and risk stratification tools in cardiovascular diseases, including understudied congenital heart diseases. The correlation between these proteomics approaches has not yet been studied among individuals with a Fontan circulation. Objective The correlation of plasma protein measurements between SomaScan and Olink platforms was evaluated in adults with a Fontan circulation. Methods We measured 491 proteins in plasma of 71 adults with a Fontan circulation using Olink and SomaScan. Missing Olink measurements (0.13%, 47/34,861) were imputed using non-parametric imputation. Spearman's rank correlation coefficient for absolute values of protein expression between platforms was calculated. Protein correlation frequencies were compared to 3 cohorts reported in the literature using Pearson's Chi-squared test of independence. Results Overall, protein correlations between Olink and SomaScan measurements were moderately strong for most proteins, (rho > 0.4 for 57.2%), but with substantial variability (median correlation = 0.457, IQR = 0.538). The distribution of protein correlations was qualitatively similar to published literature in non-Fontan cohorts. Both Olink and SomaScan identified proteins with sex-based differences; both identified differences in myostatin and leptin, but each identified additional nonoverlapping sexually dimorphic proteins (n = 14 Olink, n = 5 SomaScan). Conclusions In adults with a Fontan circulation, correlations between plasma proteins measured by Olink and SomaScan varied widely, approximately in line with prior reports in other populations. While these tools may be uniquely useful to generate hypotheses, specifically regarding potential molecular mechanisms, more definitive inference requires independent validation.
Collapse
Affiliation(s)
- Ismael Z. Assi
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael J. Landzberg
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kristian C. Becker
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David Renaud
- Fundamental and Biomedical Sciences, Paris-Cité University, Paris, France
- Health Sciences Faculty, Universidad Europea Miguel de Cervantes, Valladolid, Spain
| | - Fernando Baraona Reyes
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - David M. Leone
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mark Benson
- Harvard Medical School, Boston, MA, USA
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Miriam Michel
- Department of Child and Adolescent Health, Division of Pediatrics III — Cardiology, Pulmonology, Allergology and Cystic Fibrosis, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert E. Gerszten
- Harvard Medical School, Boston, MA, USA
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Alexander R. Opotowsky
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
4
|
Commissati S, Cagigas ML, Masedunskas A, Petrucci G, Tosti V, De Ciutiis I, Rajakumar G, Kirmess KM, Meyer MR, Goldhamer A, Kennedy BK, Hatem D, Rocca B, Fiorito G, Fontana L. Prolonged fasting promotes systemic inflammation and platelet activation in humans: A medically supervised, water-only fasting and refeeding study. Mol Metab 2025; 96:102152. [PMID: 40268190 PMCID: PMC12088818 DOI: 10.1016/j.molmet.2025.102152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVE Prolonged fasting (PF), defined as abstaining from energy intake for ≥4 consecutive days, has gained interest as a potential health intervention. However, the biological effects of PF on the plasma proteome are not well understood. METHODS In this study, we investigated the effects of a medically supervised water-only fast (mean duration: 9.8 ± 3.1 days), followed by 5.3 ± 2.4 days of guided refeeding, in 20 middle-aged volunteers (mean age: 52.2 ± 11.8 years; BMI: 28.8 ± 6.4 kg/m2). RESULTS Fasting resulted in a 7.7% mean weight loss and significant increases in serum beta-hydroxybutyrate (BHB), confirming adherence. Untargeted high-dimensional plasma proteomics (SOMAScan, 1,317 proteins) revealed multiple adaptations to PF, including preservation of skeletal muscle and bone, enhanced lysosomal biogenesis, increased lipid metabolism via PPARα signaling, and reduced amyloid fiber formation. Notably, PF significantly reduced circulating amyloid beta proteins Aβ40 and Aβ42, key components of brain amyloid plaques. In addition, PF induced an acute inflammatory response, characterized by elevated plasma C-reactive protein (CRP), hepcidin, midkine, and interleukin 8 (IL-8), among others. A retrospective cohort analysis of 1,422 individuals undergoing modified fasting confirmed increased CRP levels (from 2.8 ± 0.1 to 4.3 ± 0.2 mg/L). The acute phase response, associated with transforming growth factor (TGF)-β signaling, was accompanied by increased platelet degranulation and upregulation of the complement and coagulation cascade, validated by ELISAs in blood and urine. CONCLUSIONS While the acute inflammatory response during PF may serve as a transient adaptive mechanism, it raises concerns regarding potential cardiometabolic effects that could persist after refeeding. Further investigation is warranted to elucidate the long-term molecular and clinical implications of PF across diverse populations.
Collapse
Affiliation(s)
| | - Maria Lastra Cagigas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Andrius Masedunskas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Giovanna Petrucci
- Section of Pharmacology, Department of Safety and Bioethics, Catholic University School of Medicine, Rome, Italy
| | - Valeria Tosti
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Isabella De Ciutiis
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Gayathiri Rajakumar
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | - Brian K Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore; Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Duaa Hatem
- Section of Pharmacology, Department of Safety and Bioethics, Catholic University School of Medicine, Rome, Italy
| | - Bianca Rocca
- Section of Pharmacology, Department of Safety and Bioethics, Catholic University School of Medicine, Rome, Italy; NeuroFarBa Department, University of Florence, Florence, Italy
| | - Giovanni Fiorito
- Clinical Bioinformatics unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy; MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia.
| |
Collapse
|
5
|
Onsaker AL, Arntzen AY, Trégouët DA, Nøst TH, Tang W, Guan W, Jonasson C, Morange PE, Hindberg KD, Folsom AR, Hveem K, Morelli VM, Hansen JB. Histo-blood group ABO system transferase plasma levels and risk of future venous thromboembolism: the HUNT study. Blood 2025; 145:2656-2665. [PMID: 40009491 PMCID: PMC12163907 DOI: 10.1182/blood.2024025923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/23/2024] [Accepted: 01/29/2025] [Indexed: 02/28/2025] Open
Abstract
ABSTRACT The non-O blood group is a well-established risk factor for venous thromboembolism (VTE). However, the association between plasma levels of the histo-blood group ABO system transferase (BGAT), the gene product of the ABO locus, and VTE risk remains unclear. We aimed to investigate the association between plasma BGAT levels and risk of future VTE, and whether this relationship was mediated by plasma von Willebrand factor (VWF) or coagulation factor VIII (FVIII), as VWF is glycosylated by BGAT. Incident VTE-cases (n = 294) and a randomly sampled age- and-sex-weighted subcohort (n = 1066) were derived from the third survey of the Trøndelag Health Study. Baseline plasma samples (2006-2008) were subjected to the SomaScan aptamer-based-7K platform for protein measurements. Weighted Cox regression was used to estimate hazard ratios (HRs) with 95% confidence intervals (CIs) across BGAT quartiles. We found that ABO haplotypes (A1/A2/B/O1/O2) explained ≈80% of the BGAT plasma variability. Participants with BGAT levels in the highest quartile had 2-fold higher VTE risk (HR, 2.12; 95% CI, 1.39-3.22) compared with those with BGAT in the lowest quartile in age-, sex-, and sample batch-adjusted models. The associations were particularly pronounced for unprovoked VTE (HR, 3.71; 95% CI, 1.79-7.67) and deep vein thrombosis (HR, 3.28; 95% CI, 1.63-6.59). The HRs were similar after further adjustment for body mass index, C-reactive protein, and estimated glomerular filtration rate, and moderately attenuated when adding VWF or FVIII plasma levels to the models. Our findings indicate that elevated BGAT plasma levels are associated with increased risk of future VTE beyond what is explained by VWF and FVIII.
Collapse
Affiliation(s)
- Asbjørn L. Onsaker
- Thrombosis Research Group, Department of Clinical Medicine, UiT–the Arctic University of Norway, Tromsø, Norway
| | - Anna Y. Arntzen
- Thrombosis Research Group, Department of Clinical Medicine, UiT–the Arctic University of Norway, Tromsø, Norway
| | - David-Alexandre Trégouët
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, Unité Mixte de Recherche 1219, ELEANOR, Bordeaux, France
| | - Therese H. Nøst
- HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
- Department of Community Medicine, UiT–the Arctic University of Norway, Tromsø, Norway
- HUNT Research Center, Levanger, Norway
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN
| | - Weihua Guan
- Division of Biostatistics and Health Data Science, University of Minnesota School of Public Health, Minneapolis, MN
| | - Christian Jonasson
- HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Center, Levanger, Norway
| | - Pierre-Emmanuel Morange
- Aix-Marseille Univ, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Centre de Recherche en CardioVasculaire et Nutrition, Laboratory of Haematology, Centre de Ressources Biologiques Assistance Publique–Hôpitaux de Marseille, HemoVasc, Marseille, France
| | - Kristian D. Hindberg
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Aaron R. Folsom
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
- HUNT Research Center, Levanger, Norway
| | - Vânia M. Morelli
- Thrombosis Research Group, Department of Clinical Medicine, UiT–the Arctic University of Norway, Tromsø, Norway
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - John-Bjarne Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT–the Arctic University of Norway, Tromsø, Norway
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
6
|
Candia J, Fantoni G, Moaddel R, Delgado-Peraza F, Shehadeh N, Tanaka T, Ferrucci L. Effects of In Vitro Hemolysis and Repeated Freeze-Thaw Cycles in Protein Abundance Quantification Using the SomaScan and Olink Assays. J Proteome Res 2025; 24:2517-2528. [PMID: 40249843 PMCID: PMC12053949 DOI: 10.1021/acs.jproteome.5c00069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/17/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
SomaScan and Olink are affinity-based platforms that aim to estimate the relative abundance of thousands of human proteins with a broad range of endogenous concentrations. In this study, we investigated the effects of in vitro hemolysis and repeated freeze-thaw cycles in protein abundance quantification across 10,776 (11 K SomaScan) and 1472 (Olink Explore 1536) analytes, respectively. Using SomaScan, we found two distinct groups, each one consisting of 4% of all aptamers, affected by either hemolysis or freeze-thaw cycles. Using Olink, we found 6% of analytes affected by freeze-thaw cycles and nearly half of all measured probes significantly impacted by hemolysis. Moreover, we observed that Olink probes affected by hemolysis target proteins with a larger number of annotated protein-protein interactions. We found that Olink probes affected by hemolysis were significantly associated with the erythrocyte proteome, whereas SomaScan probes were not. Given the extent of the observed nuisance effects, we propose that unbiased, quantitative methods of evaluating hemolysis, such as the hemolysis index successfully implemented in many clinical laboratories, should be adopted in proteomics studies. We provide detailed results for each SomaScan and Olink probe in the form of extensive Supporting Information files to be used as resources for the growing user communities of both platforms.
Collapse
Affiliation(s)
| | | | | | - Francheska Delgado-Peraza
- Intramural Research Program, National Institute on Aging, National Institutes of
Health, Baltimore 21224, Maryland, United States
| | - Nader Shehadeh
- Intramural Research Program, National Institute on Aging, National Institutes of
Health, Baltimore 21224, Maryland, United States
| | - Toshiko Tanaka
- Intramural Research Program, National Institute on Aging, National Institutes of
Health, Baltimore 21224, Maryland, United States
| | - Luigi Ferrucci
- Intramural Research Program, National Institute on Aging, National Institutes of
Health, Baltimore 21224, Maryland, United States
| |
Collapse
|
7
|
Hoofnagle AN, MacCoss MJ. The Need for Better Validation: Evaluating Aptamer and Proximity Extension Assays for Large-Scale Clinical Proteomics Studies. Clin Chem 2025:hvaf046. [PMID: 40272410 DOI: 10.1093/clinchem/hvaf046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025]
Affiliation(s)
- Andrew N Hoofnagle
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, United States
| |
Collapse
|
8
|
Candia J, Fantoni G, Moaddel R, Delgado-Peraza F, Shehadeh N, Tanaka T, Ferrucci L. Effects of in vitro hemolysis and repeated freeze-thaw cycles in protein abundance quantification using the SomaScan and Olink assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.21.613295. [PMID: 40166260 PMCID: PMC11956925 DOI: 10.1101/2024.09.21.613295] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
SomaScan and Olink are affinity-based platforms that aim to estimate the relative abundance of thousands of human proteins with a broad range of endogenous concentrations. In this study, we investigated the effects of in vitro hemolysis and repeated freeze-thaw cycles in protein abundance quantification across 10,776 (11K SomaScan) and 1472 (Olink Explore 1536) analytes, respectively. Using SomaScan, we found two distinct groups, each one consisting of 4% of all aptamers, affected by either hemolysis or freeze-thaw cycles. Using Olink, we found 6% of analytes affected by freeze-thaw cycles and nearly half of all measured probes significantly impacted by hemolysis. Moreover, we observed that Olink probes affected by hemolysis target proteins with a larger number of annotated protein-protein interactions. We found that Olink probes affected by hemolysis were significantly associated with the erythrocyte proteome, whereas SomaScan probes were not. Given the extent of the observed nuisance effects, we propose that unbiased, quantitative methods of evaluating hemolysis, such as the hemolysis index successfully implemented in many clinical laboratories, should be adopted in proteomics studies. We provide detailed results for each SomaScan and Olink probe in the form of extensive Supplementary Data files to be used as resources for the growing user communities of both platforms.
Collapse
Affiliation(s)
- Julián Candia
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Giovanna Fantoni
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ruin Moaddel
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Francheska Delgado-Peraza
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Nader Shehadeh
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Toshiko Tanaka
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
9
|
Godina C, Rosendahl AH, Gonçalves de Oliveira K, Khazaei S, Björner S, Jirström K, Isaksson K, Pollak MN, Jernström H. Genetic determinants and clinical significance of circulating and tumor-specific levels of insulin-like growth factor binding protein 7 (IGFBP7) in a Swedish breast cancer cohort. Carcinogenesis 2025; 46:bgaf020. [PMID: 40230015 PMCID: PMC12066007 DOI: 10.1093/carcin/bgaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025] Open
Abstract
Previous research indicates that insulin-like growth factor binding protein 7 (IGFBP7) protein levels in breast cancer tissue and blood are prognostic. However, genetic determinants of IGFBP7 in breast cancer remain largely unexplored. We examined IGFBP7 in a cohort of 1701 patients with first breast cancer from Sweden, enrolled prior to surgery 2002-16 and followed for up to 15 years. Genotyping was performed on blood samples using OncoArray. Tumor-specific protein levels of IGFBP7, insulin receptor (InsR), and IGF-I receptor (IGFIR) were assessed on tumor tissue microarrays in 964 patients. Furthermore, 275 patients had plasma IGFBP7 levels measured. A genetic proxy marker for circulating IGFBP7 levels was constructed from five candidate single-nucleotide polymorphisms (SNPs) (rs6852762, rs1714014, rs9992658, rs10004910, and rs4865180) based on number of recessive genotypes. Age-adjusted linear regression was used to evaluate SNPs and tumor-specific IGFBP7 levels in relation to circulating IGFBP7 levels. Cox regression adjusted for age, tumor characteristics, and adjuvant treatments was used to assess associations with clinical outcomes. Circulating and tumor-specific IGFBP7 levels were significantly positively correlated. High circulating and genetically predicted IGFBP7 levels were associated with increased risk for distant metastasis and all-cause mortality. A significant interaction between high tumor-specific IGFBP7 levels and membrane-bound InsR resulted in a four-fold increased risk of breast cancer events and distant metastases. Both measured and genetically predicted IGFBP7 levels were independent prognostic biomarkers in breast cancer.
Collapse
Affiliation(s)
- Christopher Godina
- Department of Clinical Sciences Lund, Oncology, Lund University Cancer Center/Kamprad, Lund University and Skåne University Hospital, Barngatan4, SE 221 85 Lund, Sweden
| | - Ann H Rosendahl
- Department of Clinical Sciences Lund, Oncology, Lund University Cancer Center/Kamprad, Lund University and Skåne University Hospital, Barngatan4, SE 221 85 Lund, Sweden
| | - Kelin Gonçalves de Oliveira
- Department of Clinical Sciences Lund, Oncology, Lund University Cancer Center/Kamprad, Lund University and Skåne University Hospital, Barngatan4, SE 221 85 Lund, Sweden
| | - Somayeh Khazaei
- Department of Clinical Sciences Lund, Oncology, Lund University Cancer Center/Kamprad, Lund University and Skåne University Hospital, Barngatan4, SE 221 85 Lund, Sweden
| | - Sofie Björner
- Department of Clinical Sciences Lund, Oncology, Lund University Cancer Center/Kamprad, Lund University and Skåne University Hospital, Barngatan4, SE 221 85 Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Oncology and Therapeutic Pathology, Lund University Cancer Center/Kamprad, Lund University, Barngatan 4, SE 221 85 Lund, Sweden
| | - Karolin Isaksson
- Department of Clinical Sciences Lund, Surgery, Lund University Cancer Center, Lund University and Kristianstad Hospital, JA Hedlundsväg 5, SE 291 33 Kristianstad, Sweden
| | - Michael N Pollak
- Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Oncology, McGill University, 3755 Côte Ste-Catherine Road, Montreal, QC H3T 1E2, Quebec, Canada
| | - Helena Jernström
- Department of Clinical Sciences Lund, Oncology, Lund University Cancer Center/Kamprad, Lund University and Skåne University Hospital, Barngatan4, SE 221 85 Lund, Sweden
| |
Collapse
|
10
|
Rooney MR, Chen J, Ballantyne CM, Hoogeveen RC, Boerwinkle E, Yu B, Walker KA, Schlosser P, Selvin E, Chatterjee N, Couper D, Grams ME, Coresh J. Correlations Within and Between Highly Multiplexed Proteomic Assays of Human Plasma. Clin Chem 2025:hvaf030. [PMID: 40172053 DOI: 10.1093/clinchem/hvaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/11/2025] [Indexed: 04/04/2025]
Abstract
INTRODUCTION The number of assays on proteomic platforms has grown rapidly. The leading platforms, SomaScan and Olink, have strengths and limitations. Comparisons of precision on the latest platforms-SomaScan 11k and Olink Explore HT-have not yet been established. METHODS Among 102 participants in the Atherosclerosis Risk in Communities Study (mean age 74 years, 53% women, 47% Black), we used split plasma samples to measure platform precision. CV and Spearman correlations were calculated for each assay. Cross-platform agreement was assessed for overlapping proteins. RESULTS SomaScan 11k demonstrated a median correlation of 0.85 for the 10 778 assays and a median CV of 6.8%, similar precision to earlier versions. The 5420 assays on Olink Explore HT exhibited a median correlation of 0.65 and median CV of 35.7%, which was higher than observed in its predecessors (e.g., 19.8% for Olink Explore 3072). Precision of Olink assays was inversely correlated with the percentage of samples above the limit of detection (LOD) (r = -0.77). Upon replacing Olink values below the LOD with values half the LOD, the median correlation for Olink assays measured in duplicate increased to 0.79; the median CV decreased to 13.3%. The distribution of between-platform correlations for the 4443 overlapping proteins had peaks at r approximately 0 and at r approximately 0.8. One-tenth of the protein pairs had cross-platform correlations r ≥ 0.8. CONCLUSIONS Precision of these 2 proteomics platforms in human plasma has diverged as the coverage has increased. These results highlight the need for careful consideration in platform selection based on specific research requirements.
Collapse
Affiliation(s)
- Mary R Rooney
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | - Ron C Hoogeveen
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Eric Boerwinkle
- Department of Epidemiology, University of Texas Health Science Center, Houston, TX, United States
| | - Bing Yu
- Department of Epidemiology, University of Texas Health Science Center, Houston, TX, United States
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, United States
| | - Pascal Schlosser
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - David Couper
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Morgan E Grams
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, 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
| |
Collapse
|
11
|
Kou M, Ma H, Wang X, Heianza Y, Qi L. Plasma proteomics-based brain aging signature and incident dementia risk. GeroScience 2025; 47:2335-2349. [PMID: 39532828 PMCID: PMC11978599 DOI: 10.1007/s11357-024-01407-6] [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: 08/07/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Investigating brain-enriched proteins with machine learning methods may enable a brain-specific understanding of brain aging and provide insights into the molecular mechanisms and pathological pathways of dementia. The study aims to analyze associations of brain-specific plasma proteomic aging signature with risks of incident dementia. In 45,429 dementia-free UK Biobank participants at baseline, we generated a brain-specific biological age using 63 brain-enriched plasma proteins with machine learning methods. The brain age gap was estimated, and Cox proportional hazards models were used to study the association with incident all-cause dementia, Alzheimer's disease (AD), and vascular dementia. Per-unit increment in the brain age gap z-score was associated with significantly higher risks of all-cause dementia (hazard ratio [95% confidence interval], 1.67 [1.56-1.79], P < 0.001), AD (1.85 [1.66-2.08], P < 0.001), and vascular dementia (1.86 [1.55-2.24], P < 0.001), respectively. Notably, 2.1% of the study population exhibited extreme old brain aging defined as brain age gap z-score > 2, correlating with over threefold increased risks of all-cause dementia and vascular dementia (3.42 [2.25-5.20], P < 0.001, and 3.41 [1.05-11.13], P = 0.042, respectively), and fourfold increased risk of AD (4.45 [2.32-8.54], P < 0.001). The associations were stronger among participants with healthier lifestyle factors (all P-interaction < 0.05). These findings were corroborated by magnetic resonance imaging assessments showing that a higher brain age gap aligns global pathophysiology of dementia, including global and regional atrophy in gray matter, and white matter lesions (P < 0.001). The brain-specific proteomic age gap is a powerful biomarker of brain aging, indicative of dementia risk and neurodegeneration.
Collapse
Affiliation(s)
- Minghao Kou
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Hao Ma
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Xuan Wang
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Yoriko Heianza
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lu Qi
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
12
|
Mischak H, Schanstra JP, Vlahou A, Beige J. Clinical Proteomics, Quo Vadis? Proteomics 2025; 25:e202400346. [PMID: 39924729 PMCID: PMC11962580 DOI: 10.1002/pmic.202400346] [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: 12/02/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/11/2025]
Abstract
The field of clinical proteomics has seen enormous growth in the past 20 years, with over 40,000 scientific manuscripts published to date. At the same time, actual clinical application of the reported findings is obviously scarce. In this viewpoint article, we discuss the key issues that may be responsible for this apparent lack of success. We conclude that success must not be assessed based on the number of publications, but via the impact on patient management and treatment. We proceed with specific suggestions for potential solutions, which include keeping a strict focus on potential patient benefit. We hope this article can help shape the field, so it can in fact deliver on its realistic promise to bring significant improvement in management and care to patients.
Collapse
Affiliation(s)
| | - Joost P. Schanstra
- Institute of Cardiovascular and Metabolic DiseaseU1297Institut National de la Santé et de la Recherche MédicaleToulouseFrance
- Université de ToulouseToulouseFrance
| | - Antonia Vlahou
- Center of Systems BiologyBiomedical Research Foundation of the Academy of AthensAthensGreece
| | - Joachim Beige
- Martin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
- Kuratorium for Dialysis and TransplantationLeipzigGermany
| |
Collapse
|
13
|
Gan YH, Ma LZ, Zhang Y, You J, Guo Y, He Y, Wang LB, He XY, Li YZ, Dong Q, Feng JF, Cheng W, Yu JT. Large-scale proteomic analyses of incident Parkinson's disease reveal new pathophysiological insights and potential biomarkers. NATURE AGING 2025; 5:642-657. [PMID: 39979637 DOI: 10.1038/s43587-025-00818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
The early pathophysiology of Parkinson's disease (PD) is poorly understood. We analyzed 2,920 Olink-measured plasma proteins in 51,804 UK Biobank participants, identifying 859 incident PD cases after 14.45 years. We found 38 PD-related proteins, with six of the top ten validated in the Parkinson's Progression Markers Initiative (PPMI) cohort. ITGAV, HNMT and ITGAM showed consistent significant association (hazard ratio: 0.11-0.57, P = 6.90 × 10-24 to 2.10 × 10-11). Lipid metabolism dysfunction was evident 15 years before PD onset, and levels of BAG3, HPGDS, ITGAV and PEPD continuously decreased before diagnosis. These proteins were linked to prodromal symptoms and brain measures. Mendelian randomization suggested ITGAM and EGFR as potential causes of PD. A predictive model using machine learning combined the top 16 proteins and demographics, achieving high accuracy for 5-year (area under the curve (AUC) = 0.887) and over-5-year PD prediction (AUC = 0.816), outperforming demographic-only models. It was externally validated in PPMI (AUC = 0.802). Our findings reveal early peripheral pathophysiological changes in PD crucial for developing early biomarkers and precision therapies.
Collapse
Affiliation(s)
- Yi-Han Gan
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling-Zhi Ma
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin-Bo Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Zhu Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science,Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Spence J, Devereaux PJ, Bashir S, Brady K, Sun T, Chan MTV, Wang CY, Lamy A, Whitlock RP, McIntyre WF, Belley-Côté E, Paré G, Chong M. Protein Alterations in Patients with Delirium after Cardiac Surgery: An Exploratory Case-Control Substudy of the VISION Cardiac Surgery Biobank. Anesthesiology 2025; 142:716-725. [PMID: 39786937 DOI: 10.1097/aln.0000000000005368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND Delirium is an acute state of confusion associated with adverse postoperative outcomes. Delirium is diagnosed clinically using screening tools; most cases go undetected. Identifying a delirium biomarker would allow for accurate diagnosis, application of therapies, and insight into causal pathways. To agnostically discover novel biomarkers of delirium, a case-control substudy was conducted using the Vascular Events in Surgery Patients Cohort Evaluation (VISION) Cardiac Surgery Biobank. The objective was to identify candidate biomarkers to investigate in future studies. METHODS The study gathered a convenience sample of 30 patients with delirium on postoperative day 1 matched to 30 controls by age, sex, ethnicity, center, and cardiopulmonary bypass time. The Olink Explore 3K platform was used to identify blood protein alterations on postoperative day 3. Protein concentrations were expressed as normalized protein expression units (log 2 fold scale). Protein expression was compared between cases and controls using a paired t test and identified significantly different biomarkers based on a false discovery rate-adjusted P value of less than 0.05. RESULTS Of 2,865 unique serum proteins, 26 (0.9%) were significantly associated with delirium status; all were elevated in cases versus controls at a false discovery rate of less than 0.05. Pathway analysis identified calcium-release channel activity ( Padj = 0.02) and GTP-binding ( Padj = 0.005) functions as characteristic of proteins associated with delirium. The top three differentially expressed biomarkers were FKBP1B ( Padj = 0.003), C2CD2L ( Padj = 0.004), and RAB6B ( Padj = 0.004). The inflammatory biomarker interleukin-8 (CXCL8; mean difference = 2.36; P = 3.6 × 10- 4 ) was also associated with delirium. CONCLUSIONS The study identified 26 biomarkers significantly associated with delirium; all are novel except for interleukin-8. An association between delirium and recognized neuroinflammatory proteins or markers of brain injury was not identifed, which supports using biomarkers to differentiate between delirium and other neurologic conditions. While exploratory, the study's findings support using biomarkers to diagnose postoperative delirium and validate using agnostic screens to identify potential delirium biomarkers.
Collapse
Affiliation(s)
- Jessica Spence
- Population Health Research Institute, Hamilton, Ontario, Canada; World Health Research Trust, Hamilton, Ontario, Canada; Departments of Anesthesia, Critical Care, and Health Research Methods, Evaluation, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - P J Devereaux
- Population Health Research Institute, Hamilton, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada; World Health Research Trust, Hamilton, Ontario, Canada; Department of Medicine (Cardiology), McMaster University, Hamilton, Ontario, Canada
| | - Shaheena Bashir
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Katheryn Brady
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Tao Sun
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chew Yin Wang
- Department of Anaesthesiology, University of Malaya, Kuala Lumpur, Wilayah Persekutuan, Malaysia
| | - Andre Lamy
- Population Health Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada; World Health Research Trust, Hamilton, Ontario, Canada; Department of Surgery (Cardiac Surgery), McMaster University, Hamilton, Ontario, Canada
| | - Richard P Whitlock
- Population Health Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada; World Health Research Trust, Hamilton, Ontario, Canada; Department of Surgery (Cardiac Surgery), McMaster University, Hamilton, Ontario, Canada
| | - William F McIntyre
- Population Health Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada; World Health Research Trust, Hamilton, Ontario, Canada; Department of Medicine (Cardiology), McMaster University, Hamilton, Ontario, Canada
| | - Emilie Belley-Côté
- Population Health Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada; World Health Research Trust, Hamilton, Ontario, Canada; Department of Medicine (Cardiology and Critical Care), McMaster University, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Population Health Research Institute, Hamilton, Ontario, Canada; World Health Research Trust, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Michael Chong
- Population Health Research Institute, Hamilton, Canada; World Health Research Trust, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
15
|
Huang T, Campos AR, Wang J, Stukalov A, Díaz R, Maurya S, Motamedchaboki K, Hornburg D, Saciloto-de-Oliveira LR, Innocente-Alves C, Calegari-Alves YP, Batzoglou S, Beys-da-Silva WO, Santi L. Deep, Unbiased, and Quantitative Mass Spectrometry-Based Plasma Proteome Analysis of Individual Responses to mRNA COVID-19 Vaccine. J Proteome Res 2025; 24:1265-1274. [PMID: 39904632 DOI: 10.1021/acs.jproteome.4c00909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Global campaign against COVID-19 have vaccinated a significant portion of the world population in recent years. Combating the COVID-19 pandemic with mRNA vaccines played a pivotal role in the global immunization effort. However, individual responses to a vaccine are diverse and lead to varying vaccination efficacy. Despite significant progress, a complete understanding of the molecular mechanisms driving the individual immune response to the COVID-19 vaccine remains elusive. To address this gap, we combined a novel nanoparticle-based proteomic workflow with tandem mass tag (TMT) labeling, to quantitatively assess the proteomic changes in a cohort of 12 volunteers following two doses of the Pfizer-BioNTech mRNA COVID-19 vaccine. This optimized protocol seamlessly integrates comprehensive proteome analysis with enhanced throughput by leveraging the enrichment of low-abundant plasma proteins by engineered nanoparticles. Our data demonstrate the ability of this workflow to quantify over 3,000 proteins, providing the deepest view into COVID-19 vaccine-related plasma proteome study. We identified 69 proteins with boosted responses post-second dose and 74 proteins differentially regulated between individuals who contracted COVID-19 despite vaccination and those who did not. These findings offer valuable insights into individual variability in response to vaccination, demonstrating the potential of personalized medicine approaches in vaccine development.
Collapse
Affiliation(s)
- Ting Huang
- Seer, Inc., Redwood City, California 94065, United States
| | - Alex Rosa Campos
- Sanford Burnham Prebys, San Diego, California 92037, United States
| | - Jian Wang
- Seer, Inc., Redwood City, California 94065, United States
| | | | - Ramón Díaz
- Sanford Burnham Prebys, San Diego, California 92037, United States
| | - Svetlana Maurya
- Sanford Burnham Prebys, San Diego, California 92037, United States
| | | | | | | | - Camila Innocente-Alves
- Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | | | | | - Walter O Beys-da-Silva
- Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Lucélia Santi
- Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| |
Collapse
|
16
|
Zhan H, Cammann D, Cummings JL, Dong X, Chen J. Biomarker identification for Alzheimer's disease through integration of comprehensive Mendelian randomization and proteomics data. J Transl Med 2025; 23:278. [PMID: 40050982 PMCID: PMC11884171 DOI: 10.1186/s12967-025-06317-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/23/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the main cause of dementia with few effective therapies. We aimed to identify potential plasma biomarkers or drug targets for AD by investigating the causal association between plasma proteins and AD by integrating comprehensive Mendelian randomization (MR) and multi-omics data. METHODS Using two-sample MR, cis protein quantitative trait loci (cis-pQTLs) for 1,916 plasma proteins were used as an exposure to infer their causal effect on AD liability in individuals of European ancestry, with two large-scale AD genome-wide association study (GWAS) datasets as the outcome for discovery and replication. Significant causal relationships were validated by sensitivity analyses, reverse MR analysis, and Bayesian colocalization analysis. Additionally, we investigated the causal associations at the transcriptional level with cis gene expression quantitative trait loci (cis-eQTLs) data across brain tissues and blood in European ancestry populations, as well as causal plasma proteins in African ancestry populations. RESULTS In those of European ancestry, the genetically predicted levels of five plasma proteins (BLNK, CD2AP, GRN, PILRA, and PILRB) were causally associated with AD. Among these five proteins, GRN was protective against AD, while the rest were risk factors. Consistent causal effects were found in the brain for cis-eQTLs of GRN, BLNK, and CD2AP, while the same was true for PILRA in the blood. None of the plasma proteins were significantly associated with AD in persons of African ancestry. CONCLUSIONS Comprehensive MR analyses with multi-omics data identified five plasma proteins that had causal effects on AD, highlighting potential biomarkers or drug targets for better diagnosis and treatment for AD.
Collapse
Affiliation(s)
- Hui Zhan
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Davis Cammann
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
- School of Life Science, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Xianjun Dong
- Stephen and Denise Adams Center for Parkinson's Disease Research, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurology and Section of Biomedical Informatics and Data Science (BIDS), Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jingchun Chen
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
- School of Life Science, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
| |
Collapse
|
17
|
Niu L, Stinson SE, Holm LA, Lund MAV, Fonvig CE, Cobuccio L, Meisner J, Juel HB, Fadista J, Thiele M, Krag A, Holm JC, Rasmussen S, Hansen T, Mann M. Plasma proteome variation and its genetic determinants in children and adolescents. Nat Genet 2025; 57:635-646. [PMID: 39972214 PMCID: PMC11906355 DOI: 10.1038/s41588-025-02089-2] [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: 05/04/2023] [Accepted: 01/13/2025] [Indexed: 02/21/2025]
Abstract
Our current understanding of the determinants of plasma proteome variation during pediatric development remains incomplete. Here, we show that genetic variants, age, sex and body mass index significantly influence this variation. Using a streamlined and highly quantitative mass spectrometry-based proteomics workflow, we analyzed plasma from 2,147 children and adolescents, identifying 1,216 proteins after quality control. Notably, the levels of 70% of these were associated with at least one of the aforementioned factors, with protein levels also being predictive. Quantitative trait loci (QTLs) regulated at least one-third of the proteins; between a few percent and up to 30-fold. Together with excellent replication in an additional 1,000 children and 558 adults, this reveals substantial genetic effects on plasma protein levels, persisting from childhood into adulthood. Through Mendelian randomization and colocalization analyses, we identified 41 causal genes for 33 cardiometabolic traits, emphasizing the value of protein QTLs in drug target identification and disease understanding.
Collapse
Affiliation(s)
- Lili Niu
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- Novo Nordisk A/S, Copenhagen, Denmark
| | - Sara Elizabeth Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
| | - Morten Asp Vonsild Lund
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cilius Esmann Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leonardo Cobuccio
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Meisner
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Helene Bæk Juel
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Maja Thiele
- Odense Liver Research Centre, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Aleksander Krag
- Odense Liver Research Centre, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon Rasmussen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
18
|
Xia F, Duan Q, Zhang Q, Feng W, Ding D, Ji DK, Wang X, Tan W. Self-assembled aptamer nanoparticles for enhanced recognition and anticancer therapy through a lysosome-independent pathway. Acta Biomater 2025; 194:364-372. [PMID: 39863148 DOI: 10.1016/j.actbio.2025.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Aptamers and aptamer-drug conjugates (ApDCs) have shown some success as targeted therapies in cancer theranostics. However, their stability in complex media and their capacity to evade lysosomal breakdown still need improvement. To address these challenges, we herein developed a one-step self-assembly strategy to improve the stability of aptamers or ApDCs, while simultaneously enhancing their delivery performance and therapeutic efficiency through a lysosome-independent pathway. This strategy involves the formation of stable complexes between disulfide monomer and aptamers (Sgc8) or ApDCs (Gem-Sgc8). Self-assembled Sgc8 NPs resisted nuclease degradation for up to 24 h, whereas the aptamer alone degraded within just 3 h. These self-assembled Sgc8 NPs, as well as Gem-Sgc8 NPs, demonstrated enhanced binding capabilities compared to Sgc8 aptamers or Gem-Sgc8 alone. Furthermore, lysosome-independent cellular uptake was significantly improved, which in turn increased the therapeutic efficacy of Gem-Sgc8 NPs by 2.5 times compared to Gem-Sgc8 alone. In vivo results demonstrated that Gem-Sgc8 NPs can effectively suppress the growth of tumors. The same self-assembly strategy was successfully applied to other aptamers, such as MJ5C and cMET, showing the generalizability of our method, Overall, this aptamer self-assembly strategy not only overcomes the limitations associated with instability and lysosomal degradation but also demonstrates its broad applicability, highlighting its potential as a promising avenue for advancing targeted cancer theranostics. STATEMENT OF SIGNIFICANCE: We developed a one-step self-assembly strategy to improve the stability of aptamers or ApDCs and enhance their drug therapeutic efficiency through a lysosome-independent pathway. The stability of self-assembled Sgc8 nanoparticles (NPs) was significantly improved. The resulting Sgc8 NPs or GEM-Sgc8 NPs exhibited enhanced binding ability compared to Sgc8 aptamers or GEM-Sgc8 alone, and they also facilitated lysosome-independent cellular uptake, resulting in a 2.5-fold increase in therapeutic efficacy of GEM-Sgc8-NPs. The same self-assembly strategy was successfully applied to other aptamers, such as MJ5C and cMET, showing the generalizability of our method.
Collapse
Affiliation(s)
- Fangfang Xia
- Shanghai Institute of virology, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Qiao Duan
- Shanghai Institute of virology, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, PR China
| | - Qing Zhang
- Shanghai Institute of virology, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Wenqi Feng
- Shanghai Institute of virology, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Ding Ding
- Shanghai Institute of virology, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Ding-Kun Ji
- Shanghai Institute of virology, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Xiang Wang
- Shanghai Institute of virology, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Weihong Tan
- Shanghai Institute of virology, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, PR China; Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, PR China.
| |
Collapse
|
19
|
Michaëlsson K, Zheng R, Baron JA, Fall T, Wolk A, Lind L, Höijer J, Brunius C, Warensjö Lemming E, Titova OE, Svennblad B, Larsson SC, Yuan S, Melhus H, Byberg L, Brooke HL. Cardio-metabolic-related plasma proteins reveal biological links between cardiovascular diseases and fragility fractures: a cohort and Mendelian randomisation investigation. EBioMedicine 2025; 113:105580. [PMID: 39919333 PMCID: PMC11848109 DOI: 10.1016/j.ebiom.2025.105580] [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: 03/05/2024] [Revised: 12/17/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND How cardiovascular diseases (CVD) predispose to a higher risk of fragility fractures is not well understood. Both contribute to significant components of disease burden and health expenditure. Poor bone quality, central obesity, sarcopenia, falls, and low grip strength are independent risk factors for hip and other fragility fractures and also for CVD and early death. METHODS We used proteomics and a cohort design combined with Mendelian randomisation analysis to understand shared mechanisms for developing CVD and fragility fractures, two significant sources of disease burden and health expenditure. We primarily aimed to discover and replicate the association of 274 cardio-metabolic-related proteins with future rates of hip and any fracture in two separate population-based cohorts, with a total of 12,314 women and men. FINDINGS The average age at baseline was 68 years in the discovery cohort of women and 74 years in the mixed-sex replication cohort. During 100,619 person-years of follow-up, 2168 had any fracture, and 538 had a hip fracture. Our analysis resulted in 24 cardiometabolic proteins associated with fracture risk: 20 with hip fracture, 9 with any fracture, and 5 with both. The associations remained even if protein concentrations were measured from specimens taken during preclinical stages of cardio-metabolic diseases, and 19 associations remained after adjustment for bone mineral density. Twenty-two of the proteins were associated with total body fat mass or lean body mass. Mendelian randomisation (MR) analysis supported causality since genetically predicted levels of SOST (Sclerostin), CCDC80 (Coiled-coil domain-containing protein 80), NT-proBNP (N-terminal prohormone brain natriuretic peptide), and BNP (Brain natriuretic peptide) were associated with risk of hip fracture. MR analysis also revealed a possible negative impact on bone mineral density (BMD) by genetically predicted higher levels of SOST, CCDC80, and TIMP4 (Metalloproteinase inhibitor 4). The MR association with BMD was positive for PTX3 (Pentraxin-related protein) and SPP1 (Osteopontin). Genetically predicted higher concentrations of SOST and lower concentrations of SPP1 also conferred a higher risk of falls and lowered grip strength. The genetically determined concentration of nine proteins influenced fat mass, and one influenced lean body mass. INTERPRETATION These data reveal biological links between cardiovascular diseases and fragility fractures. The proteins should be further evaluated as shared targets for developing pharmacological interventions to prevent fractures and cardiovascular disease. FUNDING The study was supported by funding from the Swedish Research Council (https://www.vr.se; grants No. 2015-03257, 2017-00644, 2017-06100, and 2019-01291 to Karl Michaëlsson) and funding from Olle Engkvist Byggmästares stiftelse (SOEB).
Collapse
Affiliation(s)
- Karl Michaëlsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Rui Zheng
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - John A Baron
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Tove Fall
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Alicja Wolk
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonas Höijer
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Carl Brunius
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Eva Warensjö Lemming
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Olga E Titova
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Bodil Svennblad
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susanna C Larsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Melhus
- Clinical Pharmacology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Liisa Byberg
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Hannah L Brooke
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
20
|
Liu Y. Mass spectrometry-based mapping of plasma protein QTLs in children and adolescents. Nat Genet 2025; 57:487-488. [PMID: 39972213 DOI: 10.1038/s41588-025-02088-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Affiliation(s)
- Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT, USA.
- Department of Biomedical Informatics & Data Science, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
21
|
Wang B, Pozarickij A, Mazidi M, Wright N, Yao P, Said S, Iona A, Kartsonaki C, Fry H, Lin K, Chen Y, Du H, Avery D, Schmidt-Valle D, Yu C, Sun D, Lv J, Hill M, Li L, Bennett DA, Collins R, Walters RG, Clarke R, Millwood IY, Chen Z. Comparative studies of 2168 plasma proteins measured by two affinity-based platforms in 4000 Chinese adults. Nat Commun 2025; 16:1869. [PMID: 39984443 PMCID: PMC11845630 DOI: 10.1038/s41467-025-56935-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/27/2025] [Indexed: 02/23/2025] Open
Abstract
Proteomics offers unique insights into human biology and drug development, but few studies have directly compared the utility of different proteomics platforms. We measured plasma levels of 2168 proteins in 3976 Chinese adults using both Olink Explore and SomaScan platforms. The correlation of protein levels between platforms was modest (median rho = 0.29), with protein abundance and data quality parameters being key factors influencing correlation. For 1694 proteins with one-to-one matched reagents, 765 Olink and 513 SomaScan proteins had cis-pQTLs, including 400 with colocalising cis-pQTLs. Moreover, 1096 Olink and 1429 SomaScan proteins were associated with BMI, while 279 and 154 proteins were associated with risk of ischaemic heart disease, respectively. Addition of Olink and SomaScan proteins to conventional risk factors for ischaemic heart disease improved C-statistics from 0.845 to 0.862 (NRI: 12.2%) and 0.863 (NRI: 16.4%), respectively. These results demonstrate the utility of these platforms and could inform the design and interpretation of future studies.
Collapse
Grants
- 82192901, 82192904, 82192900 National Natural Science Foundation of China (National Science Foundation of China)
- CH/1996001/9454 British Heart Foundation (BHF)
- MC-PC-13049, MC-PC-14135 RCUK | Medical Research Council (MRC)
- FS/18/23/33512 British Heart Foundation (BHF)
- C16077/A29186, C500/A16896 Cancer Research UK (CRUK)
- Wellcome Trust
- 212946/Z/18/Z, 202922/Z/16/Z, 104085/Z/14/Z, 088158/Z/09/Z Wellcome Trust (Wellcome)
- The CKB baseline survey and the first re-survey were supported by the Kadoorie Charitable Foundation in Hong Kong. The long-term follow-up and subsequent resurveys have been supported by Wellcome grants to Oxford University (212946/Z/18/Z, 202922/Z/16/Z, 104085/Z/14/Z, 088158/Z/09/Z) and grants from the National Natural Science Foundation of China (82192901, 82192904, 82192900) and from the National Key Research and Development Program of China (2016YFC0900500).The UK Medical Research Council (MC_UU_00017/1, MC_UU_12026/2, MC_U137686851), Cancer Research UK (C16077/A29186, C500/A16896) and British Heart Foundation (CH/1996001/9454), provide core funding to the Clinical Trial Service Unit and Epidemiological Studies Unit, Oxford University for the project. The proteomic assays were supported by BHF (FS/18/23/33512), Novo Nordisk, Olink, SomaScan and NDPH. DNA extraction and genotyping were supported by GlaxoSmithKline and the UK Medical Research Council (MC-PC-13049, MC-PC-14135). Computation used the Oxford Biomedical Research Computing (BMRC) facility, a joint development between the Wellcome Centre for Human Genetics and the Big Data Institute supported by Health Data Research UK and the NIHR Oxford Biomedical Research Centre. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health.
Collapse
Affiliation(s)
- Baihan Wang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Alfred Pozarickij
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Mohsen Mazidi
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Pang Yao
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Saredo Said
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Andri Iona
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Hannah Fry
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Kuang Lin
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Huaidong Du
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel Avery
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dan Schmidt-Valle
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Canqing Yu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dianjianyi Sun
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Jun Lv
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Derrick A Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rory Collins
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Iona Y Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
22
|
Nicholas JC, Katz DH, Tahir UA, Debban CL, Aguet F, Blackwell T, Bowler RP, Broadaway KA, Chen J, Clish CB, Coresh J, Cornell E, Cruz DE, Deo R, Doyle MF, Durda P, Ekunwe L, Floyd JS, Gill D, Guo X, Hoogeveen RC, Johnson C, Lange LA, Li Y, Manning A, Meigs JB, Mi MY, Mychaleckyj JC, Olson NC, Pratte KA, Psaty BM, Reiner AP, Ruan P, Sevilla-Gonzalez M, Shah AM, Sun Q, Tracy RP, Wen J, Wood AC, Wilson JG, Young KL, Yu B, Rooney MR, Manichaikul A, Dubin R, Mohlke KL, Rich SS, Rotter JI, Ganz P, Gerszten RE, Taylor KD, Raffield LM. Cross-Ancestry Comparison of Aptamer and Antibody Proteomics Measures. RESEARCH SQUARE 2025:rs.3.rs-5968391. [PMID: 39989965 PMCID: PMC11844639 DOI: 10.21203/rs.3.rs-5968391/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Measures from affinity-proteomics platforms often correlate poorly, challenging interpretation of protein associations with genetic variants (pQTL) and phenotypes. Here, we examined 2,157 proteins measured on both SomaScan 7k and Olink Explore 3072 across 1,930 participants with genetic similarity to European, African, East Asian, and Admixed American ancestry references. Inter-platform correlation coefficients for these 2,157 proteins followed a bimodal distribution (median r=0.30). Protein measures from each platform were associated with genetic variants (pQTLs), and one-third of the pQTL signals were driven by protein-altering variants (PAVs). We highlight 80 proteins that correlate differently across ancestry groups likely due to differing PAV frequencies by ancestry. Furthermore, adjustment for PAVs with opposite directions of effect by platform improved inter-platform protein measure correlation and resulted in more concordant genetic and phenotypic associations. Hence, PAVs need to be accounted for across ancestries to facilitate platform-concordant and accurate protein measurement.
Collapse
Affiliation(s)
- Jayna C Nicholas
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel H Katz
- Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Usman A Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Catherine L Debban
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | | | | | | | - K Alaine Broadaway
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jingsha Chen
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Clary B Clish
- Metabolomics Platform, Broad Institute, Cambridge, MA, USA
| | - Josef Coresh
- Department of Population Health, Institute for Optimal Aging, New York, NY, USA
| | - Elaine Cornell
- Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Daniel E Cruz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rajat Deo
- Division of Cardiovascular Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Margaret F Doyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Lynette Ekunwe
- University of Mississippi Medical Center, Jackson, MS, USA
| | - James S Floyd
- School of Medicine, University of Washington, Seattle, WA, USA
| | | | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ron C Hoogeveen
- Medicine, Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
| | | | - Leslie A Lange
- School of Medicine, Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alisa Manning
- Broad Institute, Harvard University, Massachusetts General Hospital, Boston, MA, USA
| | - James B Meigs
- Department of Medicine, Division of General Internal Medicine, Broad Institute, Boston, MA, USA
| | - Michael Y Mi
- Department of Medicine, Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Josyf C Mychaleckyj
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Nels C Olson
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | | | - Brucy M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology, University of Washington, Seattle, WA, USA
| | - Alexander P Reiner
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| | | | - Magdalena Sevilla-Gonzalez
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute, Harvard University, Massachusetts General Hospital, Cambridge, MA, USA
| | | | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Jia Wen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexis C Wood
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - James G Wilson
- Deparment of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kristin L Young
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bing Yu
- UT Health, School of Public Health, Houston, TX, USA
| | - Mary R Rooney
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ani Manichaikul
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | | | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen S Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Peter Ganz
- Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Wang Q, Li Y, Yao L, Li H, Zhang L, Wang Y, Li J, Chen T, Chai K, Gao J, Gao J, Su L, Li X. High-affinity ssDNA aptamer and chemiluminescent aptasensor for TIMP-1 detection in human serum. ANAL SCI 2025; 41:119-126. [PMID: 39742443 PMCID: PMC11750923 DOI: 10.1007/s44211-024-00673-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/17/2024] [Indexed: 01/03/2025]
Abstract
TIMP-1 (Tissue Inhibitor of Metalloproteinases-1) is a protein involved in regulating extracellular matrix (ECM) degradation. It is recognized as a significant biomarker for cancer diagnosis. This study aimed to develop and characterize a single-stranded DNA (ssDNA) aptamer targeting human TIMP-1 protein with high affinity and specificity. A magnetic beads-based SELEX process combined with qPCR was used to select aptamers over seven rounds. The enriched ssDNA library was analyzed using high-throughput sequencing to identify candidate sequences, and these sequences were characterized using surface plasmon resonance (SPR) and binding assays to evaluate their affinity and specificity. The selected ssDNA aptamer demonstrated a dissociation equilibrium constant (KD) of 0.41 nM and a very slow off-rate, enabling effective capture of TIMP-1 in serum samples. Furthermore, a chemiluminescent aptasensor was developed for TIMP-1 detection, which exhibited high specificity and a broad linear detection range from 1 to 500 ng/mL in human serum. The developed ssDNA aptamer targeting TIMP-1 shows high affinity and specificity, and the chemiluminescent aptasensor demonstrates promising potential for clinical diagnosis of TIMP-1 levels in human serum.
Collapse
Affiliation(s)
- Qin Wang
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Yanli Li
- The First People's Hospital of Shangqiu, Shangqiu, 476000, China
| | - Lige Yao
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Huiqin Li
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Liuyan Zhang
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Yingjie Wang
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Jiayin Li
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Tian Chen
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Kun Chai
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou, 311200, China
| | - Junli Gao
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou, 311200, China
| | - Junshun Gao
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou, 311200, China
| | - Li Su
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China.
| | - Xueming Li
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou, 311200, China.
| |
Collapse
|
24
|
Reed ER, Chandler KB, Lopez P, Costello CE, Andersen SL, Perls TT, Li M, Bae H, Soerensen M, Monti S, Sebastiani P. Cross-platform proteomics signatures of extreme old age. GeroScience 2025; 47:1199-1220. [PMID: 39048883 PMCID: PMC11872828 DOI: 10.1007/s11357-024-01286-x] [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: 04/10/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
In previous work, we used a SomaLogic platform targeting approximately 5000 proteins to generate a serum protein signature of centenarians that we validated in independent studies that used the same technology. We set here to validate and possibly expand the results by profiling the serum proteome of a subset of individuals included in the original study using liquid chromatography tandem mass spectrometry (LC-MS/MS). Following pre-processing, the LC-MS/MS data provided quantification of 398 proteins, with only 266 proteins shared by both platforms. At 1% FDR statistical significance threshold, the analysis of LC-MS/MS data detected 44 proteins associated with extreme old age, including 23 of the original analysis. To identify proteins for which associations between expression and extreme-old age were conserved across platforms, we performed inter-study conservation testing of the 266 proteins quantified by both platforms using a method that accounts for the correlation between the results. From these tests, a total of 80 proteins reached 5% FDR statistical significance, and 26 of these proteins had concordant pattern of gene expression in whole blood generated in an independent set. This signature of 80 proteins points to blood coagulation, IGF signaling, extracellular matrix (ECM) organization, and complement cascade as important pathways whose protein level changes provide evidence for age-related adjustments that distinguish centenarians from younger individuals. The comparison with blood transcriptomics also highlights a possible role for neutrophil degranulation in aging.
Collapse
Affiliation(s)
- Eric R Reed
- Data Intensive Study Center, Tufts University, Boston, MA, USA
| | - Kevin B Chandler
- Center for Biomedical Mass Spectrometry, Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL, USA
| | - Prisma Lopez
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| | - Catherine E Costello
- Center for Biomedical Mass Spectrometry, Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Stacy L Andersen
- Geriatric Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Thomas T Perls
- Geriatric Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Mengze Li
- Division of Computational Biomedicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Harold Bae
- Biostatistics Program, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Mette Soerensen
- Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Stefano Monti
- Division of Computational Biomedicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Paola Sebastiani
- Data Intensive Study Center, Tufts University, Boston, MA, USA.
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA.
- Department of Medicine, School of Medicine, Tufts University, Boston, MA, USA.
| |
Collapse
|
25
|
Sim MA, Doecke JD, Liew OW, Wong LL, Tan ESJ, Chan SP, Chong JRF, Cai Y, Hilal S, Venketasubramanian N, Tan BY, Alzheimer's Disease Neuroimaging Initiative, Lai MKP, Choi H, Masters CL, Richards AM, Chen CLH. Plasma proteomics for cognitive decline and dementia-A Southeast Asian cohort study. Alzheimers Dement 2025; 21:e14577. [PMID: 39998981 PMCID: PMC11854348 DOI: 10.1002/alz.14577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/27/2024] [Accepted: 01/12/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION The prognostic utility of plasma proteomics for cognitive decline and dementia in a Southeast Asian population characterized by high cerebrovascular disease (CeVD) burden is underexplored. METHODS We examined this in a Singaporean memory clinic cohort of 528 subjects (n = 300, CeVD; n = 167, incident cognitive decline) followed-up for 4 years. RESULTS Of 1441 plasma proteins surveyed, a 12-protein signature significantly predicted cognitive decline (q-value < .05). Sixteen diverse biological processes were implicated in cognitive decline. Ten proteins independently predicted incident dementia (q-value < .05). A unified prediction model combining plasma proteins with clinical risk factors increased the area under the curve for outcome prediction from 0.62 to 0.85. External validation in the cerebrospinal fluid proteome of an independent Caucasian cohort replicated four of the significantly predictive plasma markers for cognitive decline namely: GFAP, NEFL, AREG, and PPY. DISCUSSION The prognostic proteins prioritized in our study provide robust signals in two different biological matrices, representing potential mechanistic targets for dementia and cognitive decline. HIGHLIGHTS A total of 1441 plasma proteins were profiled in a Singaporean memory clinic cohort. We report prognostic plasma protein signatures for cognitive decline and dementia. External validation was performed in the cerebrospinal fluid proteome of a Caucasian cohort. A concordant proteomic signature was identified across both biofluids and cohorts. Further studies are needed to explore the therapeutic implications of these proteins for dementia.
Collapse
Affiliation(s)
- Ming Ann Sim
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of AnesthesiaNational University Health SystemSingaporeSingapore
| | - James D. Doecke
- Australian E‐Health Research CentreCSIROHerstonQueenslandAustralia
| | - Oi Wah Liew
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Lee Lee Wong
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Eugene S. J. Tan
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of CardiologyNational University Heart CentreSingaporeSingapore
| | - Siew Pang Chan
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Joyce R. F. Chong
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| | - Yuan Cai
- Department of Medicine and Therapeutics, Faculty of MedicineDivision of NeurologyThe Chinese University of Hong KongMa Liu ShuiHong Kong
| | - Saima Hilal
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemSingaporeSingapore
| | | | - Boon Yeow Tan
- Department of MedicineSt Luke's HospitalSingaporeSingapore
| | | | - Mitchell K. P Lai
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| | - Hyungwon Choi
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
- Department of MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Singapore Lipidomics IncubatorLife Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Arthur Mark Richards
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
- Christchurch Heart InstituteUniversity of OtagoDunedinNew Zealand
| | - Christopher L. H. Chen
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| |
Collapse
|
26
|
Guo T, Steen JA, Mann M. Mass-spectrometry-based proteomics: from single cells to clinical applications. Nature 2025; 638:901-911. [PMID: 40011722 DOI: 10.1038/s41586-025-08584-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 01/02/2025] [Indexed: 02/28/2025]
Abstract
Mass-spectrometry (MS)-based proteomics has evolved into a powerful tool for comprehensively analysing biological systems. Recent technological advances have markedly increased sensitivity, enabling single-cell proteomics and spatial profiling of tissues. Simultaneously, improvements in throughput and robustness are facilitating clinical applications. In this Review, we present the latest developments in proteomics technology, including novel sample-preparation methods, advanced instrumentation and innovative data-acquisition strategies. We explore how these advances drive progress in key areas such as protein-protein interactions, post-translational modifications and structural proteomics. Integrating artificial intelligence into the proteomics workflow accelerates data analysis and biological interpretation. We discuss the application of proteomics to single-cell analysis and spatial profiling, which can provide unprecedented insights into cellular heterogeneity and tissue architecture. Finally, we examine the transition of proteomics from basic research to clinical practice, including biomarker discovery in body fluids and the promise and challenges of implementing proteomics-based diagnostics. This Review provides a broad and high-level overview of the current state of proteomics and its potential to revolutionize our understanding of biology and transform medical practice.
Collapse
Affiliation(s)
- Tiannan Guo
- State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, China.
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China.
| | - Judith A Steen
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
27
|
Pirola CJ, Fernández Gianotti T, Sookoian S. The Proteomics of MASLD Progression: Insights From Functional Analysis to Drive the Development of New Therapeutic Solutions. Aliment Pharmacol Ther 2025; 61:614-627. [PMID: 39744897 DOI: 10.1111/apt.18468] [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: 10/15/2024] [Revised: 11/22/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading chronic liver disease worldwide, with alarming prevalence reaching epidemic proportions. AIMS AND METHODS The objective of this study is to provide a comprehensive review of the latest blood proteomics studies on MASLD and metabolic dysfunction-associated steatohepatitis (MASH), with emphasis on fibrosis. Furthermore, our objective is to conduct an analysis of protein pathways and interactions by integrating proteomics data using functional enrichment analysis of the deregulated proteins. RESULTS Notwithstanding the considerable discrepancies in the methodology and the number of proteins examined in the circulation, the analysis reveals a consistent pattern among the list of proteins that are decreased or increased in the blood of the affected patients. The relevant biological processes (BP) associated with down- and upregulated proteins are high-density lipoprotein remodelling and complement activation, respectively. The protein families identified include not only those expected to be involved in the immune system and cell adhesion and migration but also ligands of glycoproteins expressed in cells that have been subjected to stress and proteins containing the Sushi domain. CONCLUSIONS The application of cutting-edge methodologies to investigate the blood proteome in MASH is yielding insights that facilitate the elucidation of disease mechanisms and the identification of optimal noninvasive biomarkers. However, several challenges remain to be addressed in future research, including the generalisation of results on a global scale, the optimisation of analytical technologies and the implementation of large longitudinal studies to gain insights into the molecular mechanisms that underpin the development of advanced disease.
Collapse
Affiliation(s)
- Carlos José Pirola
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Systems Biology of Complex Diseases, Translational Research in Health Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| | - Tomas Fernández Gianotti
- Systems Biology of Complex Diseases, Translational Research in Health Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| | - Silvia Sookoian
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Clinical and Molecular Hepatology, Translational Research in Health Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| |
Collapse
|
28
|
Yu K, Li W, Long W, Li Y, Li Y, Liao H, Liu J. Proteome-wide mendelian randomization identifies causal plasma proteins in interstitial lung disease. Sci Rep 2025; 15:2293. [PMID: 39824903 PMCID: PMC11748740 DOI: 10.1038/s41598-025-85338-y] [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: 04/09/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
Interstitial lung disease (ILD) has shown limited treatment advancements, with minimal exploration of circulating protein biomarkers causally linked to ILD and its subtypes beyond idiopathic pulmonary fibrosis (IPF). In this study, we aimed to identify potential drug targets and circulating protein biomarkers for ILD and its subtypes. We utilized the most recent large-scale plasma protein quantitative trait loci (pQTL) data detected from the antibody-based method and ILD and its subtypes' GWAS data from the updated FinnGen database for Mendelian randomization analysis. To enhance the reliability of causal associations, we conducted external validation and sensitivity analyses, including Bayesian colocalization and bidirectional Mendelian randomization analysis. Our study identified eight plasma proteins genetically associated with ILD or its subtypes. Among these, three proteins-CDH15 (Cadherin-15), LTBR (Lymphotoxin-beta receptor), and ADAM15 (A disintegrin and metalloproteinase 15)-emerged as priority biomarkers and potential therapeutic targets, demonstrating more reliable associations by passing a series of sensitivity analyses compared to the others. Based on these findings, we propose for the first time that CDH15, ADAM15, and LTBR hold promise as novel potential circulating protein biomarkers and therapeutic targets for the diagnosis and treatment of ILD, IPF, and sarcoidosis, respectively, especially ADAM15, and these findings have the potential to provide new perspectives for advancing the research on the heterogeneity of ILD.
Collapse
Affiliation(s)
- Kunrong Yu
- Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Wanying Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Wenjie Long
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yijia Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yanting Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Huili Liao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Jianhong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
29
|
Castro C, Delwarde C, Shi Y, Roh J. Geroscience in heart failure: the search for therapeutic targets in the shared pathobiology of human aging and heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2025; 5:10.20517/jca.2024.15. [PMID: 40297496 PMCID: PMC12036312 DOI: 10.20517/jca.2024.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Age is a major risk factor for heart failure, but one that has been historically viewed as non-modifiable. Emerging evidence suggests that the biology of aging is malleable, and can potentially be intervened upon to treat age-associated chronic diseases, such as heart failure. While aging biology represents a new frontier for therapeutic target discovery in heart failure, the challenges of translating Geroscience research to the clinic are multifold. In this review, we propose a strategy that prioritizes initial target discovery in human biology. We review the rationale for starting with human omics, which has generated important insights into the shared (patho)biology of human aging and heart failure. We then discuss how this knowledge can be leveraged to identify the mechanisms of aging biology most relevant to heart failure. Lastly, we provide examples of how this human-first Geroscience approach, when paired with rigorous functional assessments in preclinical models, is leading to early-stage clinical development of gerotherapeutic approaches for heart failure.
Collapse
Affiliation(s)
- Claire Castro
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Constance Delwarde
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Yanxi Shi
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jason Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
30
|
Smits HM, Delemarre EM, Pandit A, Schoneveld AH, Oldenburg B, van Wijk F, Nierkens S, Drylewicz J. The BAMBOO method for correcting batch effects in high throughput proximity extension assays for proteomic studies. Sci Rep 2025; 15:1498. [PMID: 39789032 PMCID: PMC11717925 DOI: 10.1038/s41598-024-84320-4] [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: 05/15/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
The proximity extension assay (PEA) enables large-scale proteomic investigations across numerous proteins and samples. However, discrepancies between measurements, known as batch-effects, potentially skew downstream statistical analyses and increase the risks of false discoveries. While implementing bridging controls (BCs) on each plate has been proposed to mitigate these effects, a clear method for utilizing this strategy remains elusive. Here, we characterized batch effects in PEA proteomics and identified three types: protein-specific, sample-specific, and plate-wide. We developed a robust regression-based method called BAMBOO (Batch Adjustments using Bridging cOntrOls) to correct them. Simulations comparing BAMBOO with established correction techniques (median centering, median of the difference (MOD), and ComBat) revealed that median centering and ComBat were significantly impacted by outliers within the BCs, whereas BAMBOO and MOD were more robust when no plate-wide effects were introduced. Optimal batch correction was achieved with 10-12 BCs. We validated the simulation results using experimental data and found that BAMBOO and MOD had a reduced incidence of false discoveries compared to alternative methods. Our findings emphasize the prevalence of batch effects in PEA proteomic studies and advocate for BAMBOO as a robust and effective tool to enhance the reliability of large-scale analyses in the proteomic field.
Collapse
Affiliation(s)
- H M Smits
- Center for Translational Immunology, University Medical Center Utrecht, KC 02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands
| | - E M Delemarre
- Center for Translational Immunology, University Medical Center Utrecht, KC 02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands
| | - A Pandit
- Center for Translational Immunology, University Medical Center Utrecht, KC 02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands
| | - A H Schoneveld
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - B Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, KC 02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands
| | - S Nierkens
- Center for Translational Immunology, University Medical Center Utrecht, KC 02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - J Drylewicz
- Center for Translational Immunology, University Medical Center Utrecht, KC 02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands.
| |
Collapse
|
31
|
Lim CG, Gradinariu V, Liang Y, Rebholz CM, Talegawkar S, Temprosa M, Min YI, Sim X, Wilson JG, van Dam RM. Proteomic analysis identifies novel biological pathways that may link dietary quality to type 2 diabetes risk: evidence from African American and Asian cohorts. Am J Clin Nutr 2025; 121:100-110. [PMID: 39566683 PMCID: PMC11747191 DOI: 10.1016/j.ajcnut.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Diet affects the development of chronic diseases such as type 2 diabetes, but the underlying biological mechanisms are only partly understood. OBJECTIVES This study aimed to identify proteomic markers of the Alternative Healthy Eating Index (AHEI) and the Dietary Approaches to Stop Hypertension (DASH) diet and their association with type 2 diabetes risk. METHODS We examined the associations between the AHEI and DASH diet quality scores and 1317 plasma proteins in African American participants of the Jackson Heart Study (JHS, n = 1878). These findings were validated in a Singapore Multi-Ethnic Cohort (n = 2395) and examined in relation to type 2 diabetes incidence (n = 539 cases). We adjusted for multiple testing by using false discovery rate-adjusted q values. RESULTS We identified 13 proteins consistently associated with the AHEI or DASH scores with the strongest associations for the AHEI score and epidermal growth factor receptor (β:0.089; SE: 0.017; q < 0.001) and for the DASH score and tissue factor (β: -0.114; SE: 0.022; q < 0.001). Most of these proteins were related to inflammation, thrombosis, adipogenesis, and glucose metabolism. Concentrations of myeloperoxidase, epidermal growth factor receptor, hepatocyte growth factor receptor, coagulation factor Xa, contactin 4, kynureninase, neurogenic locus notch homolog protein 1, and vesicular integral-membrane protein VIP36 were associated with the risk of type 2 diabetes in the Asian cohort. The diabetes odds ratio for a 2-fold higher protein abundance concentration ranged from 0.03 (95% CI: 0.01, 0.08) for neurogenic locus notch homolog protein 1 to 3.04 (95% CI: 2.13, 4.33) for kynureninase. Furthermore, genetic markers for myeloperoxidase and hepatocyte growth factor receptor were significantly associated with diabetes risk. CONCLUSIONS Our study across geographically and ethnically diverse populations identified robust protein biomarkers for healthy dietary patterns. Furthermore, our findings suggest novel biological mechanisms linking dietary patterns with type 2 diabetes development.
Collapse
Affiliation(s)
- Charlie Gy Lim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore.
| | - Vlad Gradinariu
- Departments of Exercise and Nutrition Sciences and Epidemiology, Milken Institute School of Public Health, The George Washington University, Newark, Washington, DC, United States
| | - Yujian Liang
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Sameera Talegawkar
- Biostatistics Center and Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Rockville, MD, United States
| | - Marinella Temprosa
- Biostatistics Center and Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Rockville, MD, United States
| | - Yuan-I Min
- Department of Medicine, University of Mississippi Medical Center, Jackson Medical Mall, Jackson, MS, United States
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - James G Wilson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore; Departments of Exercise and Nutrition Sciences and Epidemiology, Milken Institute School of Public Health, The George Washington University, Newark, Washington, DC, United States.
| |
Collapse
|
32
|
Tahir UA. Pathways to Precision Medicine in Hypertrophic Cardiomyopathy: Opportunities and Challenges in Plasma Proteomics. Circ Heart Fail 2025; 18:e012593. [PMID: 39697179 PMCID: PMC11891881 DOI: 10.1161/circheartfailure.124.012593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Affiliation(s)
- Usman A Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
33
|
Strelnikova PA, Zakharova NV, Kononikhin AS, Bugrova AE, Indeykina MI, Mitkevich VA, Makarov AA, Nikolaev EN. Blood plasma proteomic markers of Alzheimer's disease, current status and application prospects. Expert Rev Proteomics 2025; 22:11-18. [PMID: 39764651 DOI: 10.1080/14789450.2025.2450804] [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: 11/08/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025]
Abstract
INTRODUCTION Identifying early risks of developing Alzheimer's disease (AD) is a major challenge as the number of patients with AD steadily increases and requires innovative solutions. Current molecular diagnostic modalities, such as cerebrospinal fluid (CSF) testing and positron emission tomography (PET) imaging, exhibit limitations in their applicability for large-scale screening. In recent years, there has been a marked shift toward the development of blood plasma-based diagnostic tests, which offer a more accessible and clinically viable alternative for widespread use. Furthermore, advances in large-scale proteomics technologies have boosted an interest in identifying novel biomarkers and developing panels of AD-associated proteins. AREAS COVERED This review mainly examines the results of recent searches for proteomic markers of AD in blood plasma (from 2022-2024 PubMed), focuses on some aspects for special attention in further studies, and discusses the prospects for their further application. EXPERT OPINION Recent advances in AD plasma/serum proteomic studies are largely driven using novel Olink/PEA and SomaScan/aptamer technologies, which complement the 'gold standard' of MS-based quantitative proteomics (MRM/SRM), and particularly expand the capabilities for studying low-abundant proteins.
Collapse
Affiliation(s)
- Polina A Strelnikova
- Project Center of Omics Technologies and Advanced Mass Spectrometry, Skolkovo Institute of Science and Technology, Moscow Russian Federation
- Emanuel Institute of Biochemical Physics, Russian Academy of Science, Moscow Russian Federation
| | - Natalia V Zakharova
- Project Center of Omics Technologies and Advanced Mass Spectrometry, Skolkovo Institute of Science and Technology, Moscow Russian Federation
- Emanuel Institute of Biochemical Physics, Russian Academy of Science, Moscow Russian Federation
| | - Alexey S Kononikhin
- Project Center of Omics Technologies and Advanced Mass Spectrometry, Skolkovo Institute of Science and Technology, Moscow Russian Federation
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow Russian Federation
| | - Anna E Bugrova
- Project Center of Omics Technologies and Advanced Mass Spectrometry, Skolkovo Institute of Science and Technology, Moscow Russian Federation
- Emanuel Institute of Biochemical Physics, Russian Academy of Science, Moscow Russian Federation
| | - Maria I Indeykina
- Project Center of Omics Technologies and Advanced Mass Spectrometry, Skolkovo Institute of Science and Technology, Moscow Russian Federation
- Emanuel Institute of Biochemical Physics, Russian Academy of Science, Moscow Russian Federation
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow Russian Federation
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow Russian Federation
| | - Evgeny N Nikolaev
- Project Center of Omics Technologies and Advanced Mass Spectrometry, Skolkovo Institute of Science and Technology, Moscow Russian Federation
| |
Collapse
|
34
|
Puerta R, Cano A, García-González P, García-Gutiérrez F, Capdevila M, de Rojas I, Olivé C, Blázquez-Folch J, Sotolongo-Grau O, Miguel A, Montrreal L, Martino-Adami P, Khan A, Orellana A, Sung YJ, Frikke-Schmidt R, Marchant N, Lambert JC, Rosende-Roca M, Alegret M, Fernández MV, Marquié M, Valero S, Tárraga L, Cruchaga C, Ramírez A, Boada M, Smets B, Cabrera-Socorro A, Ruiz A. Head-to-Head Comparison of Aptamer- and Antibody-Based Proteomic Platforms in Human Cerebrospinal Fluid Samples from a Real-World Memory Clinic Cohort. Int J Mol Sci 2024; 26:286. [PMID: 39796148 PMCID: PMC11720409 DOI: 10.3390/ijms26010286] [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: 10/29/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
High-throughput proteomic platforms are crucial to identify novel Alzheimer's disease (AD) biomarkers and pathways. In this study, we evaluated the reproducibility and reliability of aptamer-based (SomaScan® 7k) and antibody-based (Olink® Explore 3k) proteomic platforms in cerebrospinal fluid (CSF) samples from the Ace Alzheimer Center Barcelona real-world cohort. Intra- and inter-platform reproducibility were evaluated through correlations between two independent SomaScan® assays analyzing the same samples, and between SomaScan® and Olink® results. Association analyses were performed between proteomic measures, CSF biological traits, sample demographics, and AD endophenotypes. Our 12-category metric of reproducibility combining correlation analyses identified 2428 highly reproducible SomaScan CSF measures, with over 600 proteins well reproduced on another proteomic platform. The association analyses among AD clinical phenotypes revealed that the significant associations mainly involved reproducible proteins. The validation of reproducibility in these novel proteomics platforms, measured using this scarce biomaterial, is essential for accurate analysis and proper interpretation of innovative results. This classification metric could enhance confidence in multiplexed proteomic platforms and improve the design of future panels.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- PhD Program in Biotecnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Fernando García-Gutiérrez
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Maria Capdevila
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Clàudia Olivé
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Josep Blázquez-Folch
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Oscar Sotolongo-Grau
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Andrea Miguel
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Pamela Martino-Adami
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.M.-A.); (A.R.)
| | - Asif Khan
- Janssen Pharmaceutica NV, a Johnson & Johnson Company, 2340 Beerse, Belgium; (A.K.); (B.S.); (A.C.-S.)
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Yun Ju Sung
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63108, USA; (Y.J.S.); (C.C.)
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Natalie Marchant
- Division of Psychiatry, University College London, London W1T 7NK, UK;
| | - Jean Charles Lambert
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Université de Lille, F-59000 Lille, France;
- Institut Pasteur de Lille, Inserm U1167, CHU de Lille, LabEx DISTALZ, Université de Lille, F-59000 Lille, France
| | - Maitée Rosende-Roca
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Montserrat Alegret
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Maria Victoria Fernández
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Carlos Cruchaga
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63108, USA; (Y.J.S.); (C.C.)
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Alfredo Ramírez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.M.-A.); (A.R.)
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, Medical Faculty, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Psychiatry and Glenn, Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX 78229, USA
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Bart Smets
- Janssen Pharmaceutica NV, a Johnson & Johnson Company, 2340 Beerse, Belgium; (A.K.); (B.S.); (A.C.-S.)
| | - Alfredo Cabrera-Socorro
- Janssen Pharmaceutica NV, a Johnson & Johnson Company, 2340 Beerse, Belgium; (A.K.); (B.S.); (A.C.-S.)
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX 77204, USA
| |
Collapse
|
35
|
Shitole SG, Heckbert SR, Marcus GM, Shah SJ, Sotoodehnia N, Walston JD, Reiner AP, Tracy RP, Psaty BM, Kizer JR. Assessment of Inflammatory Biomarkers and Incident Atrial Fibrillation in Older Adults. J Am Heart Assoc 2024; 13:e035710. [PMID: 39644101 PMCID: PMC11935547 DOI: 10.1161/jaha.124.035710] [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: 04/17/2024] [Accepted: 10/24/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Available evidence supports the importance of inflammation in atrial fibrillation (AF) pathogenesis, yet general anti-inflammatory therapies have failed to show benefit for prevention of the arrhythmia. Better understanding of the specific inflammatory pathways involved is necessary to advance therapeutics. METHODS AND RESULTS We evaluated 9 circulating markers of inflammation measured by immunoassays and incidence of AF in a population-based older cohort. Biomarkers included measures of general inflammation and the NLR (nucleotide-binding oligomerization domain-like receptor) family pyrin domain containing 3 inflammasome, TNF-α (tumor necrosis factor α), monocyte activation markers, and sIL-2 (soluble interleukin-2). Among 5726 participants (median age 72 years), 1836 developed AF over median follow-up of 11.5 years. After adjustment for conventional risk factors, 5 biomarkers were positively associated with incident AF: IL-6 (interleukin-6), hazard ratio (HR), 1.14 (95% CI, 1.07-1.21); hs-CRP (high-sensitivity C-reactive protein), HR, 1.05 (95% CI, 1.01-1.09); white blood cell count, HR, 1.18 (95% CI, 1.04-1.35); sTNFR1 (soluble TNF receptor 1), HR, 1.21 (95% CI, 1.05-1.39); and sIL-2Rα (sIL-2 receptor α), HR, 1.16 (95% CI, 1.05-1.29) (all per doubling of biomarker). sCD14, sCD163, IL-18, and IL-1 receptor antagonist showed no association with AF. Upon concurrent adjustment for all biomarkers, only IL-6 remained significantly associated with the arrhythmia, HR, 1.17 (95% CI, 1.07-1.26). CONCLUSIONS Among older adults, IL-6, hs-CRP, white blood cell count, sTNFR1, and sIL-2Rα were positively associated with incident AF, but only IL-6 retained significance on concurrent adjustment. These findings newly document associations for sTNFR1 and sIL-2Rα and lend support to a preeminent role for IL-6 in development of this arrhythmia. The efficacy of IL-6 blockade for AF prevention awaits completion of appropriate clinical trials.
Collapse
Affiliation(s)
- Sanyog G. Shitole
- Cardiology SectionSan Francisco Veterans Affairs Health Care SystemSan FranciscoCAUSA
- Department of MedicineUniversity of California San FranciscoSan FranciscoCAUSA
- Department of MedicineAlbert Einstein College of MedicineBronxNYUSA
| | | | - Gregory M. Marcus
- Department of MedicineUniversity of California San FranciscoSan FranciscoCAUSA
| | - Sanjiv J. Shah
- Department of MedicineNorthwestern UniversityChicagoILUSA
| | - Nona Sotoodehnia
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
- Department of MedicineUniversity of WashingtonSeattleWAUSA
| | | | | | - Russell P. Tracy
- Department of Pathology and Laboratory MedicineThe University of VermontBurlingtonVTUSA
| | - Bruce M. Psaty
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
- Department of Health Systems and Population HealthUniversity of WashingtonSeattleWAUSA
| | - Jorge R. Kizer
- Cardiology SectionSan Francisco Veterans Affairs Health Care SystemSan FranciscoCAUSA
- Department of MedicineUniversity of California San FranciscoSan FranciscoCAUSA
- Department of Epidemiology & BiostatisticsUniversity of California San FranciscoSan FranciscoCAUSA
| |
Collapse
|
36
|
Geyer PE, Hornburg D, Pernemalm M, Hauck SM, Palaniappan KK, Albrecht V, Dagley LF, Moritz RL, Yu X, Edfors F, Vandenbrouck Y, Mueller-Reif JB, Sun Z, Brun V, Ahadi S, Omenn GS, Deutsch EW, Schwenk JM. The Circulating Proteome─Technological Developments, Current Challenges, and Future Trends. J Proteome Res 2024; 23:5279-5295. [PMID: 39479990 PMCID: PMC11629384 DOI: 10.1021/acs.jproteome.4c00586] [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: 07/09/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/02/2024]
Abstract
Recent improvements in proteomics technologies have fundamentally altered our capacities to characterize human biology. There is an ever-growing interest in using these novel methods for studying the circulating proteome, as blood offers an accessible window into human health. However, every methodological innovation and analytical progress calls for reassessing our existing approaches and routines to ensure that the new data will add value to the greater biomedical research community and avoid previous errors. As representatives of HUPO's Human Plasma Proteome Project (HPPP), we present our 2024 survey of the current progress in our community, including the latest build of the Human Plasma Proteome PeptideAtlas that now comprises 4608 proteins detected in 113 data sets. We then discuss the updates of established proteomics methods, emerging technologies, and investigations of proteoforms, protein networks, extracellualr vesicles, circulating antibodies and microsamples. Finally, we provide a prospective view of using the current and emerging proteomics tools in studies of circulating proteins.
Collapse
Affiliation(s)
- Philipp E. Geyer
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Daniel Hornburg
- Seer,
Inc., Redwood City, California 94065, United States
- Bruker
Scientific, San Jose, California 95134, United States
| | - Maria Pernemalm
- Department
of Oncology and Pathology/Science for Life Laboratory, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Stefanie M. Hauck
- Metabolomics
and Proteomics Core, Helmholtz Zentrum München
GmbH, German Research Center for Environmental Health, 85764 Oberschleissheim,
Munich, Germany
| | | | - Vincent Albrecht
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Laura F. Dagley
- The
Walter and Eliza Hall Institute for Medical Research, Parkville, VIC 3052, Australia
- Department
of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Robert L. Moritz
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Xiaobo Yu
- State
Key Laboratory of Medical Proteomics, Beijing
Proteome Research Center, National Center for Protein Sciences-Beijing
(PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Fredrik Edfors
- Science
for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 17121 Solna, Sweden
| | | | - Johannes B. Mueller-Reif
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Zhi Sun
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Virginie Brun
- Université Grenoble
Alpes, CEA, Leti, Clinatec, Inserm UA13
BGE, CNRS FR2048, Grenoble, France
| | - Sara Ahadi
- Alkahest, Inc., Suite
D San Carlos, California 94070, United States
| | - Gilbert S. Omenn
- Institute
for Systems Biology, Seattle, Washington 98109, United States
- Departments
of Computational Medicine & Bioinformatics, Internal Medicine,
Human Genetics and Environmental Health, University of Michigan, Ann Arbor, Michigan 48109-2218, United States
| | - Eric W. Deutsch
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Jochen M. Schwenk
- Science
for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 17121 Solna, Sweden
| |
Collapse
|
37
|
Ma W, Tang W, Kwok JS, Tong AH, Lo CW, Chu AT, Chung BH, Hong Kong Genome Project. A review on trends in development and translation of omics signatures in cancer. Comput Struct Biotechnol J 2024; 23:954-971. [PMID: 38385061 PMCID: PMC10879706 DOI: 10.1016/j.csbj.2024.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
The field of cancer genomics and transcriptomics has evolved from targeted profiling to swift sequencing of individual tumor genome and transcriptome. The steady growth in genome, epigenome, and transcriptome datasets on a genome-wide scale has significantly increased our capability in capturing signatures that represent both the intrinsic and extrinsic biological features of tumors. These biological differences can help in precise molecular subtyping of cancer, predicting tumor progression, metastatic potential, and resistance to therapeutic agents. In this review, we summarized the current development of genomic, methylomic, transcriptomic, proteomic and metabolic signatures in the field of cancer research and highlighted their potentials in clinical applications to improve diagnosis, prognosis, and treatment decision in cancer patients.
Collapse
Affiliation(s)
- Wei Ma
- Hong Kong Genome Institute, Hong Kong, China
| | - Wenshu Tang
- Hong Kong Genome Institute, Hong Kong, China
| | | | | | | | | | - Brian H.Y. Chung
- Hong Kong Genome Institute, Hong Kong, China
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hong Kong Genome Project
- Hong Kong Genome Institute, Hong Kong, China
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
38
|
Albrecht V, Müller-Reif J, Nordmann TM, Mund A, Schweizer L, Geyer PE, Niu L, Wang J, Post F, Oeller M, Metousis A, Bach Nielsen A, Steger M, Wewer Albrechtsen NJ, Mann M. Bridging the Gap From Proteomics Technology to Clinical Application: Highlights From the 68th Benzon Foundation Symposium. Mol Cell Proteomics 2024; 23:100877. [PMID: 39522756 DOI: 10.1016/j.mcpro.2024.100877] [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: 10/03/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
The 68th Benzon Foundation Symposium brought together leading experts to explore the integration of mass spectrometry-based proteomics and artificial intelligence to revolutionize personalized medicine. This report highlights key discussions on recent technological advances in mass spectrometry-based proteomics, including improvements in sensitivity, throughput, and data analysis. Particular emphasis was placed on plasma proteomics and its potential for biomarker discovery across various diseases. The symposium addressed critical challenges in translating proteomic discoveries to clinical practice, including standardization, regulatory considerations, and the need for robust "business cases" to motivate adoption. Promising applications were presented in areas such as cancer diagnostics, neurodegenerative diseases, and cardiovascular health. The integration of proteomics with other omics technologies and imaging methods was explored, showcasing the power of multimodal approaches in understanding complex biological systems. Artificial intelligence emerged as a crucial tool for the acquisition of large-scale proteomic datasets, extracting meaningful insights, and enhancing clinical decision-making. By fostering dialog between academic researchers, industry leaders in proteomics technology, and clinicians, the symposium illuminated potential pathways for proteomics to transform personalized medicine, advancing the cause of more precise diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Vincent Albrecht
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Johannes Müller-Reif
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Thierry M Nordmann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Andreas Mund
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; BioInnovation Institute, OmicVision Biosciences, Copenhagen, Denmark
| | - Lisa Schweizer
- BioInnovation Institute, OmicVision Biosciences, Copenhagen, Denmark
| | - Philipp E Geyer
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany; ions.bio GmbH, Planegg, Germany
| | - Lili Niu
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Computational Biomarker Discovery, Novo Nordisk, Copenhagen, Denmark
| | - Juanjuan Wang
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frederik Post
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marc Oeller
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Andreas Metousis
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Annelaura Bach Nielsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Clinical Biochemistry, University Hospital Copenhagen - Bispebjerg, Copenhagen, Copenhagen, Denmark
| | - Medini Steger
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Nicolai J Wewer Albrechtsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Clinical Biochemistry, University Hospital Copenhagen - Bispebjerg, Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany; NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
39
|
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
|
40
|
Walker KA, An Y, Moghekar A, Moaddel R, Duggan MR, Peng Z, Tian Q, Pilling LC, Drouin SM, Espeland MA, Rapp SR, Hayden KM, Shadyab AH, Casanova R, Thambisetty M, Rapp PR, Kapogiannis D, Ferrucci L, Resnick SM. Proteomic analysis of APOEε4 carriers implicates lipid metabolism, complement and lymphocyte signaling in cognitive resilience. Mol Neurodegener 2024; 19:81. [PMID: 39482741 PMCID: PMC11526661 DOI: 10.1186/s13024-024-00772-2] [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: 03/30/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late onset Alzheimer's disease (AD). This case-cohort study used targeted plasma biomarkers and large-scale proteomics to examine the biological mechanisms that allow some APOEε4 carriers to maintain normal cognitive functioning in older adulthood. METHODS APOEε4 carriers and APOEε3 homozygotes enrolled in the Women's Health Initiative Memory Study (WHIMS) from 1996 to 1999 were classified as resilient if they remained cognitively unimpaired beyond age 80, and as non-resilient if they developed cognitive impairment before or at age 80. AD pathology (Aß42/40) and neurodegeneration (NfL, tau) biomarkers, as well as 1007 proteins (Olink) were quantified in blood collected at study enrollment (on average 14 years prior) when participants were cognitively normal. We identified plasma proteins that distinguished between resilient and non-resilient APOEε4 carriers, examined whether these associations generalized to APOEε3 homozygotes, and replicated these findings in the UK Biobank. RESULTS A total of 1610 participants were included (baseline age: 71.3 [3.8 SD] years; all White; 42% APOEε4 carriers). Compared to resilient APOEε4 carriers, non-resilient APOEε4 carriers had lower Aß42/40/tau ratio and greater NfL at baseline. Proteomic analyses identified four proteins differentially expressed between resilient and non-resilient APOEε4 carriers at an FDR-corrected P < 0.05. While one of the candidate proteins, a marker of neuronal injury (NfL), also distinguished resilient from non-resilient APOEε3 homozygotes, the other three proteins, known to be involved in lipid metabolism (ANGPTL4) and immune signaling (PTX3, NCR1), only predicted resilient vs. non-resilient status among APOEε4 carriers (protein*genotype interaction-P < 0.05). Three of these four proteins also predicted 14-year dementia risk among APOEε4 carriers in the UK Biobank validation sample (N = 9420). While the candidate proteins showed little to no association with targeted biomarkers of AD pathology, protein network and enrichment analyses suggested that natural killer (NK) cell and T lymphocyte signaling (via PKC-θ) distinguished resilient from non-resilient APOEε4 carriers. CONCLUSIONS We identified and replicated a plasma proteomic signature associated with cognitive resilience among APOEε4 carriers. These proteins implicate specific immune processes in the preservation of cognitive status despite elevated genetic risk for AD. Future studies in diverse cohorts will be needed to assess the generalizability of these results.
Collapse
Affiliation(s)
- Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA.
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruin Moaddel
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Qu Tian
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Luke C Pilling
- Department of Clinical & Biomedical Sciences, Faculty of Health & Life Science, University of Exeter, Exeter, UK
| | - Shannon M Drouin
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Mark A Espeland
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Stephen R Rapp
- Department of Psychiatry & Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Social Science & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kathleen M Hayden
- Department of Social Science & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Aladdin H Shadyab
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, and Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
| | - Ramon Casanova
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Peter R Rapp
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
41
|
van Vugt M, Finan C, Chopade S, Providencia R, Bezzina CR, Asselbergs FW, van Setten J, Schmidt AF. Integrating metabolomics and proteomics to identify novel drug targets for heart failure and atrial fibrillation. Genome Med 2024; 16:120. [PMID: 39434187 PMCID: PMC11492627 DOI: 10.1186/s13073-024-01395-4] [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: 11/30/2023] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Altered metabolism plays a role in the pathophysiology of cardiac diseases, such as atrial fibrillation (AF) and heart failure (HF). We aimed to identify novel plasma metabolites and proteins associating with cardiac disease. METHODS Mendelian randomisation (MR) was used to assess the association of 174 metabolites measured in up to 86,507 participants with AF, HF, dilated cardiomyopathy (DCM), and non-ischemic cardiomyopathy (NICM). Subsequently, we sourced data on 1567 plasma proteins and performed cis MR to identify proteins affecting the identified metabolites as well as the cardiac diseases. Proteins were prioritised on cardiac expression and druggability, and mapped to biological pathways. RESULTS We identified 35 metabolites associating with cardiac disease. AF was affected by seventeen metabolites, HF by nineteen, DCM by four, and NCIM by taurine. HF was particularly enriched for phosphatidylcholines (p = 0.029) and DCM for acylcarnitines (p = 0.001). Metabolite involvement with AF was more uniform, spanning for example phosphatidylcholines, amino acids, and acylcarnitines. We identified 38 druggable proteins expressed in cardiac tissue, with a directionally concordant effect on metabolites and cardiac disease. We recapitulated known associations, for example between the drug target of digoxin (AT1B2), taurine and NICM risk. Additionally, we identified numerous novel findings, such as higher RET values associating with phosphatidylcholines and decreasing AF and HF. RET is targeted by drugs such as regorafenib which has known cardiotoxic side-effects. Pathway analysis implicated involvement of GDF15 signalling through RET, and ghrelin regulation of energy homeostasis in cardiac pathogenesis. CONCLUSIONS This study identified 35 plasma metabolites involved with cardiac diseases and linked these to 38 druggable proteins, providing actionable leads for drug development.
Collapse
Affiliation(s)
- Marion van Vugt
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands.
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK.
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands.
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands.
| | - Chris Finan
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL British Heart Foundation Research Accelerator, London, UK
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Sandesh Chopade
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL British Heart Foundation Research Accelerator, London, UK
| | - Rui Providencia
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Connie R Bezzina
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- Department of Experimental Cardiology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- European Reference Network for rare, low prevalence and complex diseases of the heart: ERN GUARD-Heart , Amsterdam, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
| | - Jessica van Setten
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
| | - A Floriaan Schmidt
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- UCL British Heart Foundation Research Accelerator, London, UK
| |
Collapse
|
42
|
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
|
43
|
Chen ML, Kho PF, Guarischi-Sousa R, Zhou J, Panyard DJ, Azizi Z, Gupte T, Watson K, Abbasi F, Assimes TL. Plasma proteomics and carotid intima-media thickness in the UK biobank cohort. Front Cardiovasc Med 2024; 11:1478600. [PMID: 39416432 PMCID: PMC11480011 DOI: 10.3389/fcvm.2024.1478600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Background and aims Ultrasound derived carotid intima-media thickness (cIMT) is valuable for cardiovascular risk stratification. We assessed the relative importance of traditional atherosclerosis risk factors and plasma proteins in predicting cIMT measured nearly a decade later. Method We examined 6,136 UK Biobank participants with 1,461 proteins profiled using the proximity extension assay applied to their baseline blood draw who subsequently underwent a cIMT measurement. We implemented linear regression, stepwise Akaike Information Criterion-based, and the least absolute shrinkage and selection operator (LASSO) models to identify potential proteomic as well as non-proteomic predictors. We evaluated our model performance using the proportion variance explained (R 2). Result The mean time from baseline assessment to cIMT measurement was 9.2 years. Age, blood pressure, and anthropometric related variables were the strongest predictors of cIMT with fat-free mass index of the truncal region being the strongest predictor among adiposity measurements. A LASSO model incorporating variables including age, assessment center, genetic risk factors, smoking, blood pressure, trunk fat-free mass index, apolipoprotein B, and Townsend deprivation index combined with 97 proteins achieved the highest R 2 (0.308, 95% C.I. 0.274, 0.341). In contrast, models built with proteins alone or non-proteomic variables alone explained a notably lower R 2 (0.261, 0.228-0.294 and 0.260, 0.226-0.293, respectively). Chromogranin b (CHGB), Cystatin-M/E (CST6), leptin (LEP), and prolargin (PRELP) were the proteins consistently selected across all models. Conclusion Plasma proteins add to the clinical and genetic risk factors in predicting a cIMT measurement. Our findings implicate blood pressure and extracellular matrix-related proteins in cIMT pathophysiology.
Collapse
Affiliation(s)
- Ming-Li Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Pik Fang Kho
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Rodrigo Guarischi-Sousa
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Palo Alto Veterans Institute for Research (PAVIR), Stanford, CA, United States
| | - Jiayan Zhou
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Daniel J. Panyard
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Zahra Azizi
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Trisha Gupte
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Kathleen Watson
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Fahim Abbasi
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Themistocles L. Assimes
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
44
|
Dib MJ, Azzo JD, Zhao L, Salman O, Gan S, De Buyzere ML, De Meyer T, Ebert C, Gunawardhana K, Liu L, Gordon D, Seiffert D, Ching-Pin C, Zamani P, Cohen JB, Pourmussa B, Kun S, Gill D, Burgess S, van Empel V, Richards AM, Dennis J, Javaheri A, Mann DL, Cappola TP, Rietzschel E, Chirinos JA. Proteome-Wide Genetic Investigation of Large Artery Stiffness. JACC Basic Transl Sci 2024; 9:1178-1191. [PMID: 39534640 PMCID: PMC11551872 DOI: 10.1016/j.jacbts.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 11/16/2024]
Abstract
The molecular mechanisms contributing to large artery stiffness (LAS) are not fully understood. The aim of this study was to investigate the association between circulating plasma proteins and LAS using complementary proteomic and genomic analyses. A total of 106 proteins associated with carotid-femoral pulse-wave velocity, a noninvasive measure of LAS, were identified in 1,178 individuals from the Asklepios study cohort. Mendelian randomization analyses revealed causal effects of 13 genetically predicted plasma proteins on pulse pressure, including cartilage intermediate layer protein-2, high-temperature requirement A serine peptidase-1, and neuronal growth factor-1. These findings suggest potential novel therapeutic targets to reduce LAS and its related diseases.
Collapse
Affiliation(s)
- Marie-Joe Dib
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joe David Azzo
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lei Zhao
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - Oday Salman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sushrima Gan
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marc L. De Buyzere
- Department of Cardiovascular Diseases, Ghent University Hospital, Ghent, Belgium
| | - Tim De Meyer
- Department of Mathematical Modelling, Statistics and Bioinformatics, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | | | | | - Laura Liu
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - David Gordon
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | | | | | - Payman Zamani
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jordana B. Cohen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bianca Pourmussa
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seavmeiyin Kun
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Stephen Burgess
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Vanessa van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - A. Mark Richards
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | | | - Ali Javaheri
- Washington University School of Medicine, St. Louis, Missouri, USA
- John J. Cochran Veterans Hospital, St. Louis, Missouri, USA
| | - Douglas L. Mann
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Thomas P. Cappola
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ernst Rietzschel
- Department of Cardiovascular Diseases, Ghent University Hospital, Ghent, Belgium
| | - Julio A. Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
45
|
Carrasco-Zanini J, Wheeler E, Uluvar B, Kerrison N, Koprulu M, Wareham NJ, Pietzner M, Langenberg C. Mapping biological influences on the human plasma proteome beyond the genome. Nat Metab 2024; 6:2010-2023. [PMID: 39327534 PMCID: PMC11496106 DOI: 10.1038/s42255-024-01133-5] [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: 01/19/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024]
Abstract
Broad-capture proteomic platforms now enable simultaneous assessment of thousands of plasma proteins, but most of these are not actively secreted and their origins are largely unknown. Here we integrate genomic with deep phenomic information to identify modifiable and non-modifiable factors associated with 4,775 plasma proteins in ~8,000 mostly healthy individuals. We create a data-driven map of biological influences on the human plasma proteome and demonstrate segregation of proteins into clusters based on major explanatory factors. For over a third (N = 1,575) of protein targets, joint genetic and non-genetic factors explain 10-77% of the variation in plasma (median 19.88%, interquartile range 14.01-31.09%), independent of technical factors (median 2.48%, interquartile range 0.78-6.41%). Together with genetically anchored causal inference methods, our map highlights potential causal associations between modifiable risk factors and plasma proteins for hundreds of protein-disease associations, for example, COL6A3, which possibly mediates the association between reduced kidney function and cardiovascular disease. We provide a map of biological and technical influences on the human plasma proteome to help contextualize findings from proteomic studies.
Collapse
Affiliation(s)
- Julia Carrasco-Zanini
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Burulça Uluvar
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Kerrison
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Mine Koprulu
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Maik Pietzner
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Claudia Langenberg
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK.
| |
Collapse
|
46
|
Tachino J, Togami Y, Matsumoto H, Matsubara T, Seno S, Ogura H, Oda J. Plasma proteomics profile-based comparison of torso versus brain injury: A prospective cohort study. J Trauma Acute Care Surg 2024; 97:557-565. [PMID: 38595266 PMCID: PMC11446512 DOI: 10.1097/ta.0000000000004356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Trauma-related deaths and posttraumatic sequelae are a global health concern, necessitating a deeper understanding of the pathophysiology to advance trauma therapy. Proteomics offers insights into identifying and analyzing plasma proteins associated with trauma and inflammatory conditions; however, current proteomic methods have limitations in accurately measuring low-abundance plasma proteins. This study compared plasma proteomics profiles of patients from different acute trauma subgroups to identify new therapeutic targets and devise better strategies for personalized medicine. METHODS This prospective observational single-center cohort study was conducted between August 2020 and September 2021 in the intensive care unit of Osaka University Hospital in Japan. Enrolling 59 consecutive patients with blunt trauma, we meticulously analyzed plasma proteomics profiles in participants with torso or head trauma, comparing them with those of controls (mild trauma). Using the Olink Explore 3072 instrument (Olink Proteomics AB, Uppsala, Sweden), we identified five endotypes (α-ε) via unsupervised hierarchical clustering. RESULTS The median time from injury to blood collection was 47 minutes [interquartile range, 36-64 minutes]. The torso trauma subgroup exhibited 26 unique proteins with significantly altered expression, while the head trauma subgroup showed 68 unique proteins with no overlap between the two. The identified endotypes included α (torso trauma, n = 8), β (young patients with brain injury, n = 5), γ (severe brain injury postsurgery, n = 8), δ (torso or brain trauma with mild hyperfibrinolysis, n = 18), and ε (minor trauma, n = 20). Patients with torso trauma showed changes in blood pressure, smooth muscle adaptation, hypermetabolism, and hypoxemia. Patients with traumatic brain injury had dysregulated blood coagulation and altered nerves regeneration and differentiation. CONCLUSION This study identified unique plasma protein expression patterns in patients with torso trauma and traumatic brain injury, helping categorize five distinct endotypes. Our findings may offer new insights for clinicians, highlighting potential strategies for personalized medicine and improved trauma-related care. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
|
47
|
Wang QS, Hasegawa T, Namkoong H, Saiki R, Edahiro R, Sonehara K, Tanaka H, Azekawa S, Chubachi S, Takahashi Y, Sakaue S, Namba S, Yamamoto K, Shiraishi Y, Chiba K, Tanaka H, Makishima H, Nannya Y, Zhang Z, Tsujikawa R, Koike R, Takano T, Ishii M, Kimura A, Inoue F, Kanai T, Fukunaga K, Ogawa S, Imoto S, Miyano S, Okada Y. Statistically and functionally fine-mapped blood eQTLs and pQTLs from 1,405 humans reveal distinct regulation patterns and disease relevance. Nat Genet 2024; 56:2054-2067. [PMID: 39317738 PMCID: PMC11525184 DOI: 10.1038/s41588-024-01896-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/06/2024] [Indexed: 09/26/2024]
Abstract
Studying the genetic regulation of protein expression (through protein quantitative trait loci (pQTLs)) offers a deeper understanding of regulatory variants uncharacterized by mRNA expression regulation (expression QTLs (eQTLs)) studies. Here we report cis-eQTL and cis-pQTL statistical fine-mapping from 1,405 genotyped samples with blood mRNA and 2,932 plasma samples of protein expression, as part of the Japan COVID-19 Task Force (JCTF). Fine-mapped eQTLs (n = 3,464) were enriched for 932 variants validated with a massively parallel reporter assay. Fine-mapped pQTLs (n = 582) were enriched for missense variations on structured and extracellular domains, although the possibility of epitope-binding artifacts remains. Trans-eQTL and trans-pQTL analysis highlighted associations of class I HLA allele variation with KIR genes. We contrast the multi-tissue origin of plasma protein with blood mRNA, contributing to the limited colocalization level, distinct regulatory mechanisms and trait relevance of eQTLs and pQTLs. We report a negative correlation between ABO mRNA and protein expression because of linkage disequilibrium between distinct nearby eQTLs and pQTLs.
Collapse
Affiliation(s)
- Qingbo S Wang
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Takanori Hasegawa
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ho Namkoong
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan.
| | - Ryunosuke Saiki
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kyuto Sonehara
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiromu Tanaka
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shuhei Azekawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shotaro Chubachi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | - Saori Sakaue
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Children's Health and Genetics, Division of Health Science, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenichi Chiba
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroko Tanaka
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideki Makishima
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Zicong Zhang
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Rika Tsujikawa
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Ryuji Koike
- Health Science Research and Development Center (HeRD), Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomomi Takano
- Laboratory of Veterinary Infectious Disease, Department of Veterinary Medicine, Kitasato University, Tokyo, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akinori Kimura
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Fumitaka Inoue
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, the Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yukinori Okada
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan.
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan.
| |
Collapse
|
48
|
Ardissino M, Truong B, Slob EAW, Schuermans A, Yoshiji S, Morley AP, Burgess S, Ng FS, de Marvao A, Natarajan P, Nicolaides K, Gaziano L, Butterworth A, Honigberg MC. Proteome- and Transcriptome-Wide Genetic Analysis Identifies Biological Pathways and Candidate Drug Targets for Preeclampsia. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004755. [PMID: 39119725 PMCID: PMC7616531 DOI: 10.1161/circgen.124.004755] [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: 04/05/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Preeclampsia is a leading cause of maternal and perinatal morbidity and mortality. However, the current understanding of its underlying biological pathways remains limited. METHODS In this study, we performed a cross-platform proteome- and transcriptome-wide genetic analysis aimed at evaluating the causal relevance of >2000 circulating proteins with preeclampsia, supported by data on the expression of over 15 000 genes across 36 tissues leveraging large-scale preeclampsia genetic association data from women of European ancestry. RESULTS We demonstrate genetic associations of 18 circulating proteins with preeclampsia (SULT1A1 [sulfotransferase 1A1], SH2B3 [SH2B adapter protein 3], SERPINE2 [serpin family E member 2], RGS18 [regulator of G-protein signaling 18], PZP [pregnancy zone protein], NOTUM [notum, palmitoleoyl-protein carboxylesterase], METAP1 [methionyl aminopeptidase 1], MANEA [mannosidase endo-alpha], jun-D [JunD proto-oncogene], GDF15 [growth differentiation factor 15], FGL1 [fibrinogen like 1], FGF5 [fibroblast growth factor 5], FES [FES proto-oncogene], APOBR [apolipoprotein B receptor], ANP [natriuretic peptide A], ALDH-E2 [aldehyde dehydrogenase 2 family member], ADAMTS13 [ADAM metallopeptidase with thrombospondin type 1 motif 13], and 3MG [N-methylpurine DNA glycosylase]), among which 11 were either directly or indirectly supported by gene expression data, 9 were supported by Bayesian colocalization analyses, and 5 (SERPINE2, PZP, FGF5, FES, and ANP) were supported by all lines of evidence examined. Protein interaction mapping identified potential shared biological pathways through natriuretic peptide signaling, blood pressure regulation, immune tolerance, and thrombin activity regulation. CONCLUSIONS This investigation identified multiple targetable proteins linked to cardiovascular, inflammatory, and coagulation pathways, with SERPINE2, PZP, FGF5, FES, and ANP identified as pivotal proteins with likely causal roles in the development of preeclampsia. The identification of these potential targets may guide the development of targeted therapies for preeclampsia.
Collapse
Affiliation(s)
- Maddalena Ardissino
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (M.A., L.G., A.B.), University of Cambridge, United Kingdom
- Victor Phillip Dahdaleh Heart and Lung Research Institute (M.A, A.B.), University of Cambridge, United Kingdom
- National Heart and Lung Institute (M.A, F.S.N.), Imperial College London, United Kingdom
- Medical Research Council, London Institute of Medical Sciences (M.A.), Imperial College London, United Kingdom
| | - Buu Truong
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative (B.T., A.S., P.N., M.C.H.)
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (B.T., A.S., P.N., M.C.H.)
| | - Eric A W Slob
- MRC Biostatistics Unit (E.A.W.S., S.B.), University of Cambridge, United Kingdom
- Department of Applied Economics, Erasmus School of Economics (E.A.W.S.), Erasmus University Rotterdam, the Netherlands
- Erasmus University Rotterdam Institute for Behavior and Biology (E.A.W.S.), Erasmus University Rotterdam, the Netherlands
- Department of Psychology, Education and Child Studies (E.A.W.S.), Erasmus University Rotterdam, the Netherlands
| | - Art Schuermans
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative (B.T., A.S., P.N., M.C.H.)
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (B.T., A.S., P.N., M.C.H.)
- Faculty of Medicine, KU Leuven, Belgium (A.S.)
| | - Satoshi Yoshiji
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA (S.Y.)
| | - Alec P Morley
- Department of Medicine, School of Clinical Medicine (A.P.M.), University of Cambridge, United Kingdom
- Gonville and Caius College (A.P.M.), University of Cambridge, United Kingdom
| | - Stephen Burgess
- MRC Biostatistics Unit (E.A.W.S., S.B.), University of Cambridge, United Kingdom
| | - Fu Siong Ng
- National Heart and Lung Institute (M.A, F.S.N.), Imperial College London, United Kingdom
| | - Antonio de Marvao
- Department of Women and Children's Health (A.d.M., K.N.)
- British Heart Foundation Center of Research Excellence, School of Cardiovascular Medicine and Sciences (A.d.M.), King's College Hospital, London, United Kingdom
- Fetal Medicine Research Institute (A.d.M., K.N.), King's College Hospital, London, United Kingdom
| | - Pradeep Natarajan
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative (B.T., A.S., P.N., M.C.H.)
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (B.T., A.S., P.N., M.C.H.)
| | - Kypros Nicolaides
- Department of Women and Children's Health (A.d.M., K.N.)
- Fetal Medicine Research Institute (A.d.M., K.N.), King's College Hospital, London, United Kingdom
| | - Liam Gaziano
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (M.A., L.G., A.B.), University of Cambridge, United Kingdom
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System (L.G.), Harvard Medical School, Boston
| | - Adam Butterworth
- Victor Phillip Dahdaleh Heart and Lung Research Institute (M.A, A.B.), University of Cambridge, United Kingdom
- British Heart Foundation Center of Research Excellence (A.B.), University of Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus (A.B.), University of Cambridge, United Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics (A.B.), University of Cambridge, United Kingdom
| | - Michael C Honigberg
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative (B.T., A.S., P.N., M.C.H.)
- Department of Medicine (M.C.H.), Harvard Medical School, Boston
| |
Collapse
|
49
|
Tahir UA, Barber JL, Cruz DE, Kars ME, Deng S, Tuftin B, Gillman MG, Benson MD, Robbins JM, Chen ZZ, Rao P, Katz DH, Farrell L, Sofer T, Hall ME, Ekunwe L, Tracy RP, Durda P, Taylor KD, Liu Y, Johnson WC, Guo X, Chen YDI, Manichaikul AW, Jain D, NHLBI Trans-Omics for Precision Medicine Consortium, Wang TJ, Reiner AP, Natarajan P, Itan Y, Rich SS, Rotter JI, Wilson JG, Raffield LM, Gerszten RE. Proteogenomic analysis integrated with electronic health records data reveals disease-associated variants in Black Americans. J Clin Invest 2024; 134:e181802. [PMID: 39316441 PMCID: PMC11527441 DOI: 10.1172/jci181802] [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: 04/12/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUNDMost GWAS of plasma proteomics have focused on White individuals of European ancestry, limiting biological insight from other ancestry-enriched protein quantitative loci (pQTL).METHODSWe conducted a discovery GWAS of approximately 3,000 plasma proteins measured by the antibody-based Olink platform in 1,054 Black adults from the Jackson Heart Study (JHS) and validated our findings in the Multi-Ethnic Study of Atherosclerosis (MESA). The genetic architecture of identified pQTLs was further explored through fine mapping and admixture association analysis. Finally, using our pQTL findings, we performed a phenome-wide association study (PheWAS) across 2 large multiethnic electronic health record (EHR) systems in All of Us and BioMe.RESULTSWe identified 1,002 pQTLs for 925 protein assays. Fine mapping and admixture analyses suggested allelic heterogeneity of the plasma proteome across diverse populations. We identified associations for variants enriched in African ancestry, many in diseases that lack precise biomarkers, including cis-pQTLs for cathepsin L (CTSL) and Siglec-9, which were linked with sarcoidosis and non-Hodgkin's lymphoma, respectively. We found concordant associations across clinical diagnoses and laboratory measurements, elucidating disease pathways, including a cis-pQTL associated with circulating CD58, WBC count, and multiple sclerosis.CONCLUSIONSOur findings emphasize the value of leveraging diverse populations to enhance biological insights from proteomics GWAS, and we have made this resource readily available as an interactive web portal.FUNDINGNIH K08 HL161445-01A1; 5T32HL160522-03; HHSN268201600034I; HL133870.
Collapse
Affiliation(s)
- Usman A. Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob L. Barber
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel E. Cruz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Shuliang Deng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Madeline G. Gillman
- University of North Carolina School of Medicine, Raleigh, North Carolina, USA
| | - Mark D. Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeremy M. Robbins
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Prashant Rao
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Laurie Farrell
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Tamar Sofer
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael E. Hall
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lynette Ekunwe
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Russell P. Tracy
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Peter Durda
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Ani W. Manichaikul
- Center for Public Health Genomics and
- Division of Biostatistics and Epidemiology, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Deepti Jain
- University of Washington, Seattle, Washington
| | | | - Thomas J. Wang
- Department of Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Yuval Itan
- University of North Carolina School of Medicine, Raleigh, North Carolina, USA
| | | | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - James G. Wilson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura M. Raffield
- University of North Carolina School of Medicine, Raleigh, North Carolina, USA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
50
|
Liu GY, Perry AS, Washko GR, Farber-Eger E, Colangelo LA, Sheng Q, Wells Q, Huang X, Thyagarajan B, Guan W, Alexandria SJ, San José Estépar R, Bowler RP, Esposito AJ, Khan SS, Shah RV, Choi B, Kalhan R. Proteomic Risk Score of Increased Respiratory Susceptibility: A Multi-Cohort Study. Am J Respir Crit Care Med 2024; 211:64-74. [PMID: 39254293 PMCID: PMC11755364 DOI: 10.1164/rccm.202403-0613oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/17/2024] [Indexed: 09/11/2024] Open
Abstract
RATIONALE Accelerated decline in lung function is associated with incident COPD, hospitalizations and death. However, identifying this trajectory with longitudinal spirometry measurements is challenging in clinical practice. OBJECTIVE To determine whether a proteomic risk score trained on accelerated decline in lung function can assess risk of future respiratory disease and mortality. METHODS In CARDIA, a population-based cohort starting in young adulthood, longitudinal measurements of FEV1 percent predicted (up to six timepoints over 30 years) were used to identify accelerated and normal decline trajectories. Protein aptamers associated with an accelerated decline trajectory were identified with multivariable logistic regression followed by LASSO regression. The proteomic respiratory susceptibility score was derived based on these circulating proteins and applied to the UK Biobank and COPDGene studies to examine associations with future respiratory morbidity and mortality. MEASUREMENTS AND RESULTS Higher susceptibility score was independently associated with all-cause mortality (UKBB: HR 1.56, 95%CI 1.50-1.61; COPDGene: HR 1.75, 95%CI 1.63-1.88), respiratory mortality (UKBB: HR 2.39, 95% CI 2.16-2.64; COPDGene: HR 1.83, 95%CI 1.33-2.51), incident COPD (UKBB: HR 1.84, 95%CI 1.71-1.98), incident respiratory exacerbation (COPDGene: OR 1.11, 95%CI 1.03-1.20), and incident exacerbation requiring hospitalization (COPDGene: OR 1.18, 95%CI 1.08-1.28). CONCLUSIONS A proteomic signature of increased respiratory susceptibility identifies people at risk of respiratory death, incident COPD, and respiratory exacerbations. This susceptibility score is comprised of proteins with well-known and novel associations with lung health and holds promise for the early detection of lung disease without requiring years of spirometry measurements.
Collapse
Affiliation(s)
- Gabrielle Y Liu
- University of California Davis School of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Sacramento, California, United States
| | - Andrew S Perry
- Vanderbilt University Medical Center, Division of Cardiology, Nashville, Tennessee, United States
| | - George R Washko
- Brigham and Women's Hospital, Division of Pulmonary and Critical Care Medicine, Boston, Massachusetts, United States
| | - Eric Farber-Eger
- Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Laura A Colangelo
- Northwestern University, Medicine/Cardiology, Chicago, Illinois, United States
| | - Quanhu Sheng
- Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Quinn Wells
- Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Xiaoning Huang
- Northwestern University Feinberg School of Medicine, Division of Cardiology, Chicago, Illinois, United States
| | | | - Weihua Guan
- University of Minnesota Twin Cities, Division of Biostatistics, Minneapolis, Minnesota, United States
| | - Shaina J Alexandria
- Northwestern University Feinberg School of Medicine, Department of Preventive Medicine, Chicago, Illinois, United States
| | | | - Russell P Bowler
- National Jewish Medical and Research Center, Department of Medicine, Denver, Colorado, United States
| | - Anthony J Esposito
- Northwestern Medicine, Division of Pulmonary and Critical Care Medicine, Chicago, Illinois, United States
| | - Sadiya S Khan
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Ravi V Shah
- Vanderbilt University Medical Center, Division of Cardiology, Nashville, Tennessee, United States
| | - Bina Choi
- Brigham and Women's Hospital, Division of Pulmonary and Critical Care Medicine, Boston, Massachusetts, United States
| | - Ravi Kalhan
- Northwestern University Feinberg School of Medicine, Division of Pulmonary and Critical Care Medicine, Chicago, Illinois, United States;
| |
Collapse
|