1
|
Harder EM, Nardelli P, Pistenmaa CL, Ash SY, Balasubramanian A, Bowler RP, Iturrioz Campo M, Diaz AA, Hassoun PM, Leopold JA, Martinez FJ, Nathan SD, Noth I, Podolanczuk AJ, Saggar R, San José Estépar R, Shlobin OA, Wang W, Waxman AB, Putman RK, Washko GR, Choi B, San José Estépar R, Rahaghi FN. Preacinar Arterial Dilation Mediates Outcomes of Quantitative Interstitial Abnormalities in the COPDGene Study. Am J Respir Crit Care Med 2024; 210:1132-1142. [PMID: 38820122 DOI: 10.1164/rccm.202312-2342oc] [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: 12/23/2023] [Accepted: 05/31/2024] [Indexed: 06/02/2024] Open
Abstract
Rationale: Quantitative interstitial abnormalities (QIAs) are a computed tomography (CT) measure of early parenchymal lung disease associated with worse clinical outcomes, including exercise capacity and symptoms. The presence of pulmonary vasculopathy in QIAs and its role in the QIA-outcome relationship is unknown. Objectives: To quantify radiographic pulmonary vasculopathy in QIAs and determine whether this vasculopathy mediates the QIA-outcome relationship. Methods: Ever-smokers with QIAs, outcomes, and pulmonary vascular mediator data were identified from the Genetic Epidemiology of COPD (COPDGene) study cohort. CT-based vascular mediators were right ventricle-to-left ventricle ratio, pulmonary artery-to-aorta ratio, and preacinar intraparenchymal arterial dilation (pulmonary artery volume, 5-20 mm2 in cross-sectional area, normalized to total arterial volume). Outcomes were 6-minute walk distance and a modified Medical Council Research Council Dyspnea Scale score of 2 or higher. Adjusted causal mediation analyses were used to determine whether the pulmonary vasculature mediated the QIA effect on outcomes. Associations of preacinar arterial dilation with select plasma biomarkers of pulmonary vascular dysfunction were examined. Measurements and Main Results: Among 8,200 participants, QIA burden correlated positively with vascular damage measures, including preacinar arterial dilation. Preacinar arterial dilation mediated 79.6% of the detrimental impact of QIA on 6-minute walk distance (56.2-100%; P < 0.001). Pulmonary artery-to-aorta ratio was a weak mediator, and right ventricle-to-left ventricle ratio was a suppressor. Similar results were observed in the relationship between QIA and modified Medical Council Research Council dyspnea score. Preacinar arterial dilation correlated with increased pulmonary vascular dysfunction biomarker levels, including angiopoietin-2 and N-terminal brain natriuretic peptide. Conclusions: Parenchymal QIAs deleteriously impact outcomes primarily through pulmonary vasculopathy. Preacinar arterial dilation may be a novel marker of pulmonary vasculopathy in QIAs.
Collapse
Affiliation(s)
| | | | | | - Samuel Y Ash
- Department of Critical Care Medicine, South Shore Hospital, South Weymouth, Massachusetts, and School of Medicine, Tufts University, Boston, Massachusetts
| | - Aparna Balasubramanian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Russell P Bowler
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | | | | | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | - Fernando J Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York City, New York
| | - Steven D Nathan
- Inova Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, Falls Church, Virginia
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Anna J Podolanczuk
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York City, New York
| | - Rajan Saggar
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, Los Angeles, Los Angeles, California; and
| | | | - Oksana A Shlobin
- Inova Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, Falls Church, Virginia
| | - Wei Wang
- Division of Sleep and Circadian Disorders, Department of Medicine, and Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Bina Choi
- Division of Pulmonary and Critical Care Medicine
| | | | | |
Collapse
|
2
|
Lacinski RA, Dziadowicz SA, Roth CA, Ma L, Melemai VK, Fitzpatrick B, Chaharbakhshi E, Heim T, Lohse I, Schoedel KE, Hu G, Llosa NJ, Weiss KR, Lindsey BA. Proteomic and transcriptomic analyses identify apo-transcobalamin-II as a biomarker of overall survival in osteosarcoma. Front Oncol 2024; 14:1417459. [PMID: 39493449 PMCID: PMC11527601 DOI: 10.3389/fonc.2024.1417459] [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: 04/15/2024] [Accepted: 09/17/2024] [Indexed: 11/05/2024] Open
Abstract
Background The large-scale proteomic platform known as the SomaScan® assay is capable of simultaneously measuring thousands of proteins in patient specimens through next-generation aptamer-based multiplexed technology. While previous studies have utilized patient peripheral blood to suggest serum biomarkers of prognostic or diagnostic value in osteosarcoma (OSA), the most common primary pediatric bone cancer, they have ultimately been limited in the robustness of their analyses. We propose utilizing this aptamer-based technology to describe the systemic proteomic milieu in patients diagnosed with this disease. Methods To determine novel biomarkers associated with overall survival in OSA, we deployed the SomaLogic SomaScan® 7k assay to investigate the plasma proteomic profile of naive primary, recurrent, and metastatic OSA patients. Following identification of differentially expressed proteins (DEPs) between 2-year deceased and survivor cohorts, publicly available databases including Survival Genie, TIGER, and KM Plotter Immunotherapy, among others, were utilized to investigate the significance of our proteomic findings. Results Apo-transcobalamin-II (APO-TCN2) was identified as the most DEP between 2-year deceased and survivor cohorts (Log2 fold change = 6.8, P-value = 0.0017). Survival analysis using the Survival Genie web-based platform indicated that increased intratumoral TCN2 expression was associated with better overall survival in both OSA (TARGET-OS) and sarcoma (TCGA-SARC) datasets. Cell-cell communication analysis using the TIGER database suggested that TCN2+ Myeloid cells likely interact with marginal zone and immunoglobin-producing B lymphocytes expressing the TCN2 receptor (CD320) to promote their proliferation and survival in both non-small cell lung cancer and melanoma tumors. Analysis of publicly available OSA scRNA-sequencing datasets identified similar populations in naive primary tumors. Furthermore, circulating APO-TCN2 levels in OSA were then associated with a plasma proteomic profile likely necessary for robust B lymphocyte proliferation, infiltration, and formation of intratumoral tertiary lymphoid structures for improved anti-tumor immunity. Conclusions Overall, APO-TCN2, a circulatory protein previously described in various lymphoproliferative disorders, was associated with 2-year survival status in patients diagnosed with OSA. The relevance of this protein and apparent immunological function (anti-tumor B lymphocyte responses) was suggested using publicly available solid tumor RNA-sequencing datasets. Further studies characterizing the biological function of APO-TCN2 and its relevance in these diseases is warranted.
Collapse
Affiliation(s)
- Ryan A. Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
- West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Clark A. Roth
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Li Ma
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Vincent K. Melemai
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Brody Fitzpatrick
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Edwin Chaharbakhshi
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Tanya Heim
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ines Lohse
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Karen E. Schoedel
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Nicolas J. Llosa
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kurt R. Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brock A. Lindsey
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Duong T, Austin TR, Brody JA, Shojaie A, Battle A, Bader JS, Hong YS, Ballantyne CM, Coresh J, Gerszten RE, Tracy RP, Psaty BM, Sotoodehnia N, Arking DE. Circulating Blood Plasma Profiling Reveals Proteomic Signature and a Causal Role for SVEP1 in Sudden Cardiac Death. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004494. [PMID: 39234668 PMCID: PMC11479847 DOI: 10.1161/circgen.123.004494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Affiliation(s)
- ThuyVy Duong
- McKusick-Nathans Inst, Dept of Genetic Medicine, Johns Hopkins Univ School of Medicine, Baltimore, MD
| | - Thomas R. Austin
- Cardiovascular Health Rsrch Unit, Dept of Medicine, Univ of Washington, Seattle, WA
- Dept of Epidemiology, Univ of Washington, Seattle, WA
| | - Jennifer A. Brody
- Cardiovascular Health Rsrch Unit, Dept of Medicine, Univ of Washington, Seattle, WA
| | - Ali Shojaie
- Dept of Biostatistics, Univ of Washington, Seattle, WA
| | - Alexis Battle
- Dept of Biomedical Engineering, Johns Hopkins Univ, Baltimore, MD
- Dept of Computer Science, Johns Hopkins Univ, Baltimore, MD
| | - Joel S. Bader
- Dept of Biomedical Engineering, Johns Hopkins Univ, Baltimore, MD
| | - Yun Soo Hong
- McKusick-Nathans Inst, Dept of Genetic Medicine, Johns Hopkins Univ School of Medicine, Baltimore, MD
| | | | - Josef Coresh
- Dept of Epidemiology, Johns Hopkins Univ Bloomberg School of Public Health, Baltimore, MD
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Ctr, Boston, MA
| | - Russell P. Tracy
- Dept of Pathology & Laboratory Medicine & Biochemistry, Larner College of Medicine, Univ of Vermont, Burlington, VT
| | - Bruce M. Psaty
- Cardiovascular Health Rsrch Unit, Dept of Medicine, Univ of Washington, Seattle, WA
- Dept of Epidemiology, Univ of Washington, Seattle, WA
- Dept of Health Systems & Population Health, Univ of Washington, Seattle, WA
| | - Nona Sotoodehnia
- Cardiovascular Health Rsrch Unit, Dept of Medicine, Univ of Washington, Seattle, WA
| | - Dan E Arking
- McKusick-Nathans Inst, Dept of Genetic Medicine, Johns Hopkins Univ School of Medicine, Baltimore, MD
| |
Collapse
|
4
|
Pleshakova TO, Ershova MO, Valueva AA, Ivanova IA, Ivanov YD, Archakov AI. AFM-fishing technology for protein detection in solutions. BIOMEDITSINSKAIA KHIMIIA 2024; 70:273-286. [PMID: 39324193 DOI: 10.18097/pbmc20247005273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The review considers the possibility of using atomic force microscopy (AFM) as a basic method for protein detection in solutions with low protein concentrations. The demand for new bioanalytical approaches is determined by the problem of insufficient sensitivity of systems used in routine practice for protein detection. Special attention is paid to demonstration of the use in bioanalysis of a combination of AFM and fishing methods as an approach of concentrating biomolecules from a large volume of the analyzed solution on a small surface area.
Collapse
Affiliation(s)
| | - M O Ershova
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A A Valueva
- Institute of Biomedical Chemistry, Moscow, Russia
| | - I A Ivanova
- Institute of Biomedical Chemistry, Moscow, Russia
| | - Yu D Ivanov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A I Archakov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
5
|
Wang S, Rao Z, Cao R, Blaes AH, Coresh J, Deo R, Dubin R, Joshu CE, Lehallier B, Lutsey PL, Pankow JS, Post WS, Rotter JI, Sedaghat S, Tang W, Thyagarajan B, Walker KA, Ganz P, Platz EA, Guan W, Prizment A. Development, characterization, and replication of proteomic aging clocks: Analysis of 2 population-based cohorts. PLoS Med 2024; 21:e1004464. [PMID: 39316596 PMCID: PMC11460707 DOI: 10.1371/journal.pmed.1004464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/08/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Biological age may be estimated by proteomic aging clocks (PACs). Previous published PACs were constructed either in smaller studies or mainly in white individuals, and they used proteomic measures from only one-time point. In this study, we created de novo PACs and compared their performance to published PACs at 2 different time points in the Atherosclerosis Risk in Communities (ARIC) study of white and black participants (around 75% white and 25% black). MEDTHODS AND FINDINGS A total of 4,712 plasma proteins were measured using SomaScan in blood samples collected in 1990 to 1992 from 11,761 midlife participants (aged 46 to 70 years) and in 2011 to 2013 from 5,183 late-life participants (aged 66 to 90 years). The de novo ARIC PACs were constructed by training them against chronological age using elastic net regression in two-thirds of healthy participants in midlife and late life and validated in the remaining one-third of healthy participants at the corresponding time point. We also computed 3 published PACs. We estimated age acceleration for each PAC as residuals after regressing each PAC on chronological age. We also calculated the change in age acceleration from midlife to late life. We examined the associations of age acceleration and change in age acceleration with mortality through 2019 from all-cause, cardiovascular disease (CVD), cancer, and lower respiratory disease (LRD) using Cox proportional hazards regression in participants (irrespective of health) after excluding the training set. The model was adjusted for chronological age, smoking, body mass index (BMI), and other confounders. We externally validated the midlife PAC using the Multi-Ethnic Study of Atherosclerosis (MESA) Exam 1 data. The ARIC PACs had a slightly stronger correlation with chronological age than published PACs in healthy participants at each time point. Associations with mortality were similar for the ARIC PACs and published PACs. For late-life and midlife age acceleration for the ARIC PACs, respectively, hazard ratios (HRs) per 1 standard deviation were 1.65 and 1.38 (both p < 0.001) for all-cause mortality, 1.37 and 1.20 (both p < 0.001) for CVD mortality, 1.21 (p = 0.028) and 1.04 (p = 0.280) for cancer mortality, and 1.68 and 1.36 (both p < 0.001) for LRD mortality. For the change in age acceleration, HRs for all-cause, CVD, and LRD mortality were comparable to the HRs for late-life age acceleration. The association between the change in age acceleration and cancer mortality was not significant. The external validation of the midlife PAC in MESA showed significant associations with mortality, as observed for midlife participants in ARIC. The main limitation is that our PACs were constructed in midlife and late-life participants. It is unknown whether these PACs could be applied to young individuals. CONCLUSIONS In this longitudinal study, we found that the ARIC PACs and published PACs were similarly associated with an increased risk of mortality. These findings suggested that PACs show promise as biomarkers of biological age. PACs may be serve as tools to predict mortality and evaluate the effect of anti-aging lifestyle and therapeutic interventions.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zexi Rao
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Rui Cao
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Josef Coresh
- Departments of Population Health and Medicine, New York University Glossman School of Medicine, New York, New York, United States of America
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ruth Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | | | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Peter Ganz
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Weihua Guan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
6
|
Oliver N, Choi MJ, Arul AB, Whitaker MD, Robinson RAS. Establishing Quality Control Metrics for Large-Scale Plasma Proteomic Sample Preparation. ACS MEASUREMENT SCIENCE AU 2024; 4:442-451. [PMID: 39184360 PMCID: PMC11342454 DOI: 10.1021/acsmeasuresciau.3c00070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 08/27/2024]
Abstract
Large-scale plasma proteomics studies have been transformed due to the multiplexing and automation of sample preparation workflows. However, these workflows can suffer from reproducibility issues, a lack of standardized quality control (QC) metrics, and the assessment of variation before liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. The incorporation of robust QC metrics in sample preparation workflows ensures better reproducibility, lower assay variation, and better-informed decisions for troubleshooting. Our laboratory conducted a plasma proteomics study of a cohort of patient samples (N = 808) using tandem mass tag (TMT) 16-plex batches (N = 58). The proteomic workflow consisted of protein depletion, protein digestion, TMT labeling, and fractionation. Five QC sample types (QCstd, QCdig, QCpool, QCTMT, and QCBSA) were created to measure the performance of sample preparation prior to the final LC-MS/MS analysis. We measured <10% CV for individual sample preparation steps in the proteomic workflow based on data from various QC sample steps. The establishment of robust measures for QC of sample preparation steps allowed for greater confidence in prepared samples for subsequent LC-MS/MS analysis. This study also provides recommendations for standardized QC metrics that can assist with future large-scale cohort sample preparation workflows.
Collapse
Affiliation(s)
- Nekesa
C. Oliver
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Min Ji Choi
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Albert B. Arul
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Marsalas D. Whitaker
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Renã A. S. Robinson
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt
Memory and Alzheimer’s Center, Vanderbilt
University Medical Center, Nashville, Tennessee 37212, United States
- Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States
- Vanderbilt
Brain Institute, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Neurology, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
7
|
Ruvuna L, Hijazi K, Guzman DE, Guo C, Loureiro J, Khokhlovich E, Morris M, Obeidat M, Pratte KA, DiLillo KM, Sharma S, Kechris K, Anzueto A, Barjaktarevic I, Bleecker ER, Casaburi R, Comellas A, Cooper CB, DeMeo DL, Foreman M, Flenaugh EL, Han MK, Hanania NA, Hersh CP, Krishnan JA, Labaki WW, Martinez FJ, O’Neal WK, Paine R, Peters SP, Woodruff PG, Wells JM, Wendt CH, Arnold KB, Barr RG, Curtis JL, Ngo D, Bowler RP. Dynamic and prognostic proteomic associations with FEV 1 decline in chronic obstructive pulmonary disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311507. [PMID: 39148837 PMCID: PMC11326337 DOI: 10.1101/2024.08.07.24311507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Rationale Identification and validation of circulating biomarkers for lung function decline in COPD remains an unmet need. Objective Identify prognostic and dynamic plasma protein biomarkers of COPD progression. Methods We measured plasma proteins using SomaScan from two COPD-enriched cohorts, the Subpopulations and Intermediate Outcomes Measures in COPD Study (SPIROMICS) and Genetic Epidemiology of COPD (COPDGene), and one population-based cohort, Multi-Ethnic Study of Atherosclerosis (MESA) Lung. Using SPIROMICS as a discovery cohort, linear mixed models identified baseline proteins that predicted future change in FEV1 (prognostic model) and proteins whose expression changed with change in lung function (dynamic model). Findings were replicated in COPDGene and MESA-Lung. Using the COPD-enriched cohorts, Gene Set Enrichment Analysis (GSEA) identified proteins shared between COPDGene and SPIROMICS. Metascape identified significant associated pathways. Measurements and Main Results The prognostic model found 7 significant proteins in common (p < 0.05) among all 3 cohorts. After applying false discovery rate (adjusted p < 0.2), leptin remained significant in all three cohorts and growth hormone receptor remained significant in the two COPD cohorts. Elevated baseline levels of leptin and growth hormone receptor were associated with slower rate of decline in FEV1. Twelve proteins were nominally but not FDR significant in the dynamic model and all were distinct from the prognostic model. Metascape identified several immune related pathways unique to prognostic and dynamic proteins. Conclusion We identified leptin as the most reproducible COPD progression biomarker. The difference between prognostic and dynamic proteins suggests disease activity signatures may be different from prognosis signatures.
Collapse
Affiliation(s)
- Lisa Ruvuna
- Pulmonary Sciences and Critical Care Medicine University of Colorado Denver, Colorado
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Kahkeshan Hijazi
- Novartis Institute of Biomedical Research, Cambridge, MA, United States
| | - Daniel E. Guzman
- Columbia University Irving Medical Center, New York Presbyterian, New York, NY, United States
| | - Claire Guo
- National Jewish Health, Denver, CO, United States
| | - Joseph Loureiro
- Novartis Institute of Biomedical Research, Cambridge, MA, United States
| | | | - Melody Morris
- Novartis Institute of Biomedical Research, Cambridge, MA, United States
| | - Ma’en Obeidat
- Novartis Institute of Biomedical Research, Cambridge, MA, United States
| | | | - Katarina M. DiLillo
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Sunita Sharma
- Pulmonary Sciences and Critical Care Medicine University of Colorado Denver, Colorado
| | - Katerina Kechris
- Department of Biostatics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Antonio Anzueto
- University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, Texas
| | - Igor Barjaktarevic
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | | | - Richard Casaburi
- Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, Torrance, California
| | | | - Christopher B. Cooper
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Marilyn Foreman
- Pulmonary and Critical Care Medicine Division, Morehouse School of Medicine, Atlanta, GA
| | - Eric L. Flenaugh
- Pulmonary and Critical Care Medicine Division, Morehouse School of Medicine, Atlanta, GA
| | - MeiLan K. Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Nicola A. Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX
| | - Craig P. Hersh
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Jerry A. Krishnan
- Breathe Chicago Center, Division of Pulmonary and Critical Care Medicine, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Wassim W. Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Fernando J. Martinez
- Department of Medicine, Weill Cornell Medical College, New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - Wanda K. O’Neal
- Marsico Lung Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Robert Paine
- Division of Respiratory, Critical Care and Occupational Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Stephen P. Peters
- Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Prescott G. Woodruff
- Division of Pulmonary Critical Care, Allergy, and Sleep Medicine, University of California San Francisco, San Francisco, California, United States
| | - J Michael Wells
- Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Christine H. Wendt
- Minneapolis VA Health Care System, Minneapolis, Minnesota
- University of Minnesota, Minneapolis, Minnesota Medical Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - R. Graham Barr
- Columbia University Irving Medical Center, New York Presbyterian, New York, NY, United States
| | - Jeffrey L. Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
- Medical Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Debby Ngo
- Novartis Institute of Biomedical Research, Cambridge, MA, United States
| | - Russell P. Bowler
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
| | | |
Collapse
|
8
|
Allen LH, Fenech M, LeVatte MA, West KP, Wishart DS. Multiomics: Functional Molecular Biomarkers of Micronutrients for Public Health Application. Annu Rev Nutr 2024; 44:125-153. [PMID: 39207879 DOI: 10.1146/annurev-nutr-062322-022751] [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: 09/04/2024]
Abstract
Adequate micronutrient intake and status are global public health goals. Vitamin and mineral deficiencies are widespread and known to impair health and survival across the life stages. However, knowledge of molecular effects, metabolic pathways, biological responses to variation in micronutrient nutriture, and abilities to assess populations for micronutrient deficiencies and their pathology remain lacking. Rapidly evolving methodological capabilities in genomics, epigenomics, proteomics, and metabolomics offer unparalleled opportunities for the nutrition research community to link micronutrient exposure to cellular health; discover new, arguably essential micronutrients of microbial origin; and integrate methods of molecular biology, epidemiology, and intervention trials to develop novel approaches to assess and prevent micronutrient deficiencies in populations. In this review article, we offer new terminology to specify nutritional application of multiomic approaches and encourage collaboration across the basic to public health sciences to advance micronutrient deficiency prevention.
Collapse
Affiliation(s)
- Lindsay H Allen
- Western Human Nutrition Research Center, United States Department of Agriculture, Agricultural Research Service, Davis, California, USA
- Department of Nutrition, University of California, Davis, California, USA
| | - Michael Fenech
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- Genome Health Foundation, North Brighton, South Australia, Australia
| | - Marcia A LeVatte
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Keith P West
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| | - David S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Joyce T, Tasci E, Jagasia S, Shephard J, Chappidi S, Zhuge Y, Zhang L, Cooley Zgela T, Sproull M, Mackey M, Camphausen K, Krauze AV. Serum CD133-Associated Proteins Identified by Machine Learning Are Connected to Neural Development, Cancer Pathways, and 12-Month Survival in Glioblastoma. Cancers (Basel) 2024; 16:2740. [PMID: 39123468 PMCID: PMC11311306 DOI: 10.3390/cancers16152740] [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: 06/17/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Glioma is the most prevalent type of primary central nervous system cancer, while glioblastoma (GBM) is its most aggressive variant, with a median survival of only 15 months when treated with maximal surgical resection followed by chemoradiation therapy (CRT). CD133 is a potentially significant GBM biomarker. However, current clinical biomarker studies rely on invasive tissue samples. These make prolonged data acquisition impossible, resulting in increased interest in the use of liquid biopsies. Our study, analyzed 7289 serum proteins from 109 patients with pathology-proven GBM obtained prior to CRT using the aptamer-based SOMAScan® proteomic assay technology. We developed a novel methodology that identified 24 proteins linked to both serum CD133 and 12-month overall survival (OS) through a multi-step machine learning (ML) analysis. These identified proteins were subsequently subjected to survival and clustering evaluations, categorizing patients into five risk groups that accurately predicted 12-month OS based on their protein profiles. Most of these proteins are involved in brain function, neural development, and/or cancer biology signaling, highlighting their significance and potential predictive value. Identifying these proteins provides a valuable foundation for future serum investigations as validation of clinically applicable GBM biomarkers can unlock immense potential for diagnostics and treatment monitoring.
Collapse
Affiliation(s)
- Thomas Joyce
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Erdal Tasci
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Sarisha Jagasia
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Jason Shephard
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Shreya Chappidi
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
- Department of Computer Science and Technology, University of Cambridge, 15 JJ Thomson Ave, Cambridge CB3 0FD, UK
| | - Ying Zhuge
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Longze Zhang
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Theresa Cooley Zgela
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Mary Sproull
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Megan Mackey
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Kevin Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Andra V. Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| |
Collapse
|
10
|
Tripp BA, Dillon ST, Yuan M, Asara JM, Vasunilashorn SM, Fong TG, Inouye SK, Ngo LH, Marcantonio ER, Xie Z, Libermann TA, Otu HH. Integrated Multi-Omics Analysis of Cerebrospinal Fluid in Postoperative Delirium. Biomolecules 2024; 14:924. [PMID: 39199312 PMCID: PMC11352186 DOI: 10.3390/biom14080924] [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: 06/29/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024] Open
Abstract
Preoperative risk biomarkers for delirium may aid in identifying high-risk patients and developing intervention therapies, which would minimize the health and economic burden of postoperative delirium. Previous studies have typically used single omics approaches to identify such biomarkers. Preoperative cerebrospinal fluid (CSF) from the Healthier Postoperative Recovery study of adults ≥ 63 years old undergoing elective major orthopedic surgery was used in a matched pair delirium case-no delirium control design. We performed metabolomics and lipidomics, which were combined with our previously reported proteomics results on the same samples. Differential expression, clustering, classification, and systems biology analyses were applied to individual and combined omics datasets. Probabilistic graph models were used to identify an integrated multi-omics interaction network, which included clusters of heterogeneous omics interactions among lipids, metabolites, and proteins. The combined multi-omics signature of 25 molecules attained an AUC of 0.96 [95% CI: 0.85-1.00], showing improvement over individual omics-based classification. We conclude that multi-omics integration of preoperative CSF identifies potential risk markers for delirium and generates new insights into the complex pathways associated with delirium. With future validation, this hypotheses-generating study may serve to build robust biomarkers for delirium and improve our understanding of its pathophysiology.
Collapse
Affiliation(s)
- Bridget A. Tripp
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Simon T. Dillon
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.)
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
| | - Min Yuan
- Division of Signal Transduction and Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - John M. Asara
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Division of Signal Transduction and Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Sarinnapha M. Vasunilashorn
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tamara G. Fong
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA 02131, USA
| | - Sharon K. Inouye
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA 02131, USA
| | - Long H. Ngo
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Edward R. Marcantonio
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Zhongcong Xie
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Towia A. Libermann
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.)
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Hasan H. Otu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
11
|
Deng J, Belyanskaya S, Prabhu N, Arico-Muendel C, Deng H, Phelps CB, Israel DI, Yang H, Boyer J, Franklin GJ, Yap JL, Lind KE, Tsai CH, Donahue C, Summerfield JD. Profiling cells with DELs: Small molecule fingerprinting of cell surfaces. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100171. [PMID: 38917882 DOI: 10.1016/j.slasd.2024.100171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
DNA-encoded small molecule library technology has recently emerged as a new paradigm for identifying ligands against drug targets. To date, it has been used to identify ligands against targets that are soluble or overexpressed on cell surfaces. Here, we report applying cell-based selection methods to profile surfaces of mouse C2C12 myoblasts and myotube cells in an unbiased, target agnostic manner. A panel of on-DNA compounds were identified and confirmed for cell binding selectivity. We optimized the cell selection protocol and employed a novel data analysis method to identify cell selective ligands against a panel of human B and T lymphocytes. We discuss the generality of using this workflow for DNA encoded small molecule library selection and data analysis against different cell types, and the feasibility of applying this method to profile cell surfaces for biomarker and target identification.
Collapse
Affiliation(s)
- Jason Deng
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Svetlana Belyanskaya
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Ninad Prabhu
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | | | - Hongfeng Deng
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Christopher B Phelps
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - David I Israel
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Hongfang Yang
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Joseph Boyer
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - G Joseph Franklin
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Jeremy L Yap
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Kenneth E Lind
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Ching-Hsuan Tsai
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Christine Donahue
- GSK Molecular Modalities Discovery, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | | |
Collapse
|
12
|
Karpov OA, Stotland A, Raedschelders K, Chazarin B, Ai L, Murray CI, Van Eyk JE. Proteomics of the heart. Physiol Rev 2024; 104:931-982. [PMID: 38300522 PMCID: PMC11381016 DOI: 10.1152/physrev.00026.2023] [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: 07/03/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Mass spectrometry-based proteomics is a sophisticated identification tool specializing in portraying protein dynamics at a molecular level. Proteomics provides biologists with a snapshot of context-dependent protein and proteoform expression, structural conformations, dynamic turnover, and protein-protein interactions. Cardiac proteomics can offer a broader and deeper understanding of the molecular mechanisms that underscore cardiovascular disease, and it is foundational to the development of future therapeutic interventions. This review encapsulates the evolution, current technologies, and future perspectives of proteomic-based mass spectrometry as it applies to the study of the heart. Key technological advancements have allowed researchers to study proteomes at a single-cell level and employ robot-assisted automation systems for enhanced sample preparation techniques, and the increase in fidelity of the mass spectrometers has allowed for the unambiguous identification of numerous dynamic posttranslational modifications. Animal models of cardiovascular disease, ranging from early animal experiments to current sophisticated models of heart failure with preserved ejection fraction, have provided the tools to study a challenging organ in the laboratory. Further technological development will pave the way for the implementation of proteomics even closer within the clinical setting, allowing not only scientists but also patients to benefit from an understanding of protein interplay as it relates to cardiac disease physiology.
Collapse
Affiliation(s)
- Oleg A Karpov
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Aleksandr Stotland
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Blandine Chazarin
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Lizhuo Ai
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Christopher I Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| |
Collapse
|
13
|
Donovan MG, Eduthan NP, Smith KP, Britton EC, Lyford HR, Araya P, Granrath RE, Waugh KA, Enriquez Estrada B, Rachubinski AL, Sullivan KD, Galbraith MD, Espinosa JM. Variegated overexpression of chromosome 21 genes reveals molecular and immune subtypes of Down syndrome. Nat Commun 2024; 15:5473. [PMID: 38942750 PMCID: PMC11213896 DOI: 10.1038/s41467-024-49781-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
Individuals with Down syndrome, the genetic condition caused by trisomy 21, exhibit strong inter-individual variability in terms of developmental phenotypes and diagnosis of co-occurring conditions. The mechanisms underlying this variable developmental and clinical presentation await elucidation. We report an investigation of human chromosome 21 gene overexpression in hundreds of research participants with Down syndrome, which led to the identification of two major subsets of co-expressed genes. Using clustering analyses, we identified three main molecular subtypes of trisomy 21, based on differential overexpression patterns of chromosome 21 genes. We subsequently performed multiomics comparative analyses among subtypes using whole blood transcriptomes, plasma proteomes and metabolomes, and immune cell profiles. These efforts revealed strong heterogeneity in dysregulation of key pathophysiological processes across the three subtypes, underscored by differential multiomics signatures related to inflammation, immunity, cell growth and proliferation, and metabolism. We also observed distinct patterns of immune cell changes across subtypes. These findings provide insights into the molecular heterogeneity of trisomy 21 and lay the foundation for the development of personalized medicine approaches for the clinical management of Down syndrome.
Collapse
Affiliation(s)
- Micah G Donovan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Neetha P Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Eleanor C Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Hannah R Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Katherine A Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Belinda Enriquez Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, USA.
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, USA.
| |
Collapse
|
14
|
Whiteaker JR, Zhao L, Schoenherr RM, Huang D, Kennedy JJ, Ivey RG, Lin C, Lorentzen TD, Colantonio S, Caceres TW, Roberts RR, Knotts JG, Reading JJ, Perry CD, Garcia-Buntley SS, Bocik W, Hewitt SM, Paulovich AG. Characterization of an expanded set of assays for immunomodulatory proteins using targeted mass spectrometry. Sci Data 2024; 11:682. [PMID: 38918394 PMCID: PMC11199596 DOI: 10.1038/s41597-024-03467-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: 02/01/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024] Open
Abstract
Immunotherapies are revolutionizing cancer care, but many patients do not achieve durable responses and immune-related adverse events are difficult to predict. Quantifying the hundreds of proteins involved in cancer immunity has the potential to provide biomarkers to monitor and predict tumor response. We previously developed robust, multiplexed quantitative assays for immunomodulatory proteins using targeted mass spectrometry, providing measurements that can be performed reproducibly and harmonized across laboratories. Here, we expand upon those efforts in presenting data from a multiplexed immuno-oncology (IO)-3 assay panel targeting 43 peptides representing 39 immune- and inflammation-related proteins. A suite of novel monoclonal antibodies was generated as assay reagents, and the fully characterized antibodies are made available as a resource to the community. The publicly available dataset contains complete characterization of the assay performance, as well as the mass spectrometer parameters and reagent information necessary for implementation of the assay. Quantification of the proteins will provide benefit to correlative studies in clinical trials, identification of new biomarkers, and improve understanding of the immune response in cancer.
Collapse
Affiliation(s)
- Jeffrey R Whiteaker
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lei Zhao
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Regine M Schoenherr
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dongqing Huang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jacob J Kennedy
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Richard G Ivey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenwei Lin
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Travis D Lorentzen
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Simona Colantonio
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tessa W Caceres
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Rhonda R Roberts
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Joseph G Knotts
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Joshua J Reading
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Candice D Perry
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Sandra S Garcia-Buntley
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - William Bocik
- Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stephen M Hewitt
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Amanda G Paulovich
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
15
|
Kim H, Bedsaul-Fryer JR, Schulze KJ, Sincerbeaux G, Baker S, Rebholz CM, Wu LSF, Gogain J, Cuddeback L, Yager JD, De Luca LM, Siddiqua TJ, West KP. An Early Gestation Plasma Inflammasome in Rural Bangladeshi Women. Biomolecules 2024; 14:736. [PMID: 39062451 PMCID: PMC11274825 DOI: 10.3390/biom14070736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Circulating α1-acid glycoprotein (AGP) and C-reactive protein (CRP) are commonly measured to assess inflammation, but these biomarkers fail to reveal the complex molecular biology of inflammation. We mined the maternal plasma proteome to detect proteins that covary with AGP and CRP. In 435 gravida predominantly in <12-week gestation, we correlated the relative quantification of plasma proteins assessed via a multiplexed aptamer assay (SOMAScan®) with AGP and CRP, quantified by immunoassay. We defined a plasma inflammasome as protein correlates meeting a false discovery rate <0.05. We examined potential pathways using principal component analysis. A total of 147 and 879 of 6431 detected plasma proteins correlated with AGP and CRP, respectively, of which 61 overlapped with both biomarkers. Positive correlates included serum amyloid, complement, interferon-induced, and immunoregulatory proteins. Negative correlates were micronutrient and lipid transporters and pregnancy-related anabolic proteins. The principal components (PCs) of AGP were dominated by negatively correlated anabolic proteins associated with gestational homeostasis, angiogenesis, and neurogenesis. The PCs of CRP were more diverse in function, reflecting cell surface and adhesion, embryogenic, and intracellular and extra-hepatic tissue leakage proteins. The plasma proteome of AGP or CRP reveals wide proteomic variation associated with early gestational inflammation, suggesting mechanisms and pathways that merit future research.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA 98105, USA
| | - Jacquelyn R. Bedsaul-Fryer
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20850, USA
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kerry J. Schulze
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Gwen Sincerbeaux
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Sarah Baker
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Lee SF Wu
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | - James D. Yager
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Luigi M. De Luca
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Keith P. West
- Department of International Health (Human Nutrition), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
16
|
Yao P, Iona A, Pozarickij A, Said S, Wright N, Lin K, Millwood I, Fry H, Kartsonaki C, Mazidi M, Chen Y, Bragg F, Liu B, Yang L, Liu J, Avery D, Schmidt D, Sun D, Pei P, Lv J, Yu C, Hill M, Bennett D, Walters R, Li L, Clarke R, Du H, Chen Z. Proteomic Analyses in Diverse Populations Improved Risk Prediction and Identified New Drug Targets for Type 2 Diabetes. Diabetes Care 2024; 47:1012-1019. [PMID: 38623619 PMCID: PMC7615965 DOI: 10.2337/dc23-2145] [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: 11/13/2023] [Accepted: 03/09/2024] [Indexed: 04/17/2024]
Abstract
OBJECTIVE Integrated analyses of plasma proteomics and genetic data in prospective studies can help assess the causal relevance of proteins, improve risk prediction, and discover novel protein drug targets for type 2 diabetes (T2D). RESEARCH DESIGN AND METHODS We measured plasma levels of 2,923 proteins using Olink Explore among ∼2,000 randomly selected participants from China Kadoorie Biobank (CKB) without prior diabetes at baseline. Cox regression assessed associations of individual protein with incident T2D (n = 92 cases). Proteomic-based risk models were developed with discrimination, calibration, reclassification assessed using area under the curve (AUC), calibration plots, and net reclassification index (NRI), respectively. Two-sample Mendelian randomization (MR) analyses using cis-protein quantitative trait loci identified in a genome-wide association study of CKB and UK Biobank for specific proteins were conducted to assess their causal relevance for T2D, along with colocalization analyses to examine shared causal variants between proteins and T2D. RESULTS Overall, 33 proteins were significantly associated (false discovery rate <0.05) with risk of incident T2D, including IGFBP1, GHR, and amylase. The addition of these 33 proteins to a conventional risk prediction model improved AUC from 0.77 (0.73-0.82) to 0.88 (0.85-0.91) and NRI by 38%, with predicted risks well calibrated with observed risks. MR analyses provided support for the causal relevance for T2D of ENTR1, LPL, and PON3, with replication of ENTR1 and LPL in Europeans using different genetic instruments. Moreover, colocalization analyses showed strong evidence (pH4 > 0.6) of shared genetic variants of LPL and PON3 with T2D. CONCLUSIONS Proteomic analyses in Chinese adults identified novel associations of multiple proteins with T2D with strong genetic evidence supporting their causal relevance and potential as novel drug targets for prevention and treatment of T2D.
Collapse
Affiliation(s)
- Pang Yao
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Andri Iona
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Alfred Pozarickij
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Saredo Said
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Neil Wright
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Kuang Lin
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Iona Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Hannah Fry
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Mohsen Mazidi
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Fiona Bragg
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Bowen Liu
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Junxi Liu
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel Avery
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dan Schmidt
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Pei Pei
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Michael Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Derrick Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Huaidong Du
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
17
|
Coorssen JR, Padula MP. Proteomics-The State of the Field: The Definition and Analysis of Proteomes Should Be Based in Reality, Not Convenience. Proteomes 2024; 12:14. [PMID: 38651373 PMCID: PMC11036260 DOI: 10.3390/proteomes12020014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
With growing recognition and acknowledgement of the genuine complexity of proteomes, we are finally entering the post-proteogenomic era. Routine assessment of proteomes as inferred correlates of gene sequences (i.e., canonical 'proteins') cannot provide the necessary critical analysis of systems-level biology that is needed to understand underlying molecular mechanisms and pathways or identify the most selective biomarkers and therapeutic targets. These critical requirements demand the analysis of proteomes at the level of proteoforms/protein species, the actual active molecular players. Currently, only highly refined integrated or integrative top-down proteomics (iTDP) enables the analytical depth necessary to provide routine, comprehensive, and quantitative proteome assessments across the widest range of proteoforms inherent to native systems. Here we provide a broad perspective of the field, taking in historical and current realities, to establish a more balanced understanding of where the field has come from (in particular during the ten years since Proteomes was launched), current issues, and how things likely need to proceed if necessary deep proteome analyses are to succeed. We base this in our firm belief that the best proteomic analyses reflect, as closely as possible, the native sample at the moment of sampling. We also seek to emphasise that this and future analytical approaches are likely best based on the broad recognition and exploitation of the complementarity of currently successful approaches. This also emphasises the need to continuously evaluate and further optimize established approaches, to avoid complacency in thinking and expectations but also to promote the critical and careful development and introduction of new approaches, most notably those that address proteoforms. Above all, we wish to emphasise that a rigorous focus on analytical quality must override current thinking that largely values analytical speed; the latter would certainly be nice, if only proteoforms could thus be effectively, routinely, and quantitatively assessed. Alas, proteomes are composed of proteoforms, not molecular species that can be amplified or that directly mirror genes (i.e., 'canonical'). The problem is hard, and we must accept and address it as such, but the payoff in playing this longer game of rigorous deep proteome analyses is the promise of far more selective biomarkers, drug targets, and truly personalised or even individualised medicine.
Collapse
Affiliation(s)
- Jens R. Coorssen
- Department of Biological Sciences, Faculty of Mathematics and Science, Brock University, St. Catharines, ON L2S 3A1, Canada
- Institute for Globally Distributed Open Research and Education (IGDORE), St. Catharines, ON L2N 4X2, Canada
| | - Matthew P. Padula
- School of Life Sciences and Proteomics, Lipidomics and Metabolomics Core Facility, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
18
|
Gedik H, Peterson R, Chatzinakos C, Dozmorov MG, Vladimirov V, Riley BP, Bacanu SA. A novel multi-omics mendelian randomization method for gene set enrichment and its application to psychiatric disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.14.24305811. [PMID: 38699366 PMCID: PMC11065030 DOI: 10.1101/2024.04.14.24305811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Genome-wide association studies (GWAS) of psychiatric disorders (PD) yield numerous loci with significant signals, but often do not implicate specific genes. Because GWAS risk loci are enriched in expression/protein/methylation quantitative loci (e/p/mQTL, hereafter xQTL), transcriptome/proteome/methylome-wide association studies (T/P/MWAS, hereafter XWAS) that integrate xQTL and GWAS information, can link GWAS signals to effects on specific genes. To further increase detection power, gene signals are aggregated within relevant gene sets (GS) by performing gene set enrichment (GSE) analyses. Often GSE methods test for enrichment of "signal" genes in curated GS while overlooking their linkage disequilibrium (LD) structure, allowing for the possibility of increased false positive rates. Moreover, no GSE tool uses xQTL information to perform mendelian randomization (MR) analysis. To make causal inference on association between PD and GS, we develop a novel MR GSE (MR-GSE) procedure. First, we generate a "synthetic" GWAS for each MSigDB GS by aggregating summary statistics for x-level (mRNA, protein or DNA methylation (DNAm) levels) from the largest xQTL studies available) of genes in a GS. Second, we use synthetic GS GWAS as exposure in a generalized summary-data-based-MR analysis of complex trait outcomes. We applied MR-GSE to GWAS of nine important PD. When applied to the underpowered opioid use disorder GWAS, none of the four analyses yielded any signals, which suggests a good control of false positive rates. For other PD, MR-GSE greatly increased the detection of GO terms signals (2,594) when compared to the commonly used (non-MR) GSE method (286). Some of the findings might be easier to adapt for treatment, e.g., our analyses suggest modest positive effects for supplementation with certain vitamins and/or omega-3 for schizophrenia, bipolar and major depression disorder patients. Similar to other MR methods, when applying MR-GSE researchers should be mindful of the confounding effects of horizontal pleiotropy on statistical inference.
Collapse
|
19
|
Xiang Y, Liu J, Chen J, Xiao M, Pei H, Li L. MoS 2-Based Sensor Array for Accurate Identification of Cancer Cells with Ensemble-Modified Aptamers. ACS APPLIED MATERIALS & INTERFACES 2024; 16:15861-15869. [PMID: 38508220 DOI: 10.1021/acsami.3c19159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
In this work, we present an array-based chemical nose sensor that utilizes a set of ensemble-modified aptamer (EMAmer) probes to sense subtle physicochemical changes on the cell surface for cancer cell identification. The EMAmer probes are engineered by domain-selective incorporation of different types and/or copies of positively charged functional groups into DNA scaffolds, and their differential interactions with cancer cells can be transduced through competitive adsorption of fluorophore-labeled EMAmer probes loaded on MoS2 nanosheets. We demonstrate that this MoS2-EMAmer-based sensor array enables rapid and effective discrimination among six types of cancer cells and their mixtures with a concentration of 104 cells within 60 min, achieving a 94.4% accuracy in identifying blinded unknown cell samples. The established MoS2-EMAmer sensing platform is anticipated to show significant promise in the advancement of cancer diagnostics.
Collapse
Affiliation(s)
- Ying Xiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Jingjing Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Jing Chen
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Hao Pei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Li Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| |
Collapse
|
20
|
Christopoulos P, Harel M, McGregor K, Brody Y, Puzanov I, Bar J, Elon Y, Sela I, Yellin B, Lahav C, Raveh S, Reiner-Benaim A, Reinmuth N, Nechushtan H, Farrugia D, Bustinza-Linares E, Lou Y, Leibowitz R, Kamer I, Zer Kuch A, Moskovitz M, Levy-Barda A, Koch I, Lotem M, Katzenelson R, Agbarya A, Price G, Cheley H, Abu-Amna M, Geldart T, Gottfried M, Tepper E, Polychronis A, Wolf I, Dicker AP, Carbone DP, Gandara DR. Plasma Proteome-Based Test for First-Line Treatment Selection in Metastatic Non-Small Cell Lung Cancer. JCO Precis Oncol 2024; 8:e2300555. [PMID: 38513170 PMCID: PMC10965206 DOI: 10.1200/po.23.00555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/15/2023] [Accepted: 01/25/2024] [Indexed: 03/23/2024] Open
Abstract
PURPOSE Current guidelines for the management of metastatic non-small cell lung cancer (NSCLC) without driver mutations recommend checkpoint immunotherapy with PD-1/PD-L1 inhibitors, either alone or in combination with chemotherapy. This approach fails to account for individual patient variability and host immune factors and often results in less-than-ideal outcomes. To address the limitations of the current guidelines, we developed and subsequently blindly validated a machine learning algorithm using pretreatment plasma proteomic profiles for personalized treatment decisions. PATIENTS AND METHODS We conducted a multicenter observational trial (ClinicalTrials.gov identifier: NCT04056247) of patients undergoing PD-1/PD-L1 inhibitor-based therapy (n = 540) and an additional patient cohort receiving chemotherapy (n = 85) who consented to pretreatment plasma and clinical data collection. Plasma proteome profiling was performed using SomaScan Assay v4.1. RESULTS Our test demonstrates a strong association between model output and clinical benefit (CB) from PD-1/PD-L1 inhibitor-based treatments, evidenced by high concordance between predicted and observed CB (R2 = 0.98, P < .001). The test categorizes patients as either PROphet-positive or PROphet-negative and further stratifies patient outcomes beyond PD-L1 expression levels. The test successfully differentiates between PROphet-negative patients exhibiting high tumor PD-L1 levels (≥50%) who have enhanced overall survival when treated with a combination of immunotherapy and chemotherapy compared with immunotherapy alone (hazard ratio [HR], 0.23 [95% CI, 0.1 to 0.51], P = .0003). By contrast, PROphet-positive patients show comparable outcomes when treated with immunotherapy alone or in combination with chemotherapy (HR, 0.78 [95% CI, 0.42 to 1.44], P = .424). CONCLUSION Plasma proteome-based testing of individual patients, in combination with standard PD-L1 testing, distinguishes patient subsets with distinct differences in outcomes from PD-1/PD-L1 inhibitor-based therapies. These data suggest that this approach can improve the precision of first-line treatment for metastatic NSCLC.
Collapse
Affiliation(s)
- Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital and National Center for Tumor Diseases, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | | | | | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- The Roswell Park Comprehensive Cancer Center Data Bank and BioRepository
| | - Jair Bar
- Institute of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | | | | | | | | | | | - Anat Reiner-Benaim
- Department of Epidemiology, Biostatistics and Community Health Sciences, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Niels Reinmuth
- Asklepios Kliniken GmbH, Asklepios Fachkliniken Muenchen, Gauting, Germany
- The German Center for Lung Research (DZL), Munich-Gauting, Germany
| | - Hovav Nechushtan
- Oncology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic School of Medicine, Jacksonville, FL
| | - Raya Leibowitz
- Shamir Medical Center, Oncology Institute, Zerifin, Israel
| | - Iris Kamer
- Institute of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Alona Zer Kuch
- Department of Oncology, Rambam Medical Center, Haifa, Israel
| | - Mor Moskovitz
- Thoracic Cancer Service, Davidoff Cancer Center, Beilinson, Petah Tikva, Israel
| | - Adva Levy-Barda
- Biobank, Department of Pathology, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | - Ina Koch
- Asklepios Kliniken GmbH, Asklepios Fachkliniken Muenchen, Gauting, Germany
| | - Michal Lotem
- Center for Melanoma and Cancer Immunotherapy, Hadassah Hebrew University Medical Center, Sharett Institute of Oncology, Jerusalem, Israel
| | | | - Abed Agbarya
- Institute of Oncology, Bnai Zion Medical Center, Haifa, Israel
| | - Gillian Price
- Department of Medical Oncology, Aberdeen Royal Infirmary NHS Grampian, Aberdeen, United Kingdom
| | | | - Mahmoud Abu-Amna
- Oncology & Hematology Division, Cancer Center, Emek Medical Center, Afula, Israel
| | | | - Maya Gottfried
- Department of Oncology, Meir Medical Center, Kfar-Saba, Israel
| | - Ella Tepper
- Department of Oncology, Assuta Hospital, Tel Aviv, Israel
| | | | - Ido Wolf
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - David P. Carbone
- Comprehensive Cancer Center, Ohio State University, Columbus, OH
| | - David R. Gandara
- Division of Hematology and Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA
| |
Collapse
|
21
|
Thorolfsdottir RB, Jonsdottir AB, Sveinbjornsson G, Aegisdottir HM, Oddsson A, Stefansson OA, Halldorsson GH, Saevarsdottir S, Thorleifsson G, Stefansdottir L, Pedersen OB, Sørensen E, Ghouse J, Raja AA, Zheng C, Silajdzija E, Rand SA, Erikstrup C, Ullum H, Mikkelsen C, Banasik K, Brunak S, Ivarsdottir EV, Sigurdsson A, Beyter D, Sturluson A, Einarsson H, Tragante V, Helgason H, Lund SH, Halldorsson BV, Sigurpalsdottir BD, Olafsson I, Arnar DO, Thorgeirsson G, Knowlton KU, Nadauld LD, Gretarsdottir S, Helgadottir A, Ostrowski SR, Gudbjartssson DF, Jonsdottir I, Bundgaard H, Holm H, Sulem P, Stefansson K. Variants at the Interleukin 1 Gene Locus and Pericarditis. JAMA Cardiol 2024; 9:165-172. [PMID: 38150231 PMCID: PMC10753444 DOI: 10.1001/jamacardio.2023.4820] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/14/2023] [Indexed: 12/28/2023]
Abstract
Importance Recurrent pericarditis is a treatment challenge and often a debilitating condition. Drugs inhibiting interleukin 1 cytokines are a promising new treatment option, but their use is based on scarce biological evidence and clinical trials of modest sizes, and the contributions of innate and adaptive immune processes to the pathophysiology are incompletely understood. Objective To use human genomics, transcriptomics, and proteomics to shed light on the pathogenesis of pericarditis. Design, Setting, and Participants This was a meta-analysis of genome-wide association studies of pericarditis from 5 countries. Associations were examined between the pericarditis-associated variants and pericarditis subtypes (including recurrent pericarditis) and secondary phenotypes. To explore mechanisms, associations with messenger RNA expression (cis-eQTL), plasma protein levels (pQTL), and CpG methylation of DNA (ASM-QTL) were assessed. Data from Iceland (deCODE genetics, 1983-2020), Denmark (Copenhagen Hospital Biobank/Danish Blood Donor Study, 1977-2022), the UK (UK Biobank, 1953-2021), the US (Intermountain, 1996-2022), and Finland (FinnGen, 1970-2022) were included. Data were analyzed from September 2022 to August 2023. Exposure Genotype. Main Outcomes and Measures Pericarditis. Results In this genome-wide association study of 4894 individuals with pericarditis (mean [SD] age at diagnosis, 51.4 [17.9] years, 2734 [67.6%] male, excluding the FinnGen cohort), associations were identified with 2 independent common intergenic variants at the interleukin 1 locus on chromosome 2q14. The lead variant was rs12992780 (T) (effect allele frequency [EAF], 31%-40%; odds ratio [OR], 0.83; 95% CI, 0.79-0.87; P = 6.67 × 10-16), downstream of IL1B and the secondary variant rs7575402 (A or T) (EAF, 45%-55%; adjusted OR, 0.89; 95% CI, 0.85-0.93; adjusted P = 9.6 × 10-8). The lead variant rs12992780 had a smaller odds ratio for recurrent pericarditis (0.76) than the acute form (0.86) (P for heterogeneity = .03) and rs7575402 was associated with CpG methylation overlapping binding sites of 4 transcription factors known to regulate interleukin 1 production: PU.1 (encoded by SPI1), STAT1, STAT3, and CCAAT/enhancer-binding protein β (encoded by CEBPB). Conclusions and Relevance This study found an association between pericarditis and 2 independent sequence variants at the interleukin 1 gene locus. This finding has the potential to contribute to development of more targeted and personalized therapy of pericarditis with interleukin 1-blocking drugs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gisli H. Halldorsson
- deCODE genetics, Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Saedis Saevarsdottir
- deCODE genetics, Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Medicine, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | | | | | - Ole B. Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jonas Ghouse
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Laboratory for Molecular Cardiology, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Anna Axelsson Raja
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Chaoqun Zheng
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Elvira Silajdzija
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Søren Albertsen Rand
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Laboratory for Molecular Cardiology, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Christina Mikkelsen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Karina Banasik
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | - Hafsteinn Einarsson
- deCODE genetics, Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Hannes Helgason
- deCODE genetics, Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Bjarni V. Halldorsson
- deCODE genetics, Amgen, Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | - Brynja D. Sigurpalsdottir
- deCODE genetics, Amgen, Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali, National University Hospital of Iceland, Reykjavik, Iceland
| | - David O. Arnar
- deCODE genetics, Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Medicine, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Kirk U. Knowlton
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, Utah
- School of Medicine, University of Utah, Salt Lake City
| | - Lincoln D. Nadauld
- Precision Genomics, Intermountain Healthcare, Saint George, Utah
- School of Medicine, Stanford University, Stanford, California
| | | | | | - Sisse R. Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Daniel F. Gudbjartssson
- deCODE genetics, Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Ingileif Jonsdottir
- deCODE genetics, Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Henning Bundgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Hilma Holm
- deCODE genetics, Amgen, Reykjavik, Iceland
| | | | - Kari Stefansson
- deCODE genetics, Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
22
|
Uhlen M, Quake SR. Sequential sequencing by synthesis and the next-generation sequencing revolution. Trends Biotechnol 2023; 41:1565-1572. [PMID: 37482467 DOI: 10.1016/j.tibtech.2023.06.007] [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: 05/08/2023] [Revised: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023]
Abstract
The impact of next-generation sequencing (NGS) cannot be overestimated. The technology has transformed the field of life science, contributing to a dramatic expansion in our understanding of human health and disease and our understanding of biology and ecology. The vast majority of the major NGS systems today are based on the concept of 'sequencing by synthesis' (SBS) with sequential detection of nucleotide incorporation using an engineered DNA polymerase. Based on this strategy, various alternative platforms have been developed, including the use of either native nucleotides or reversible terminators and different strategies for the attachment of DNA to a solid support. In this review, some of the key concepts leading to this remarkable development are discussed.
Collapse
Affiliation(s)
- Mathias Uhlen
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden; Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Stephen R Quake
- Departments of Bioengineering and Applied Physics, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, California, USA, Stanford, CA, USA
| |
Collapse
|
23
|
Sopic M, Vilne B, Gerdts E, Trindade F, Uchida S, Khatib S, Wettinger SB, Devaux Y, Magni P. Multiomics tools for improved atherosclerotic cardiovascular disease management. Trends Mol Med 2023; 29:983-995. [PMID: 37806854 DOI: 10.1016/j.molmed.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023]
Abstract
Multiomics studies offer accurate preventive and therapeutic strategies for atherosclerotic cardiovascular disease (ASCVD) beyond traditional risk factors. By using artificial intelligence (AI) and machine learning (ML) approaches, it is possible to integrate multiple 'omics and clinical data sets into tools that can be utilized for the development of personalized diagnostic and therapeutic approaches. However, currently multiple challenges in data quality, integration, and privacy still need to be addressed. In this opinion, we emphasize that joined efforts, exemplified by the AtheroNET COST Action, have a pivotal role in overcoming the challenges to advance multiomics approaches in ASCVD research, with the aim to foster more precise and effective patient care.
Collapse
Affiliation(s)
- Miron Sopic
- Cardiovascular Research Unit, Department of Precision Health, 1A-B rue Edison, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, 11000, Serbia
| | - Baiba Vilne
- Bioinformatics Laboratory, Rīga Stradiņš University, Rīga, LV-1007, Latvia
| | - Eva Gerdts
- Center for Research on Cardiac Disease in Women, Department of Clinical Science, University of Bergen, Bergen, 5020, Norway
| | - Fábio Trindade
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, 4099-002, Portugal
| | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen, SV, DK-2450, Denmark
| | - Soliman Khatib
- Natural Compounds and Analytical Chemistry Laboratory, MIGAL-Galilee Research Institute, Kiryat Shemona, 11016, Israel; Department of Biotechnology, Tel-Hai College, Upper Galilee 12210, Israel
| | - Stephanie Bezzina Wettinger
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, Malta
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, 1A-B rue Edison, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg.
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via G. Balzaretti 9, 20133 Milano, Italy; IRCCS MultiMedica, Via Milanese 300, 20099 Sesto S. Giovanni, Milan, Italy.
| |
Collapse
|
24
|
Lee D, Rapp V CG, Loureiro J, Patel MT, Mikhailov D, Gusev AI. Decentralized clinical trial design using blood microsampling technology for serum bioanalysis. Bioanalysis 2023; 15:1287-1303. [PMID: 37855231 DOI: 10.4155/bio-2023-0136] [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: 10/20/2023] Open
Abstract
Background: Alternatives to phlebotomy in clinical trials increase options for patients and clinicians by simplifying and increasing accessibility to clinical trials. The authors investigated the technical and logistical considerations of one technology compared with phlebotomy. Methodology: Paired samples were collected from 16 donors via a second-generation serum gel microsampling device and conventional phlebotomy. Microsamples were subject to alternative sample handling conditions and were evaluated for quality, clinical testing and proteome profiling. Results: Timely centrifugation of blood serum microsamples largely preserved analyte stability. Conclusion: Centrifugation timing of serum microsamples impacts the quality of specific clinical chemistry and protein biomarkers. Microsampling devices with remote centrifugation and refrigerated shipping can decrease patient burden, expand clinical trial populations and aid clinical decisions.
Collapse
Affiliation(s)
- Dana Lee
- Biomarker Development, Novartis Institutes for BioMedical Research, 220 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Charles G Rapp V
- Biomarker & Bioanalytical Science & Technology, Takeda Pharmaceutical Company, 40 Landsdowne St., Cambridge, MA 02139, USA
| | - Joseph Loureiro
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, 250 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Michael T Patel
- Biomarker Development, Novartis Institutes for BioMedical Research, 220 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Dmitri Mikhailov
- Biomarker Development, Novartis Institutes for BioMedical Research, 220 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Arkady I Gusev
- Biomarker Development, Novartis Institutes for BioMedical Research, 220 Massachusetts Ave., Cambridge, MA 02139, USA
| |
Collapse
|
25
|
Dowling P, Swandulla D, Ohlendieck K. Mass Spectrometry-Based Proteomic Technology and Its Application to Study Skeletal Muscle Cell Biology. Cells 2023; 12:2560. [PMID: 37947638 PMCID: PMC10649384 DOI: 10.3390/cells12212560] [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: 10/06/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Voluntary striated muscles are characterized by a highly complex and dynamic proteome that efficiently adapts to changed physiological demands or alters considerably during pathophysiological dysfunction. The skeletal muscle proteome has been extensively studied in relation to myogenesis, fiber type specification, muscle transitions, the effects of physical exercise, disuse atrophy, neuromuscular disorders, muscle co-morbidities and sarcopenia of old age. Since muscle tissue accounts for approximately 40% of body mass in humans, alterations in the skeletal muscle proteome have considerable influence on whole-body physiology. This review outlines the main bioanalytical avenues taken in the proteomic characterization of skeletal muscle tissues, including top-down proteomics focusing on the characterization of intact proteoforms and their post-translational modifications, bottom-up proteomics, which is a peptide-centric method concerned with the large-scale detection of proteins in complex mixtures, and subproteomics that examines the protein composition of distinct subcellular fractions. Mass spectrometric studies over the last two decades have decisively improved our general cell biological understanding of protein diversity and the heterogeneous composition of individual myofibers in skeletal muscles. This detailed proteomic knowledge can now be integrated with findings from other omics-type methodologies to establish a systems biological view of skeletal muscle function.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Dieter Swandulla
- Institute of Physiology, Faculty of Medicine, University of Bonn, D53115 Bonn, Germany;
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
26
|
Gonçalves M, Khera T, Otu HH, Narayanan S, Dillon ST, Shanker A, Gu X, Jung Y, Ngo LH, Marcantonio ER, Libermann TA, Subramaniam B. Multivariable model of postoperative delirium in cardiac surgery patients: proteomic and demographic contributions. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.30.23289741. [PMID: 37333093 PMCID: PMC10274980 DOI: 10.1101/2023.05.30.23289741] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Delirium following cardiac surgery is common, morbid, and costly, but may be prevented with risk stratification and targeted intervention. Preoperative protein signatures may identify patients at increased risk for worse postoperative outcomes, including delirium. In this study, we aimed to identify plasma protein biomarkers and develop a predictive model for postoperative delirium in older patients undergoing cardiac surgery, while also uncovering possible pathophysiological mechanisms. Methods SOMAscan analysis of 1,305 proteins in the plasma from 57 older adults undergoing cardiac surgery requiring cardiopulmonary bypass was conducted to define delirium-specific protein signatures at baseline (PREOP) and postoperative day 2 (POD2). Selected proteins were validated in 115 patients using the ELLA multiplex immunoassay platform. Proteins were combined with clinical and demographic variables to build multivariable models that estimate the risk of postoperative delirium and bring light to the underlying pathophysiology. Results A total of 115 and 85 proteins from SOMAscan analyses were found altered in delirious patients at PREOP and POD2, respectively (p<0.05). Using four criteria including associations with surgery, delirium, and biological plausibility, 12 biomarker candidates (Tukey's fold change (|tFC|)>1.4, Benjamini-Hochberg (BH)-p<0.01) were selected for ELLA multiplex validation. Eight proteins were significantly altered at PREOP, and seven proteins at POD2 (p<0.05), in patients who developed postoperative delirium compared to non-delirious patients. Statistical analyses of model fit resulted in the selection of a combination of age, sex, and three proteins (angiopoietin-2 (ANGPT2); C-C motif chemokine 5 (CCL5); and metalloproteinase inhibitor 1 (TIMP1); AUC=0.829) as the best performing predictive model for delirium at PREOP. The delirium-associated proteins identified as biomarker candidates are involved with inflammation, glial dysfunction, vascularization, and hemostasis, highlighting the multifactorial pathophysiology of delirium. Conclusion Our study proposes a model of postoperative delirium that includes a combination of older age, female sex, and altered levels of three proteins. Our results support the identification of patients at higher risk of developing postoperative delirium after cardiac surgery and provide insights on the underlying pathophysiology. ClinicalTrials.gov ( NCT02546765 ).
Collapse
|
27
|
Krauze AV, Zhao Y, Li MC, Shih J, Jiang W, Tasci E, Cooley Zgela T, Sproull M, Mackey M, Shankavaram U, Tofilon P, Camphausen K. Revisiting Concurrent Radiation Therapy, Temozolomide, and the Histone Deacetylase Inhibitor Valproic Acid for Patients with Glioblastoma-Proteomic Alteration and Comparison Analysis with the Standard-of-Care Chemoirradiation. Biomolecules 2023; 13:1499. [PMID: 37892181 PMCID: PMC10604983 DOI: 10.3390/biom13101499] [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: 08/24/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common brain tumor with an overall survival (OS) of less than 30% at two years. Valproic acid (VPA) demonstrated survival benefits documented in retrospective and prospective trials, when used in combination with chemo-radiotherapy (CRT). PURPOSE The primary goal of this study was to examine if the differential alteration in proteomic expression pre vs. post-completion of concurrent chemoirradiation (CRT) is present with the addition of VPA as compared to standard-of-care CRT. The second goal was to explore the associations between the proteomic alterations in response to VPA/RT/TMZ correlated to patient outcomes. The third goal was to use the proteomic profile to determine the mechanism of action of VPA in this setting. MATERIALS AND METHODS Serum obtained pre- and post-CRT was analyzed using an aptamer-based SOMAScan® proteomic assay. Twenty-nine patients received CRT plus VPA, and 53 patients received CRT alone. Clinical data were obtained via a database and chart review. Tests for differences in protein expression changes between radiation therapy (RT) with or without VPA were conducted for individual proteins using two-sided t-tests, considering p-values of <0.05 as significant. Adjustment for age, sex, and other clinical covariates and hierarchical clustering of significant differentially expressed proteins was carried out, and Gene Set Enrichment analyses were performed using the Hallmark gene sets. Univariate Cox proportional hazards models were used to test the individual protein expression changes for an association with survival. The lasso Cox regression method and 10-fold cross-validation were employed to test the combinations of expression changes of proteins that could predict survival. Predictiveness curves were plotted for significant proteins for VPA response (p-value < 0.005) to show the survival probability vs. the protein expression percentiles. RESULTS A total of 124 proteins were identified pre- vs. post-CRT that were differentially expressed between the cohorts who received CRT plus VPA and those who received CRT alone. Clinical factors did not confound the results, and distinct proteomic clustering in the VPA-treated population was identified. Time-dependent ROC curves for OS and PFS for landmark times of 20 months and 6 months, respectively, revealed AUC of 0.531, 0.756, 0.774 for OS and 0.535, 0.723, 0.806 for PFS for protein expression, clinical factors, and the combination of protein expression and clinical factors, respectively, indicating that the proteome can provide additional survival risk discrimination to that already provided by the standard clinical factors with a greater impact on PFS. Several proteins of interest were identified. Alterations in GALNT14 (increased) and CCL17 (decreased) (p = 0.003 and 0.003, respectively, FDR 0.198 for both) were associated with an improvement in both OS and PFS. The pre-CRT protein expression revealed 480 proteins predictive for OS and 212 for PFS (p < 0.05), of which 112 overlapped between OS and PFS. However, FDR-adjusted p values were high, with OS (the smallest p value of 0.586) and PFS (the smallest p value of 0.998). The protein PLCD3 had the lowest p-value (p = 0.002 and 0.0004 for OS and PFS, respectively), and its elevation prior to CRT predicted superior OS and PFS with VPA administration. Cancer hallmark genesets associated with proteomic alteration observed with the administration of VPA aligned with known signal transduction pathways of this agent in malignancy and non-malignancy settings, and GBM signaling, and included epithelial-mesenchymal transition, hedgehog signaling, Il6/JAK/STAT3, coagulation, NOTCH, apical junction, xenobiotic metabolism, and complement signaling. CONCLUSIONS Differential alteration in proteomic expression pre- vs. post-completion of concurrent chemoirradiation (CRT) is present with the addition of VPA. Using pre- vs. post-data, prognostic proteins emerged in the analysis. Using pre-CRT data, potentially predictive proteins were identified. The protein signals and hallmark gene sets associated with the alteration in the proteome identified between patients who received VPA and those who did not, align with known biological mechanisms of action of VPA and may allow for the identification of novel biomarkers associated with outcomes that can help advance the study of VPA in future prospective trials.
Collapse
Affiliation(s)
- Andra V. Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Yingdong Zhao
- Computational and Systems Biology Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, USA; (Y.Z.); (M.-C.L.); (J.S.)
| | - Ming-Chung Li
- Computational and Systems Biology Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, USA; (Y.Z.); (M.-C.L.); (J.S.)
| | - Joanna Shih
- Computational and Systems Biology Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, USA; (Y.Z.); (M.-C.L.); (J.S.)
| | - Will Jiang
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Erdal Tasci
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Theresa Cooley Zgela
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Mary Sproull
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Megan Mackey
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Uma Shankavaram
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Philip Tofilon
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Kevin Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| |
Collapse
|
28
|
Kussmann M. Mass spectrometry as a lens into molecular human nutrition and health. EUROPEAN JOURNAL OF MASS SPECTROMETRY (CHICHESTER, ENGLAND) 2023; 29:370-379. [PMID: 37587732 DOI: 10.1177/14690667231193555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Mass spectrometry (MS) has developed over the last decades into the most informative and versatile analytical technology in molecular and structural biology (). The platform enables discovery, identification, and characterisation of non-volatile biomolecules, such as proteins, peptides, DNA, RNA, nutrients, metabolites, and lipids at both speed and scale and can elucidate their interactions and effects. The versatility, robustness, and throughput have rendered MS a major research and development platform in molecular human health and biomedical science. More recently, MS has also been established as the central tool for 'Molecular Nutrition', enabling comprehensive and rapid identification and characterisation of macro- and micronutrients, bioactives, and other food compounds. 'Molecular Nutrition' thereby helps understand bioaccessibility, bioavailability, and bioefficacy of macro- and micronutrients and related health effects. Hence, MS provides a lens through which the fate of nutrients can be monitored along digestion via absorption to metabolism. This in turn provides the bioanalytical foundation for 'Personalised Nutrition' or 'Precision Nutrition' in which design and development of diets and nutritional products is tailored towards consumer and patient groups sharing similar genetic and environmental predisposition, health/disease conditions and lifestyles, and/or objectives of performance and wellbeing. The next level of integrated nutrition science is now being built as 'Systems Nutrition' where public and personal health data are correlated with life condition and lifestyle factors, to establish directional relationships between nutrition, lifestyle, environment, and health, eventually translating into science-based public and personal heath recommendations and actions. This account provides a condensed summary of the contributions of MS to a precise, quantitative, and comprehensive nutrition and health science and sketches an outlook on its future role in this fascinating and relevant field.
Collapse
Affiliation(s)
- Martin Kussmann
- Abteilung Wissenschaft, Kompetenzzentrum für Ernährung (KErn), Germany
- Kussmann Biotech GmbH, Germany
| |
Collapse
|
29
|
Yao P, Iona A, Kartsonaki C, Said S, Wright N, Lin K, Pozarickij A, Millwood I, Fry H, Mazidi M, Chen Y, Du H, Bennett D, Avery D, Schmidt D, Pei P, Lv J, Yu C, Hill M, Chen J, Peto R, Walters R, Collins R, Li L, Clarke R, Chen Z. Conventional and genetic associations of adiposity with 1463 proteins in relatively lean Chinese adults. Eur J Epidemiol 2023; 38:1089-1103. [PMID: 37676424 PMCID: PMC10570181 DOI: 10.1007/s10654-023-01038-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/28/2023] [Indexed: 09/08/2023]
Abstract
Adiposity is associated with multiple diseases and traits, but little is known about the causal relevance and mechanisms underlying these associations. Large-scale proteomic profiling, especially when integrated with genetic data, can clarify mechanisms linking adiposity with disease outcomes. We examined the associations of adiposity with plasma levels of 1463 proteins in 3977 Chinese adults, using measured and genetically-instrumented BMI. We further used two-sample bi-directional MR analyses to assess if certain proteins influenced adiposity, along with other (e.g. enrichment) analyses to clarify possible mechanisms underlying the observed associations. Overall, the mean (SD) baseline BMI was 23.9 (3.3) kg/m2, with only 6% being obese (i.e. BMI ≥ 30 kg/m2). Measured and genetically-instrumented BMI was significantly associated at FDR < 0.05 with levels of 1096 (positive/inverse: 826/270) and 307 (positive/inverse: 270/37) proteins, respectively, with FABP4, LEP, IL1RN, LSP1, GOLM2, TNFRSF6B, and ADAMTS15 showing the strongest positive and PON3, NCAN, LEPR, IGFBP2 and MOG showing the strongest inverse genetic associations. These associations were largely linear, in adiposity-to-protein direction, and replicated (> 90%) in Europeans of UKB (mean BMI 27.4 kg/m2). Enrichment analyses of the top > 50 BMI-associated proteins demonstrated their involvement in atherosclerosis, lipid metabolism, tumour progression and inflammation. Two-sample bi-directional MR analyses using cis-pQTLs identified in CKB GWAS found eight proteins (ITIH3, LRP11, SCAMP3, NUDT5, OGN, EFEMP1, TXNDC15, PRDX6) significantly affect levels of BMI, with NUDT5 also showing bi-directional association. The findings among relatively lean Chinese adults identified novel pathways by which adiposity may increase disease risks and novel potential targets for treatment of obesity and obesity-related diseases.
Collapse
Affiliation(s)
- Pang Yao
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Andri Iona
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research 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, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Kuang Lin
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Alfred Pozarickij
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Iona Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research 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, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research 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, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research 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, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Derrick Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research 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, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dan Schmidt
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Pei Pei
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
| | - Jun Lv
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Canqing Yu
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Richard Peto
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Robin Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- Medical Research Council Health Research 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, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Liming Li
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK.
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK.
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
30
|
Vasunilashorn SM, Dillon ST, Marcantonio ER, Libermann TA. Application of Multiple Omics to Understand Postoperative Delirium Pathophysiology in Humans. Gerontology 2023; 69:1369-1384. [PMID: 37722373 PMCID: PMC10711777 DOI: 10.1159/000533789] [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: 01/24/2023] [Accepted: 08/23/2023] [Indexed: 09/20/2023] Open
Abstract
Delirium, an acute change in cognition, is common, morbid, and costly, particularly among hospitalized older adults. Despite growing knowledge of its epidemiology, far less is known about delirium pathophysiology. Initial work understanding delirium pathogenesis has focused on assaying single or a limited subset of molecules or genetic loci. Recent technological advances at the forefront of biomarker and drug target discovery have facilitated application of multiple "omics" approaches aimed to provide a more complete understanding of complex disease processes such as delirium. At its basic level, "omics" involves comparison of genes (genomics, epigenomics), transcripts (transcriptomics), proteins (proteomics), metabolites (metabolomics), or lipids (lipidomics) in biological fluids or tissues obtained from patients who have a certain condition (i.e., delirium) and those who do not. Multi-omics analyses of these various types of molecules combined with machine learning and systems biology enable the discovery of biomarkers, biological pathways, and predictors of delirium, thus elucidating its pathophysiology. This review provides an overview of the most recent omics techniques, their current impact on identifying delirium biomarkers, and future potential in enhancing our understanding of delirium pathogenesis. We summarize challenges in identification of specific biomarkers of delirium and, more importantly, in discovering the mechanisms underlying delirium pathophysiology. Based on mounting evidence, we highlight a heightened inflammatory response as one common pathway in delirium risk and progression, and we suggest other promising biological mechanisms that have recently emerged. Advanced multiple omics approaches coupled with bioinformatics methodologies have great promise to yield important discoveries that will advance delirium research.
Collapse
Affiliation(s)
- Sarinnapha M. Vasunilashorn
- Division of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Simon T. Dillon
- Harvard Medical School, Boston, MA, USA
- Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, BIDMC, Boston, MA, USA
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, BIDMC, Boston, MA, USA
| | - Edward R. Marcantonio
- Division of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Gerontology, Department of Medicine, BIDMC, Boston, MA, USA
| | - Towia A. Libermann
- Harvard Medical School, Boston, MA, USA
- Division of Interdisciplinary Medicine and Biotechnology, Department of Medicine, BIDMC, Boston, MA, USA
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, BIDMC, Boston, MA, USA
| |
Collapse
|
31
|
Dillon ST, Vasunilashorn SM, Otu HH, Ngo L, Fong T, Gu X, Cavallari M, Touroutoglou A, Shafi M, Inouye SK, Xie Z, Marcantonio ER, Libermann TA. Aptamer-Based Proteomics Measuring Preoperative Cerebrospinal Fluid Protein Alterations Associated with Postoperative Delirium. Biomolecules 2023; 13:1395. [PMID: 37759795 PMCID: PMC10526755 DOI: 10.3390/biom13091395] [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: 08/10/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Delirium is a common postoperative complication among older patients with many adverse outcomes. Due to a lack of validated biomarkers, prediction and monitoring of delirium by biological testing is not currently feasible. Circulating proteins in cerebrospinal fluid (CSF) may reflect biological processes causing delirium. Our goal was to discover and investigate candidate protein biomarkers in preoperative CSF that were associated with the development of postoperative delirium in older surgical patients. We employed a nested case-control study design coupled with high multiplex affinity proteomics analysis to measure 1305 proteins in preoperative CSF. Twenty-four matched delirium cases and non-delirium controls were selected from the Healthier Postoperative Recovery (HiPOR) cohort, and the associations between preoperative protein levels and postoperative delirium were assessed using t-test statistics with further analysis by systems biology to elucidate delirium pathophysiology. Proteomics analysis identified 32 proteins in preoperative CSF that significantly associate with delirium (t-test p < 0.05). Due to the limited sample size, these proteins did not remain significant by multiple hypothesis testing using the Benjamini-Hochberg correction and q-value method. Three algorithms were applied to separate delirium cases from non-delirium controls. Hierarchical clustering classified 40/48 case-control samples correctly, and principal components analysis separated 43/48. The receiver operating characteristic curve yielded an area under the curve [95% confidence interval] of 0.91 [0.80-0.97]. Systems biology analysis identified several key pathways associated with risk of delirium: inflammation, immune cell migration, apoptosis, angiogenesis, synaptic depression and neuronal cell death. Proteomics analysis of preoperative CSF identified 32 proteins that might discriminate individuals who subsequently develop postoperative delirium from matched control samples. These proteins are potential candidate biomarkers for delirium and may play a role in its pathophysiology.
Collapse
Affiliation(s)
- Simon T. Dillon
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.); (X.G.)
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
| | - Sarinnapha M. Vasunilashorn
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Divisions of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Hasan H. Otu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA;
| | - Long Ngo
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Divisions of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Tamara Fong
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew Senior Life, Boston, MA 02131, USA;
| | - Xuesong Gu
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.); (X.G.)
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
| | - Michele Cavallari
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Center for Neurological Imaging, Department of Radiology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Alexandra Touroutoglou
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mouhsin Shafi
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Sharon K. Inouye
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew Senior Life, Boston, MA 02131, USA;
- Divisions of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Zhongcong Xie
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Edward R. Marcantonio
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Divisions of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Divisions of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Towia A. Libermann
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.); (X.G.)
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
| |
Collapse
|
32
|
Wang S, Rao Z, Cao R, Blaes AH, Coresh J, Joshu CE, Lehallier B, Lutsey PL, Pankow JS, Sedaghat S, Tang W, Thyagarajan B, Walker KA, Ganz P, Platz EA, Guan W, Prizment A. Development and Characterization of Proteomic Aging Clocks in the Atherosclerosis Risk in Communities (ARIC) Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.06.23295174. [PMID: 37732184 PMCID: PMC10508816 DOI: 10.1101/2023.09.06.23295174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Biological age may be estimated by proteomic aging clocks (PACs). Previous published PACs were constructed either in smaller studies or mainly in White individuals, and they used proteomic measures from only one-time point. In the Atherosclerosis Risk in Communities (ARIC) study of about 12,000 persons followed for 30 years (around 75% White, 25% Black), we created de novo PACs and compared their performance to published PACs at two different time points. We measured 4,712 plasma proteins by SomaScan in 11,761 midlife participants, aged 46-70 years (1990-92), and 5,183 late-life pariticpants, aged 66-90 years (2011-13). All proteins were log2-transformed to correct for skewness. We created de novo PACs by training them against chronological age using elastic net regression in two-thirds of healthy participants in midlife and late life and compared their performance to three published PACs. We estimated age acceleration (by regressing each PAC on chronological age) and its change from midlife to late life. We examined their associations with mortality from all-cause, cardiovascular disease (CVD), cancer, and lower respiratory disease (LRD) using Cox proportional hazards regression in all remaining participants irrespective of health. The model was adjusted for chronological age, smoking, body mass index (BMI), and other confounders. The ARIC PACs had a slightly stronger correlation with chronological age than published PACs in healthy participants at each time point. Associations with mortality were similar for the ARIC and published PACs. For late-life and midlife age acceleration for the ARIC PACs, respectively, hazard ratios (HRs) per one standard deviation were 1.65 and 1.38 (both p<0.001) for all-cause mortality, 1.37 and 1.20 (both p<0.001) for CVD mortality, 1.21 (p=0.03) and 1.04 (p=0.19) for cancer mortality, and 1.46 and 1.68 (both p<0.001) for LRD mortality. For the change in age acceleration, HRs for all-cause, CVD, and LRD mortality were comparable to those observed for late-life age acceleration. The association between the change in age acceleration and cancer mortality was insignificant. In this prospective study, the ARIC and published PACs were similarly associated with an increased risk of mortality and advanced testing in relation to various age-related conditions in future studies is suggested.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Zexi Rao
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Rui Cao
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, MN
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Benoit Lehallier
- Alkahest Inc, San Carlos, CA, United States, Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, Baltimore, MD
| | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco, CA
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| |
Collapse
|
33
|
Zasedateleva OA, Surzhikov SA, Kuznetsova VE, Shershov VE, Barsky VE, Zasedatelev AS, Chudinov AV. Non-Covalent Interactions between dUTP C5-Substituents and DNA Polymerase Decrease PCR Efficiency. Int J Mol Sci 2023; 24:13643. [PMID: 37686447 PMCID: PMC10487964 DOI: 10.3390/ijms241713643] [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: 07/31/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/10/2023] Open
Abstract
The approach based on molecular modeling was developed to study dNTP derivatives characterized by new polymerase-specific properties. For this purpose, the relative efficiency of PCR amplification with modified dUTPs was studied using Taq, Tth, Pfu, Vent, Deep Vent, Vent (exo-), and Deep Vent (exo-) DNA polymerases. The efficiency of PCR amplification with modified dUTPs was compared with the results of molecular modeling using the known 3D structures of KlenTaq polymerase-DNA-dNTP complexes. The dUTPs were C5-modified with bulky functional groups (the Cy5 dye analogs) or lighter aromatic groups. Comparing the experimental data and the results of molecular modeling revealed the decrease in PCR efficiency in the presence of modified dUTPs with an increase in the number of non-covalent bonds between the substituents and the DNA polymerase (about 15% decrease per one extra non-covalent bond). Generalization of the revealed patterns to all the studied polymerases of the A and B families is discussed herein. The number of non-covalent bonds between the substituents and polymerase amino acid residues is proposed to be a potentially variable parameter for regulating enzyme activity.
Collapse
Affiliation(s)
- Olga A. Zasedateleva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov Street, 119991 Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
34
|
Krauze AV, Sierk M, Nguyen T, Chen Q, Yan C, Hu Y, Jiang W, Tasci E, Zgela TC, Sproull M, Mackey M, Shankavaram U, Meerzaman D, Camphausen K. Glioblastoma survival is associated with distinct proteomic alteration signatures post chemoirradiation in a large-scale proteomic panel. Front Oncol 2023; 13:1127645. [PMID: 37637066 PMCID: PMC10448824 DOI: 10.3389/fonc.2023.1127645] [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: 12/19/2022] [Accepted: 06/20/2023] [Indexed: 08/29/2023] Open
Abstract
Background Glioblastomas (GBM) are rapidly progressive, nearly uniformly fatal brain tumors. Proteomic analysis represents an opportunity for noninvasive GBM classification and biological understanding of treatment response. Purpose We analyzed differential proteomic expression pre vs. post completion of concurrent chemoirradiation (CRT) in patient serum samples to explore proteomic alterations and classify GBM by integrating clinical and proteomic parameters. Materials and methods 82 patients with GBM were clinically annotated and serum samples obtained pre- and post-CRT. Serum samples were then screened using the aptamer-based SOMAScan® proteomic assay. Significant traits from uni- and multivariate Cox models for overall survival (OS) were designated independent prognostic factors and principal component analysis (PCA) was carried out. Differential expression of protein signals was calculated using paired t-tests, with KOBAS used to identify associated KEGG pathways. GSEA pre-ranked analysis was employed on the overall list of differentially expressed proteins (DEPs) against the MSigDB Hallmark, GO Biological Process, and Reactome databases with weighted gene correlation network analysis (WGCNA) and Enrichr used to validate pathway hits internally. Results 3 clinical clusters of patients with differential survival were identified. 389 significantly DEPs pre vs. post-treatment were identified, including 284 upregulated and 105 downregulated, representing several pathways relevant to cancer metabolism and progression. The lowest survival group (median OS 13.2 months) was associated with DEPs affiliated with proliferative pathways and exhibiting distinct oppositional response including with respect to radiation therapy related pathways, as compared to better-performing groups (intermediate, median OS 22.4 months; highest, median OS 28.7 months). Opposite signaling patterns across multiple analyses in several pathways (notably fatty acid metabolism, NOTCH, TNFα via NF-κB, Myc target V1 signaling, UV response, unfolded protein response, peroxisome, and interferon response) were distinct between clinical survival groups and supported by WGCNA. 23 proteins were statistically signficant for OS with 5 (NETO2, CST7, SEMA6D, CBLN4, NPS) supported by KM. Conclusion Distinct proteomic alterations with hallmarks of cancer, including progression, resistance, stemness, and invasion, were identified in serum samples obtained from GBM patients pre vs. post CRT and corresponded with clinical survival. The proteome can potentially be employed for glioma classification and biological interrogation of cancer pathways.
Collapse
Affiliation(s)
- Andra Valentina Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Michael Sierk
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology, National Cancer Institute, NIH, Rockville, MD, United States
| | - Trinh Nguyen
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology, National Cancer Institute, NIH, Rockville, MD, United States
| | - Qingrong Chen
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology, National Cancer Institute, NIH, Rockville, MD, United States
| | - Chunhua Yan
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology, National Cancer Institute, NIH, Rockville, MD, United States
| | - Ying Hu
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology, National Cancer Institute, NIH, Rockville, MD, United States
| | - William Jiang
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Erdal Tasci
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Theresa Cooley Zgela
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Mary Sproull
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Megan Mackey
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Uma Shankavaram
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Daoud Meerzaman
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology, National Cancer Institute, NIH, Rockville, MD, United States
| | - Kevin Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
35
|
Ngo D, Pratte KA, Flexeder C, Petersen H, Dang H, Ma Y, Keyes MJ, Gao Y, Deng S, Peterson BD, Farrell LA, Bhambhani VM, Palacios C, Quadir J, Gillenwater L, Xu H, Emson C, Gieger C, Suhre K, Graumann J, Jain D, Conomos MP, Tracy RP, Guo X, Liu Y, Johnson WC, Cornell E, Durda P, Taylor KD, Papanicolaou GJ, Rich SS, Rotter JI, Rennard SI, Curtis JL, Woodruff PG, Comellas AP, Silverman EK, Crapo JD, Larson MG, Vasan RS, Wang TJ, Correa A, Sims M, Wilson JG, Gerszten RE, O’Connor GT, Barr RG, Couper D, Dupuis J, Manichaikul A, O’Neal WK, Tesfaigzi Y, Schulz H, Bowler RP. Systemic Markers of Lung Function and Forced Expiratory Volume in 1 Second Decline across Diverse Cohorts. Ann Am Thorac Soc 2023; 20:1124-1135. [PMID: 37351609 PMCID: PMC10405603 DOI: 10.1513/annalsats.202210-857oc] [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: 10/11/2022] [Accepted: 06/13/2023] [Indexed: 06/24/2023] Open
Abstract
Rationale: Chronic obstructive pulmonary disease (COPD) is a complex disease characterized by airway obstruction and accelerated lung function decline. Our understanding of systemic protein biomarkers associated with COPD remains incomplete. Objectives: To determine what proteins and pathways are associated with impaired pulmonary function in a diverse population. Methods: We studied 6,722 participants across six cohort studies with both aptamer-based proteomic and spirometry data (4,566 predominantly White participants in a discovery analysis and 2,156 African American cohort participants in a validation). In linear regression models, we examined protein associations with baseline forced expiratory volume in 1 second (FEV1) and FEV1/forced vital capacity (FVC). In linear mixed effects models, we investigated the associations of baseline protein levels with rate of FEV1 decline (ml/yr) in 2,777 participants with up to 7 years of follow-up spirometry. Results: We identified 254 proteins associated with FEV1 in our discovery analyses, with 80 proteins validated in the Jackson Heart Study. Novel validated protein associations include kallistatin serine protease inhibitor, growth differentiation factor 2, and tumor necrosis factor-like weak inducer of apoptosis (discovery β = 0.0561, Q = 4.05 × 10-10; β = 0.0421, Q = 1.12 × 10-3; and β = 0.0358, Q = 1.67 × 10-3, respectively). In longitudinal analyses within cohorts with follow-up spirometry, we identified 15 proteins associated with FEV1 decline (Q < 0.05), including elafin leukocyte elastase inhibitor and mucin-associated TFF2 (trefoil factor 2; β = -4.3 ml/yr, Q = 0.049; β = -6.1 ml/yr, Q = 0.032, respectively). Pathways and processes highlighted by our study include aberrant extracellular matrix remodeling, enhanced innate immune response, dysregulation of angiogenesis, and coagulation. Conclusions: In this study, we identify and validate novel biomarkers and pathways associated with lung function traits in a racially diverse population. In addition, we identify novel protein markers associated with FEV1 decline. Several protein findings are supported by previously reported genetic signals, highlighting the plausibility of certain biologic pathways. These novel proteins might represent markers for risk stratification, as well as novel molecular targets for treatment of COPD.
Collapse
Affiliation(s)
- Debby Ngo
- Cardiovascular Research Institute
- Division of Pulmonary, Critical Care, and Sleep Medicine, and
| | | | - Claudia Flexeder
- Institute of Epidemiology and
- Comprehensive Pneumology Center Munich (CPC-M) as member of the German Center for Lung Research (DZL), Munich, Germany
- Institute and Clinic for Occupational, Social, and Environmental Medicine, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Hans Petersen
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Hong Dang
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yanlin Ma
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | | | - Yan Gao
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; and
- Institute and Clinic for Occupational, Social, and Environmental Medicine, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Claire Emson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Christian Gieger
- Institute of Epidemiology and
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Doha, Qatar
| | | | - Deepti Jain
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Matthew P. Conomos
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA (University of California, Los Angeles) Medical Center, Torrance, California
| | - Yongmei Liu
- Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Elaine Cornell
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA (University of California, Los Angeles) Medical Center, Torrance, California
| | - George J. Papanicolaou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA (University of California, Los Angeles) Medical Center, Torrance, California
| | - Steven I. Rennard
- Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | - Prescott G. Woodruff
- Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | | | | | - Martin G. Larson
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts
| | - Ramachandran S. Vasan
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts
- Division of Preventive Medicine and
- Division of Cardiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Thomas J. Wang
- Department of Medicine, UT (University of Texas) Southwestern Medical Center, Dallas, Texas
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, and
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, and
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - James G. Wilson
- Cardiovascular Research Institute
- Jackson Heart Study, Department of Medicine, and
| | - Robert E. Gerszten
- Cardiovascular Research Institute
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - George T. O’Connor
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts
- Pulmonary Center, Department of Medicine, Boston University, Boston, Massachusetts
| | - R. Graham Barr
- Department of Medicine and
- Department of Epidemiology, Columbia University, New York, New York
| | - David Couper
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Wanda K. O’Neal
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yohannes Tesfaigzi
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Holger Schulz
- Institute of Epidemiology and
- Comprehensive Pneumology Center Munich (CPC-M) as member of the German Center for Lung Research (DZL), Munich, Germany
| | | |
Collapse
|
36
|
Álvez MB, Edfors F, von Feilitzen K, Zwahlen M, Mardinoglu A, Edqvist PH, Sjöblom T, Lundin E, Rameika N, Enblad G, Lindman H, Höglund M, Hesselager G, Stålberg K, Enblad M, Simonson OE, Häggman M, Axelsson T, Åberg M, Nordlund J, Zhong W, Karlsson M, Gyllensten U, Ponten F, Fagerberg L, Uhlén M. Next generation pan-cancer blood proteome profiling using proximity extension assay. Nat Commun 2023; 14:4308. [PMID: 37463882 DOI: 10.1038/s41467-023-39765-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
A comprehensive characterization of blood proteome profiles in cancer patients can contribute to a better understanding of the disease etiology, resulting in earlier diagnosis, risk stratification and better monitoring of the different cancer subtypes. Here, we describe the use of next generation protein profiling to explore the proteome signature in blood across patients representing many of the major cancer types. Plasma profiles of 1463 proteins from more than 1400 cancer patients are measured in minute amounts of blood collected at the time of diagnosis and before treatment. An open access Disease Blood Atlas resource allows the exploration of the individual protein profiles in blood collected from the individual cancer patients. We also present studies in which classification models based on machine learning have been used for the identification of a set of proteins associated with each of the analyzed cancers. The implication for cancer precision medicine of next generation plasma profiling is discussed.
Collapse
Affiliation(s)
- María Bueno Álvez
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Kalle von Feilitzen
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Martin Zwahlen
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Per-Henrik Edqvist
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Tobias Sjöblom
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Emma Lundin
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Natallia Rameika
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Henrik Lindman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Martin Höglund
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Göran Hesselager
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Karin Stålberg
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Malin Enblad
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Oscar E Simonson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Michael Häggman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Tomas Axelsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and SciLifeLab Affinity Proteomics, Uppsala University, Uppsala, Sweden
| | - Jessica Nordlund
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Wen Zhong
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Max Karlsson
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fredrik Ponten
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden.
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
37
|
Kim H, Lichtenstein AH, Ganz P, Miller ER, Coresh J, Appel LJ, Rebholz CM. Associations of circulating proteins with lipoprotein profiles: proteomic analyses from the OmniHeart randomized trial and the Atherosclerosis Risk in Communities (ARIC) Study. Clin Proteomics 2023; 20:27. [PMID: 37400771 PMCID: PMC10316599 DOI: 10.1186/s12014-023-09416-x] [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: 11/05/2022] [Accepted: 06/19/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Within healthy dietary patterns, manipulation of the proportion of macronutrient can reduce CVD risk. However, the biological pathways underlying healthy diet-disease associations are poorly understood. Using an untargeted, large-scale proteomic profiling, we aimed to (1) identify proteins mediating the association between healthy dietary patterns varying in the proportion of macronutrient and lipoproteins, and (2) validate the associations between diet-related proteins and lipoproteins in the Atherosclerosis Risk in Communities (ARIC) Study. METHODS In 140 adults from the OmniHeart trial, a randomized, cross-over, controlled feeding study with 3 intervention periods (carbohydrate-rich; protein-rich; unsaturated fat-rich dietary patterns), 4,958 proteins were quantified at the end of each diet intervention period using an aptamer assay (SomaLogic). We assessed differences in log2-transformed proteins in 3 between-diet comparisons using paired t-tests, examined the associations between diet-related proteins and lipoproteins using linear regression, and identified proteins mediating these associations using a causal mediation analysis. Levels of diet-related proteins and lipoprotein associations were validated in the ARIC study (n = 11,201) using multivariable linear regression models, adjusting for important confounders. RESULTS Three between-diet comparisons identified 497 significantly different proteins (protein-rich vs. carbohydrate-rich = 18; unsaturated fat-rich vs. carbohydrate-rich = 335; protein-rich vs. unsaturated fat-rich dietary patterns = 398). Of these, 9 proteins [apolipoprotein M, afamin, collagen alpha-3(VI) chain, chitinase-3-like protein 1, inhibin beta A chain, palmitoleoyl-protein carboxylesterase NOTUM, cathelicidin antimicrobial peptide, guanylate-binding protein 2, COP9 signalosome complex subunit 7b] were positively associated with lipoproteins [high-density lipoprotein (HDL)-cholesterol (C) = 2; triglyceride = 5; non-HDL-C = 3; total cholesterol to HDL-C ratio = 1]. Another protein, sodium-coupled monocarboxylate transporter 1, was inversely associated with HDL-C and positively associated with total cholesterol to HDL-C ratio. The proportion of the association between diet and lipoproteins mediated by these 10 proteins ranged from 21 to 98%. All of the associations between diet-related proteins and lipoproteins were significant in the ARIC study, except for afamin. CONCLUSIONS We identified proteins that mediate the association between healthy dietary patterns varying in macronutrients and lipoproteins in a randomized feeding study and an observational study. TRIAL REGISTRATION NCT00051350 at clinicaltrials.gov.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 2024 East Monument Street, Suite 2-500, Baltimore, MD 21287 USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD USA
| | - Alice H. Lichtenstein
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA USA
| | - Peter Ganz
- Department of Medicine, University of California San Francisco, San Francisco, CA USA
| | - Edgar R. Miller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 2024 East Monument Street, Suite 2-500, Baltimore, MD 21287 USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 2024 East Monument Street, Suite 2-500, Baltimore, MD 21287 USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Lawrence J. Appel
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 2024 East Monument Street, Suite 2-500, Baltimore, MD 21287 USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 2024 East Monument Street, Suite 2-500, Baltimore, MD 21287 USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| |
Collapse
|
38
|
Dagher M, Ongo G, Robichaud N, Kong J, Rho W, Teahulos I, Tavakoli A, Bovaird S, Merjaneh S, Tan A, Edwardson K, Scheepers C, Ng A, Hajjar A, Sow B, Vrouvides M, Lee A, DeCorwin-Martin P, Rasool S, Huang J, Han Y, Erps T, Coffin S, Chandrasekaran SN, Miller L, Kost-Alimova M, Skepner A, Singh S, Carpenter AE, Munzar J, Juncker D. nELISA: A high-throughput, high-plex platform enables quantitative profiling of the secretome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.535914. [PMID: 37131604 PMCID: PMC10153206 DOI: 10.1101/2023.04.17.535914] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
We present the nELISA, a high-throughput, high-fidelity, and high-plex protein profiling platform. DNA oligonucleotides are used to pre-assemble antibody pairs on spectrally encoded microparticles and perform displacement-mediated detection. Spatial separation between non-cognate antibodies prevents the rise of reagent-driven cross-reactivity, while read-out is performed cost-efficiently and at high-throughput using flow cytometry. We assembled an inflammatory panel of 191 targets that were multiplexed without cross-reactivity or impact on performance vs 1-plex signals, with sensitivities as low as 0.1pg/mL and measurements spanning 7 orders of magnitude. We then performed a large-scale secretome perturbation screen of peripheral blood mononuclear cells (PBMCs), with cytokines as both perturbagens and read-outs, measuring 7,392 samples and generating ~1.5M protein datapoints in under a week, a significant advance in throughput compared to other highly multiplexed immunoassays. We uncovered 447 significant cytokine responses, including multiple putatively novel ones, that were conserved across donors and stimulation conditions. We also validated the nELISA's use in phenotypic screening, and propose its application to drug discovery.
Collapse
|
39
|
Galbraith MD, Rachubinski AL, Smith KP, Araya P, Waugh KA, Enriquez-Estrada B, Worek K, Granrath RE, Kinning KT, Paul Eduthan N, Ludwig MP, Hsieh EW, Sullivan KD, Espinosa JM. Multidimensional definition of the interferonopathy of Down syndrome and its response to JAK inhibition. SCIENCE ADVANCES 2023; 9:eadg6218. [PMID: 37379383 PMCID: PMC10306300 DOI: 10.1126/sciadv.adg6218] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023]
Abstract
Individuals with Down syndrome (DS) display chronic hyperactivation of interferon signaling. However, the clinical impacts of interferon hyperactivity in DS are ill-defined. Here, we describe a multiomics investigation of interferon signaling in hundreds of individuals with DS. Using interferon scores derived from the whole blood transcriptome, we defined the proteomic, immune, metabolic, and clinical features associated with interferon hyperactivity in DS. Interferon hyperactivity associates with a distinct proinflammatory phenotype and dysregulation of major growth signaling and morphogenic pathways. Individuals with the highest interferon activity display the strongest remodeling of the peripheral immune system, including increased cytotoxic T cells, B cell depletion, and monocyte activation. Interferon hyperactivity accompanies key metabolic changes, most prominently dysregulated tryptophan catabolism. High interferon signaling stratifies a subpopulation with elevated rates of congenital heart disease and autoimmunity. Last, a longitudinal case study demonstrated that JAK inhibition normalizes interferon signatures with therapeutic benefit in DS. Together, these results justify the testing of immune-modulatory therapies in DS.
Collapse
Affiliation(s)
- Matthew D. Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angela L. Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Keith P. Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine A. Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Belinda Enriquez-Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kayleigh Worek
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross E. Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kohl T. Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael P. Ludwig
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elena W. Y. Hsieh
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Division of Allergy/Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
40
|
Gedik H, Nguyen TH, Peterson RE, Chatzinakos C, Vladimirov VI, Riley BP, Bacanu SA. Identifying potential risk genes and pathways for neuropsychiatric and substance use disorders using intermediate molecular mediator information. Front Genet 2023; 14:1191264. [PMID: 37415601 PMCID: PMC10320396 DOI: 10.3389/fgene.2023.1191264] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/23/2023] [Indexed: 07/08/2023] Open
Abstract
Neuropsychiatric and substance use disorders (NPSUDs) have a complex etiology that includes environmental and polygenic risk factors with significant cross-trait genetic correlations. Genome-wide association studies (GWAS) of NPSUDs yield numerous association signals. However, for most of these regions, we do not yet have a firm understanding of either the specific risk variants or the effects of these variants. Post-GWAS methods allow researchers to use GWAS summary statistics and molecular mediators (transcript, protein, and methylation abundances) infer the effect of these mediators on risk for disorders. One group of post-GWAS approaches is commonly referred to as transcriptome/proteome/methylome-wide association studies, which are abbreviated as T/P/MWAS (or collectively as XWAS). Since these approaches use biological mediators, the multiple testing burden is reduced to the number of genes (∼20,000) instead of millions of GWAS SNPs, which leads to increased signal detection. In this work, our aim is to uncover likely risk genes for NPSUDs by performing XWAS analyses in two tissues-blood and brain. First, to identify putative causal risk genes, we performed an XWAS using the Summary-data-based Mendelian randomization, which uses GWAS summary statistics, reference xQTL data, and a reference LD panel. Second, given the large comorbidities among NPSUDs and the shared cis-xQTLs between blood and the brain, we improved XWAS signal detection for underpowered analyses by performing joint concordance analyses between XWAS results i) across the two tissues and ii) across NPSUDs. All XWAS signals i) were adjusted for heterogeneity in dependent instruments (HEIDI) (non-causality) p-values and ii) used to test for pathway enrichment. The results suggest that there were widely shared gene/protein signals within the major histocompatibility complex region on chromosome 6 (BTN3A2 and C4A) and elsewhere in the genome (FURIN, NEK4, RERE, and ZDHHC5). The identification of putative molecular genes and pathways underlying risk may offer new targets for therapeutic development. Our study revealed an enrichment of XWAS signals in vitamin D and omega-3 gene sets. So, including vitamin D and omega-3 in treatment plans may have a modest but beneficial effect on patients with bipolar disorder.
Collapse
Affiliation(s)
- Huseyin Gedik
- Integrative Life Sciences, Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Tan Hoang Nguyen
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Roseann E. Peterson
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
| | - Christos Chatzinakos
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Department of Psychiatry, McLean Hospital and Harvard Medical School, Belmont, MA, United States
| | - Vladimir I. Vladimirov
- Department of Psychiatry, College of Medicine, University of Arizona Phoenix, Phoenix, AZ, United States
| | - Brien P. Riley
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Silviu-Alin Bacanu
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
41
|
Paranjpe I, Jayaraman P, Su CY, Zhou S, Chen S, Thompson R, Del Valle DM, Kenigsberg E, Zhao S, Jaladanki S, Chaudhary K, Ascolillo S, Vaid A, Gonzalez-Kozlova E, Kauffman J, Kumar A, Paranjpe M, Hagan RO, Kamat S, Gulamali FF, Xie H, Harris J, Patel M, Argueta K, Batchelor C, Nie K, Dellepiane S, Scott L, Levin MA, He JC, Suarez-Farinas M, Coca SG, Chan L, Azeloglu EU, Schadt E, Beckmann N, Gnjatic S, Merad M, Kim-Schulze S, Richards B, Glicksberg BS, Charney AW, Nadkarni GN. Proteomic characterization of acute kidney injury in patients hospitalized with SARS-CoV2 infection. COMMUNICATIONS MEDICINE 2023; 3:81. [PMID: 37308534 PMCID: PMC10258469 DOI: 10.1038/s43856-023-00307-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/18/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a known complication of COVID-19 and is associated with an increased risk of in-hospital mortality. Unbiased proteomics using biological specimens can lead to improved risk stratification and discover pathophysiological mechanisms. METHODS Using measurements of ~4000 plasma proteins in two cohorts of patients hospitalized with COVID-19, we discovered and validated markers of COVID-associated AKI (stage 2 or 3) and long-term kidney dysfunction. In the discovery cohort (N = 437), we identified 413 higher plasma abundances of protein targets and 30 lower plasma abundances of protein targets associated with COVID-AKI (adjusted p < 0.05). Of these, 62 proteins were validated in an external cohort (p < 0.05, N = 261). RESULTS We demonstrate that COVID-AKI is associated with increased markers of tubular injury (NGAL) and myocardial injury. Using estimated glomerular filtration (eGFR) measurements taken after discharge, we also find that 25 of the 62 AKI-associated proteins are significantly associated with decreased post-discharge eGFR (adjusted p < 0.05). Proteins most strongly associated with decreased post-discharge eGFR included desmocollin-2, trefoil factor 3, transmembrane emp24 domain-containing protein 10, and cystatin-C indicating tubular dysfunction and injury. CONCLUSIONS Using clinical and proteomic data, our results suggest that while both acute and long-term COVID-associated kidney dysfunction are associated with markers of tubular dysfunction, AKI is driven by a largely multifactorial process involving hemodynamic instability and myocardial damage.
Collapse
Affiliation(s)
- Ishan Paranjpe
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Pushkala Jayaraman
- The Charles Bronfman Institute for Personalized Medicine (CBIPM), Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chen-Yang Su
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Computer Science, Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Sirui Zhou
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Steven Chen
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ryan Thompson
- The Charles Bronfman Institute for Personalized Medicine (CBIPM), Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Marie Del Valle
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ephraim Kenigsberg
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shan Zhao
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Suraj Jaladanki
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kumardeep Chaudhary
- Clinical Informatics, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Steven Ascolillo
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Akhil Vaid
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edgar Gonzalez-Kozlova
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Justin Kauffman
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arvind Kumar
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manish Paranjpe
- Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Ross O Hagan
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Kamat
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Faris F Gulamali
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hui Xie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joceyln Harris
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manishkumar Patel
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kimberly Argueta
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Craig Batchelor
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergio Dellepiane
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leisha Scott
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew A Levin
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Cijiang He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mayte Suarez-Farinas
- Department of Biostatistics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven G Coca
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lili Chan
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evren U Azeloglu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noam Beckmann
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miram Merad
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brent Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Computer Science, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Department of Twin Research, King's College London, London, GB, UK
| | | | - Alexander W Charney
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine (CBIPM), Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada.
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
42
|
Wang S, Onyeaghala GC, Pankratz N, Nelson HH, Thyagarajan B, Tang W, Norby FL, Ugoji C, Joshu CE, Gomez CR, Couper DJ, Coresh J, Platz EA, Prizment AE. Associations between MICA and MICB Genetic Variants, Protein Levels, and Colorectal Cancer: Atherosclerosis Risk in Communities (ARIC). Cancer Epidemiol Biomarkers Prev 2023; 32:784-794. [PMID: 36958849 PMCID: PMC10239349 DOI: 10.1158/1055-9965.epi-22-1113] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/24/2023] [Accepted: 03/20/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND The MHC class I chain-related protein A (MICA) and protein B (MICB) participate in tumor immunosurveillance and may be important in colorectal cancer, but have not been examined in colorectal cancer development. METHODS sMICA and sMICB blood levels were measured by SomaScan in Visit 2 (1990-92, baseline) and Visit 3 (1993-95) samples in cancer-free participants in the Atherosclerosis Risk in Communities Study. We selected rs1051792, rs1063635, rs2516448, rs3763288, rs1131896, rs2596542, and rs2395029 that were located in or in the vicinity of MICA or MICB and were associated with cancer or autoimmune diseases in published studies. SNPs were genotyped by the Affymetrix Genome-Wide Human SNP Array. We applied linear and Cox proportional hazards regressions to examine the associations of preselected SNPs with sMICA and sMICB levels and colorectal cancer risk (236 colorectal cancers, 8,609 participants) and of sMICA and sMICB levels with colorectal cancer risk (312 colorectal cancers, 10,834 participants). In genetic analyses, estimates adjusted for ancestry markers were meta-analyzed. RESULTS Rs1051792-A, rs1063635-A, rs2516448-C, rs3763288-A, rs2596542-T, and rs2395029-G were significantly associated with decreased sMICA levels. Rs2395029-G, in the vicinity of MICA and MICB, was also associated with increased sMICB levels. Rs2596542-T was significantly associated with decreased colorectal cancer risk. Lower sMICA levels were associated with lower colorectal cancer risk in males (HR = 0.68; 95% confidence interval, 0.49-0.96) but not in females (Pinteraction = 0.08). CONCLUSIONS Rs2596542-T associated with lower sMICA levels was associated with decreased colorectal cancer risk. Lower sMICA levels were associated with lower colorectal cancer risk in males. IMPACT These findings support an importance of immunosurveillance in colorectal cancer.
Collapse
Affiliation(s)
- Shuo Wang
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, MN
| | - Guillaume C. Onyeaghala
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Heather H Nelson
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Faye L. Norby
- Department of Cardiology, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Chinenye Ugoji
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Christian R. Gomez
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS
| | - David J. Couper
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Anna E. Prizment
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, MN
| |
Collapse
|
43
|
Tasci E, Jagasia S, Zhuge Y, Sproull M, Cooley Zgela T, Mackey M, Camphausen K, Krauze AV. RadWise: A Rank-Based Hybrid Feature Weighting and Selection Method for Proteomic Categorization of Chemoirradiation in Patients with Glioblastoma. Cancers (Basel) 2023; 15:2672. [PMID: 37345009 PMCID: PMC10216128 DOI: 10.3390/cancers15102672] [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: 03/27/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 06/23/2023] Open
Abstract
Glioblastomas (GBM) are rapidly growing, aggressive, nearly uniformly fatal, and the most common primary type of brain cancer. They exhibit significant heterogeneity and resistance to treatment, limiting the ability to analyze dynamic biological behavior that drives response and resistance, which are central to advancing outcomes in glioblastoma. Analysis of the proteome aimed at signal change over time provides a potential opportunity for non-invasive classification and examination of the response to treatment by identifying protein biomarkers associated with interventions. However, data acquired using large proteomic panels must be more intuitively interpretable, requiring computational analysis to identify trends. Machine learning is increasingly employed, however, it requires feature selection which has a critical and considerable effect on machine learning problems when applied to large-scale data to reduce the number of parameters, improve generalization, and find essential predictors. In this study, using 7k proteomic data generated from the analysis of serum obtained from 82 patients with GBM pre- and post-completion of concurrent chemoirradiation (CRT), we aimed to select the most discriminative proteomic features that define proteomic alteration that is the result of administering CRT. Thus, we present a novel rank-based feature weighting method (RadWise) to identify relevant proteomic parameters using two popular feature selection methods, least absolute shrinkage and selection operator (LASSO) and the minimum redundancy maximum relevance (mRMR). The computational results show that the proposed method yields outstanding results with very few selected proteomic features, with higher accuracy rate performance than methods that do not employ a feature selection process. While the computational method identified several proteomic signals identical to the clinical intuitive (heuristic approach), several heuristically identified proteomic signals were not selected while other novel proteomic biomarkers not selected with the heuristic approach that carry biological prognostic relevance in GBM only emerged with the novel method. The computational results show that the proposed method yields promising results, reducing 7k proteomic data to 8 selected proteomic features with a performance value of 96.364%, comparing favorably with techniques that do not employ feature selection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Andra Valentina Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (E.T.); (S.J.); (Y.Z.); (M.S.); (T.C.Z.); (M.M.); (K.C.)
| |
Collapse
|
44
|
Shalon D, Culver RN, Grembi JA, Folz J, Treit PV, Shi H, Rosenberger FA, Dethlefsen L, Meng X, Yaffe E, Aranda-Díaz A, Geyer PE, Mueller-Reif JB, Spencer S, Patterson AD, Triadafilopoulos G, Holmes SP, Mann M, Fiehn O, Relman DA, Huang KC. Profiling the human intestinal environment under physiological conditions. Nature 2023; 617:581-591. [PMID: 37165188 PMCID: PMC10191855 DOI: 10.1038/s41586-023-05989-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/21/2023] [Indexed: 05/12/2023]
Abstract
The spatiotemporal structure of the human microbiome1,2, proteome3 and metabolome4,5 reflects and determines regional intestinal physiology and may have implications for disease6. Yet, little is known about the distribution of microorganisms, their environment and their biochemical activity in the gut because of reliance on stool samples and limited access to only some regions of the gut using endoscopy in fasting or sedated individuals7. To address these deficiencies, we developed an ingestible device that collects samples from multiple regions of the human intestinal tract during normal digestion. Collection of 240 intestinal samples from 15 healthy individuals using the device and subsequent multi-omics analyses identified significant differences between bacteria, phages, host proteins and metabolites in the intestines versus stool. Certain microbial taxa were differentially enriched and prophage induction was more prevalent in the intestines than in stool. The host proteome and bile acid profiles varied along the intestines and were highly distinct from those of stool. Correlations between gradients in bile acid concentrations and microbial abundance predicted species that altered the bile acid pool through deconjugation. Furthermore, microbially conjugated bile acid concentrations exhibited amino acid-dependent trends that were not apparent in stool. Overall, non-invasive, longitudinal profiling of microorganisms, proteins and bile acids along the intestinal tract under physiological conditions can help elucidate the roles of the gut microbiome and metabolome in human physiology and disease.
Collapse
Affiliation(s)
| | - Rebecca Neal Culver
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Jessica A Grembi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacob Folz
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, USA
| | - Peter V Treit
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Handuo Shi
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Florian A Rosenberger
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Les Dethlefsen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Eitan Yaffe
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Philipp E Geyer
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Johannes B Mueller-Reif
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Sean Spencer
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - George Triadafilopoulos
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
- Silicon Valley Neurogastroenterology and Motility Center, Mountain View, CA, USA
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, USA.
- Department of Food Science and Technology, University of California, Davis, Davis, CA, USA.
| | - David A Relman
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Kerwyn Casey Huang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
45
|
Blatt S, Kämmerer PW, Krüger M, Surabattula R, Thiem DGE, Dillon ST, Al-Nawas B, Libermann TA, Schuppan D. High-Multiplex Aptamer-Based Serum Proteomics to Identify Candidate Serum Biomarkers of Oral Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15072071. [PMID: 37046731 PMCID: PMC10093013 DOI: 10.3390/cancers15072071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Improved serological biomarkers are needed for the early detection, risk stratification and treatment surveillance of patients with oral squamous cell carcinoma (OSCC). We performed an exploratory study using advanced, highly specific, DNA-aptamer-based serum proteomics (SOMAscan, 1305-plex) to identify distinct proteomic changes in patients with OSCC pre- vs. post-resection and compared to healthy controls. A total of 63 significantly differentially expressed serum proteins (each p < 0.05) were found that could discriminate between OSCC and healthy controls with 100% accuracy. Furthermore, 121 proteins were detected that were significantly altered between pre- and post-resection sera, and 12 OSCC-associated proteins reversed to levels equivalent to healthy controls after resection. Of these, 6 were increased and 6 were decreased relative to healthy controls, highlighting the potential relevance of these proteins as OSCC tumor markers. Pathway analyses revealed potential pathophysiological mechanisms associated with OSCC. Hence, quantitative proteome analysis using SOMAscan technology is promising and may aid in the development of defined serum marker assays to predict tumor occurrence, progression and recurrence in OSCC, and to guide personalized therapies.
Collapse
|
46
|
Steffen BT, Pankow JS, Norby FL, Lutsey PL, Demmer RT, Guan W, Pankratz N, Li A, Liu G, Matsushita K, Tin A, Tang W. Proteomics Analysis of Genetic Liability of Abdominal Aortic Aneurysm Identifies Plasma Neogenin and Kit Ligand: The ARIC Study. Arterioscler Thromb Vasc Biol 2023; 43:367-378. [PMID: 36579647 PMCID: PMC9995137 DOI: 10.1161/atvbaha.122.317984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Genome-wide association studies have reported 23 gene loci related to abdominal aortic aneurysm (AAA)-a potentially lethal condition characterized by a weakened dilated vessel wall. This study aimed to identify proteomic signatures and pathways related to these risk loci to better characterize AAA genetic susceptibility. METHODS Plasma concentrations of 4870 proteins were determined using a DNA aptamer-based array. Linear regression analysis estimated the associations between the 23 risk alleles and plasma protein levels with adjustments for potential confounders in a race-stratified analysis of 1671 Black and 7241 White participants. Significant proteins were then evaluated for their prediction of clinical AAA (454 AAA events in 11 064 individuals), and those significantly associated with AAA were further interrogated using Mendelian randomization analysis. RESULTS Risk variants proximal to PSRC1-CELSR2-SORT1, PCIF1-ZNF335-MMP9, RP11-136O12.2/TRIB1, ZNF259/APOA5, IL6R, PCSK9, LPA, and APOE were associated with 118 plasma proteins in Whites and 59 were replicated in Black participants. Novel associations with clinical AAA incidence were observed for kit ligand (HR, 0.59 [95% CI, 0.42-0.82] for top versus first quintiles) and neogenin (HR, 0.64 [95% CI, 0.46-0.88]) over a median 21.2-year follow-up; neogenin was also associated with ultrasound-detected asymptomatic AAA (N=4295; 57 asymptomatic AAA cases). Mendelian randomization inverse variance weighted estimates suggested that AAA risk is promoted by lower levels of kit ligand (OR per SD=0.67; P=1.4×10-5) and neogenin (OR per SD=0.50; P=0.03). CONCLUSIONS Low levels of neogenin and kit ligand may be novel risk factors for AAA development in potentially causal pathways. These findings provide insights and potential targets to reduce AAA susceptibility.
Collapse
Affiliation(s)
- Brian T. Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
- Division of Health Data Science, Department of Surgery, University of Minnesota, Minneapolis, MN 55455
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Faye L. Norby
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, CA 90048
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Ryan T. Demmer
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, 55455
| | - Nathan Pankratz
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Aixin Li
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Guning Liu
- Division of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center, School of Public Health, Houston, TX 77030
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD 21205
| | - Adrienne Tin
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| |
Collapse
|
47
|
Steffen BT, Tang W, Lutsey PL, Demmer RT, Selvin E, Matsushita K, Morrison AC, Guan W, Rooney MR, Norby FL, Pankratz N, Couper D, Pankow JS. Proteomic analysis of diabetes genetic risk scores identifies complement C2 and neuropilin-2 as predictors of type 2 diabetes: the Atherosclerosis Risk in Communities (ARIC) Study. Diabetologia 2023; 66:105-115. [PMID: 36194249 PMCID: PMC9742300 DOI: 10.1007/s00125-022-05801-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Genetic predisposition to type 2 diabetes is well-established, and genetic risk scores (GRS) have been developed that capture heritable liabilities for type 2 diabetes phenotypes. However, the proteins through which these genetic variants influence risk have not been thoroughly investigated. This study aimed to identify proteins and pathways through which type 2 diabetes risk variants may influence pathophysiology. METHODS Using a proteomics data-driven approach in a discovery sample of 7241 White participants in the Atherosclerosis Risk in Communities Study (ARIC) cohort and a replication sample of 1674 Black ARIC participants, we interrogated plasma levels of 4870 proteins and four GRS of specific type 2 diabetes phenotypes related to beta cell function, insulin resistance, lipodystrophy, BMI/blood lipid abnormalities and a composite score of all variants combined. RESULTS Twenty-two plasma proteins were identified in White participants after Bonferroni correction. Of the 22 protein-GRS associations that were statistically significant, 10 were replicated in Black participants and all but one were directionally consistent. In a secondary analysis, 18 of the 22 proteins were found to be associated with prevalent type 2 diabetes and ten proteins were associated with incident type 2 diabetes. Two-sample Mendelian randomisation indicated that complement C2 may be causally related to greater type 2 diabetes risk (inverse variance weighted estimate: OR 1.65 per SD; p=7.0 × 10-3), while neuropilin-2 was inversely associated (OR 0.44 per SD; p=8.0 × 10-3). CONCLUSIONS/INTERPRETATION Identified proteins may represent viable intervention or pharmacological targets to prevent, reverse or slow type 2 diabetes progression, and further research is needed to pursue these targets.
Collapse
Affiliation(s)
- Brian T Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Pamela L Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Mary R Rooney
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Faye L Norby
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, CA, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | - David Couper
- University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
48
|
Hshieh TT, Schmitt EM, Fong TG, Arnold S, Cavallari M, Dickerson BC, Dillon ST, Jones RN, Libermann TA, Marcantonio ER, Pascual-Leone A, Shafi MM, Touroutoglou A, Travison TG, Gou RY, Tommet D, Abdeen A, Earp B, Kunze L, Lange J, Vlassakov K, Inouye SK. Successful aging after elective surgery II: Study design and methods. J Am Geriatr Soc 2023; 71:46-61. [PMID: 36214228 PMCID: PMC9870853 DOI: 10.1111/jgs.18065] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/30/2022] [Accepted: 09/04/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND The Successful Aging after Elective Surgery (SAGES) II study was designed to increase knowledge of the pathophysiology and linkages between delirium and dementia. We examine novel biomarkers potentially associated with delirium, including inflammation, Alzheimer's disease (AD) pathology and neurodegeneration, neuroimaging markers, and neurophysiologic markers. The goal of this paper is to describe the study design and methods for the SAGES II study. METHODS The SAGES II study is a 5-year prospective observational study of 400-420 community dwelling persons, aged 65 years and older, assessed prior to scheduled surgery and followed daily throughout hospitalization to observe for development of delirium and other clinical outcomes. Delirium is measured with the Confusion Assessment Method (CAM), long form, after cognitive testing. Cognitive function is measured with a detailed neuropsychologic test battery, summarized as a weighted composite, the General Cognitive Performance (GCP) score. Other key measures include magnetic resonance imaging (MRI), transcranial magnetic stimulation (TMS)/electroencephalography (EEG), and Amyloid positron emission tomography (PET) imaging. We describe the eligibility criteria, enrollment flow, timing of assessments, and variables collected at baseline and during repeated assessments at 1, 2, 6, 12, and 18 months. RESULTS This study describes the hospital and surgery-related variables, delirium, long-term cognitive decline, clinical outcomes, and novel biomarkers. In inter-rater reliability assessments, the CAM ratings (weighted kappa = 0.91, 95% confidence interval, CI = 0.74-1.0) in 50 paired assessments and GCP ratings (weighted kappa = 0.99, 95% CI 0.94-1.0) in 25 paired assessments. We describe procedures for data quality assurance and Covid-19 adaptations. CONCLUSIONS This complex study presents an innovative effort to advance our understanding of the inter-relationship between delirium and dementia via novel biomarkers, collected in the context of major surgery in older adults. Strengths include the integration of MRI, TMS/EEG, PET modalities, and high-quality longitudinal data.
Collapse
Affiliation(s)
- Tammy T. Hshieh
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Eva M. Schmitt
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | - Tamara G. Fong
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Steve Arnold
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michele Cavallari
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
- Department of Radiology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | - Simon T. Dillon
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Richard N. Jones
- Department of Psychiatry and Human Behavior, Department of Neurology, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Towia A. Libermann
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Edward R. Marcantonio
- Division of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Alvaro Pascual-Leone
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
- Deanna and Sidney Wolk Center for Memory Health, HebrewSeniorLife, Boston, Massachusetts, USA
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Mouhsin M. Shafi
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Thomas G. Travison
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | - Ray Yun Gou
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | - Douglas Tommet
- Department of Psychiatry and Human Behavior, Department of Neurology, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Ayesha Abdeen
- Department of Orthopedic Surgery, Boston Medical Center, Boston, Massachusetts, USA
| | - Brandon Earp
- Department of Orthopedic Surgery, Brigham and Women’s Faulkner Hospital, Boston, Massachusetts, USA
| | - Lisa Kunze
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jeffrey Lange
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Kamen Vlassakov
- Department of Anesthesia, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Sharon K. Inouye
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | | |
Collapse
|
49
|
Chen J, Xiang Y, Wang P, Liu J, Lai W, Xiao M, Pei H, Fan C, Li L. Ensemble Modified Aptamer Based Pattern Recognition for Adaptive Target Identification. NANO LETTERS 2022; 22:10057-10065. [PMID: 36524831 DOI: 10.1021/acs.nanolett.2c03808] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The difficulty of the molecular design and chemical synthesis of artificial sensing receptors restricts their diagnostic and proteomic applications. Herein, we report a concept of "ensemble modified aptamers" (EMAmers) that exploits the collective recognition abilities of a small set of protein-like side-chain-modified nucleic acid ligands for discriminative identification of molecular or cellular targets. Different types and numbers of hydrophobic functional groups were incorporated at designated positions on nucleic acid scaffolds to mimic amino acid side chains. We successfully assayed 18 EMAmer probes with differential binding affinities to seven proteins. We constructed an EMAmer-based chemical nose sensor and demonstrated its application in blinded unknown protein identification, giving a 92.9% accuracy. Additionally, the sensor is generalizable to the detection of blinded unknown bacterial and cellular samples, which enabled identification accuracies of 96.3% and 94.8%, respectively. This sensing platform offers a discriminative means for adaptive target identification and holds great potential for diverse applications.
Collapse
Affiliation(s)
- Jing Chen
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Ying Xiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Peipei Wang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Jingjing Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Wei Lai
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Hao Pei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201240, People's Republic of China
| | - Li Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| |
Collapse
|
50
|
Kosa P, Barbour C, Varosanec M, Wichman A, Sandford M, Greenwood M, Bielekova B. Molecular models of multiple sclerosis severity identify heterogeneity of pathogenic mechanisms. Nat Commun 2022; 13:7670. [PMID: 36509784 PMCID: PMC9744737 DOI: 10.1038/s41467-022-35357-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
While autopsy studies identify many abnormalities in the central nervous system (CNS) of subjects dying with neurological diseases, without their quantification in living subjects across the lifespan, pathogenic processes cannot be differentiated from epiphenomena. Using machine learning (ML), we searched for likely pathogenic mechanisms of multiple sclerosis (MS). We aggregated cerebrospinal fluid (CSF) biomarkers from 1305 proteins, measured blindly in the training dataset of untreated MS patients (N = 129), into models that predict past and future speed of disability accumulation across all MS phenotypes. Healthy volunteers (N = 24) data differentiated natural aging and sex effects from MS-related mechanisms. Resulting models, validated (Rho 0.40-0.51, p < 0.0001) in an independent longitudinal cohort (N = 98), uncovered intra-individual molecular heterogeneity. While candidate pathogenic processes must be validated in successful clinical trials, measuring them in living people will enable screening drugs for desired pharmacodynamic effects. This will facilitate drug development making, it hopefully more efficient and successful.
Collapse
Affiliation(s)
- Peter Kosa
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Christopher Barbour
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Mihael Varosanec
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Alison Wichman
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Mary Sandford
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Mark Greenwood
- grid.41891.350000 0001 2156 6108Department of Mathematical Sciences, Montana State University, Bozeman, MT USA
| | - Bibiana Bielekova
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|