1
|
Seyedmohammad S, Rivas A, Zhgamadze M, Haghani A, Kreimer S, Bharadwaj A, Sundararaman N, Vasantgadkar S, Pal K, Daviso E, Stotland A, Murray C, Raedschelders K, Savant S, Van Eyk JE. High-Throughput Workflow for Detergent-free Cell-Based Proteomic Characterization. J Proteome Res 2025. [PMID: 40255039 DOI: 10.1021/acs.jproteome.4c00892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
We have developed an automated cell-based workflow for the quantification of proteins by liquid chromatography-mass spectrometry (LC-MS) that facilitates large-scale perturbation studies carried out in a 96-well plate format and enables the preparation of one full plate in approximately 4 h, showcasing a high-throughput (HTP) concept. Cells were grown in a 96-well plate and lysed via ultrasonication. Proteins were subsequently solubilized, extracted, and processed into tryptic peptides for 2 h before being acquired by data-independent acquisition mass spectrometry (DIA-MS). This workflow leverages adaptive focused acoustics (AFA) technology for ultrasonication to aid cell lysis and protein solubilization on an automated liquid handling platform. As proof of principle, AC16 human cardiomyocyte-like cells were cultured in a 96-well plate under optimized conditions that were compatible with the downstream HTP pipeline. Over 30,000 peptides were identified, corresponding to the detection of 5100 unique proteins. 50% of measured proteins had an average coefficient of variation (CV) under 25% from approximately 30,000 cells. Our optimized detergent-free buffer consisting of ammonium bicarbonate yielded comparable findings. For the same number of cells, 5000 proteins were identified from 29,000 peptides, 40% of which demonstrated a CV under 25%.
Collapse
Affiliation(s)
- Saeed Seyedmohammad
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Alejandro Rivas
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Maxim Zhgamadze
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Ali Haghani
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Simion Kreimer
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Ajay Bharadwaj
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Niveda Sundararaman
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Sameer Vasantgadkar
- Covaris LLC., 14 Gill St Unit H, Woburn 01801-1721, Massachusetts, United States
| | - Kasturi Pal
- Covaris LLC., 14 Gill St Unit H, Woburn 01801-1721, Massachusetts, United States
| | - Eugenio Daviso
- Covaris LLC., 14 Gill St Unit H, Woburn 01801-1721, Massachusetts, United States
| | - Aleksandr Stotland
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Chris Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| | - Sudha Savant
- Beckman Coulter Life Sciences, 5350 Lakeview Parkway Drive South, Suite A, Indianapolis 46268, Indiana, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical 7 Center, 8700 Beverly Blvd, Los Angeles 90048, California, United States
| |
Collapse
|
2
|
Kulig P, Brazauskas P, Suffiotti M, Raoult E, Babilonski U, Renault B, Grieder U, Vezzali E, Blattmann P, Martinic MM, Murphy MJ. Efficacy of IDOR-1117-2520, a novel, orally available CCR6 antagonist in preclinical models of skin dermatitis. Br J Pharmacol 2025. [PMID: 40156059 DOI: 10.1111/bph.70025] [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: 10/18/2024] [Revised: 01/22/2025] [Accepted: 02/11/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND AND PURPOSE The chemokine receptor CCR6 guides pathogenic T17 cells, implicated in autoimmune diseases including psoriasis, to sites of inflammation via the chemokine CCL20. Therefor, pharmacological inhibition of CCR6+ immune cell migration provides a novel therapeutic approach. Translatability of such an intervention has not yet been assessed in detail. We evaluated the translatability of the Aldara® mouse model induced skin inflammation to psoriasis, with particular focus on immune cell trafficking and assessed the efficacy of IDOR-1117-2520, a highly selective, potent and orally available CCR6 small inhibitor. EXPERIMENTAL APPROACH Effects of IDOR-1117-2520 were investigated in the Aldara® and IL23 mouse models of skin inflammation using flow cytometry, RNA sequencing and transcriptome-based cell type deconvolution approaches to characterise immune cell migration patterns. These results were compared to human psoriasis transcriptomics data. KEY RESULTS IDOR-1117-2520 dose dependently reduced infiltration of CCR6+ immune cells into inflamed skin, and was equally efficacious as IL-17 and IL-23 inhibition in models of skin inflammation. Pathway analysis showed molecular similarities in the immune response between human psoriasis and the Aldara® mouse model. IL-17/IL-23 pathway genes were expressed in both human psoriasis and the mouse model. CCR6 inhibition modulated multiple pathways associated with inflammation beyond the proximal IL-17/IL-23 pathway. A chemokine-chemokine receptor interaction map implicated CCL20-CCR6 as the dominant axis in recruiting pathogenic T17 cells in both the model and in human psoriasis. CONCLUSION AND IMPLICATIONS IDOR-1117-2520 could provide a promising novel targeted approach to treating psoriasis and, potentially, other autoimmune diseases involving the CCR6/CCL20 axis and the IL-17/IL-23 pathway. IDOR-1117-2520 is currently being evaluated in a clinical phase 1 trial (ISRCTN28892128).
Collapse
Affiliation(s)
- Paulina Kulig
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Pijus Brazauskas
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Madeleine Suffiotti
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Emilie Raoult
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Ulrike Babilonski
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Bérengère Renault
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Ursula Grieder
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Enrico Vezzali
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Peter Blattmann
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Marianne M Martinic
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Mark J Murphy
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| |
Collapse
|
3
|
Vegesna M, Sundararaman N, Bharadwaj A, Washington K, Pandey R, Haghani A, Chazarin B, Binek A, Fu Q, Cheng S, Herrington D, Van Eyk JE. Enhancing Proteomics Quality Control: Insights from the Visualization Tool QCeltis. J Proteome Res 2025; 24:1148-1160. [PMID: 39992359 DOI: 10.1021/acs.jproteome.4c00777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Large-scale mass-spectrometry-based proteomics experiments are complex and prone to analytical variability, requiring rigorous quality checks across each step in the workflow: sample preparation, chromatography, mass spectrometry, and the bioinformatics stages. This includes quality control (QC) measures that address biological and technical variation. Most QC approaches involve detecting sample outliers and monitoring parameters related to sample preparation and mass spectrometer performance. Evaluating these parameters regularly is essential for reliable downstream analysis and proteomics research. Here, we introduce "QCeltis", a Python package designed to facilitate automated QC analysis across the proteomics workflow, aiding in the identification of technical biases and consistency verification. QCeltis is a versatile tool for detecting QC issues in large-scale data-independent acquisition proteomics experiments by not only identifying sample preparation and acquisition issues but also aiding in differentiating between QC issues vs batch effects. QCeltis is available for command-line use in Windows and Linux environments. We present three case studies showcasing QCeltis's capabilities across different data sets, including depleted plasma, whole blood vs plasma, and dried blood spot samples, emphasizing its potential impact on large-scale proteomics projects. This package can be used to enhance data reliability and enable nuanced downstream analysis and interpretation for proteomics studies.
Collapse
Affiliation(s)
- Manasa Vegesna
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Niveda Sundararaman
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Ajay Bharadwaj
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Kirstin Washington
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Rakhi Pandey
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Ali Haghani
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Blandine Chazarin
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Aleksandra Binek
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Qin Fu
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - David Herrington
- Department of Cardiovascular Medicine, Wake Forest University, Winston-Salem, North Carolina 27101, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| |
Collapse
|
4
|
Liu Y, Mei L, Liang C, Zhong CQ, Tong M, Yu R. Cross-Run Hybrid Features Improve the Identification of Data-Independent Acquisition Proteomics. ACS OMEGA 2024; 9:46362-46372. [PMID: 39583733 PMCID: PMC11579728 DOI: 10.1021/acsomega.4c07398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 11/26/2024]
Abstract
The analysis of data-independent acquisition (DIA) mass spectrometry data is crucial for comprehensive proteomics studies. However, traditional single-run methods often fall short in terms of identification depth and consistency. We present HFDiscrim, a specialized multirun DIA analysis tool aimed at enhancing the depth and consistency of reliable peptide identifications of DIA analysis tools. HFDiscrim was extensively benchmarked on multiple data sets, including the MCB data set, the ccRCC data set, and a three-species benchmark mixture. Compared to PyProphet, HFDiscrim identified 22.04% more precursors, 19.1% more peptides, and 13.2% more proteins while maintaining a controllable false discovery rate. Furthermore, HFDiscrim demonstrated higher identification rates and improved reproducibility across multiple runs. HFDiscrim is publicly available at https://github.com/yachliu/HFDiscrim.
Collapse
Affiliation(s)
- Yachen Liu
- School
of Informatics, Xiamen University, Xiamen, Fujian 361000, China
- National
Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Longfei Mei
- School
of Informatics, Xiamen University, Xiamen, Fujian 361000, China
| | - Chenyu Liang
- National
Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Chuan-Qi Zhong
- School
of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Mengsha Tong
- National
Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China
- School
of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Rongshan Yu
- School
of Informatics, Xiamen University, Xiamen, Fujian 361000, China
- National
Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China
- Aginome
Scientific, Xiamen, Fujian 361005, China
| |
Collapse
|
5
|
Bundgaard L, Årman F, Åhrman E, Walters M, auf dem Keller U, Malmström J, Jacobsen S. An Equine Protein Atlas Highlights Synovial Fluid Proteome Dynamics during Experimentally LPS-Induced Arthritis. J Proteome Res 2024; 23:4849-4863. [PMID: 39395021 PMCID: PMC11536436 DOI: 10.1021/acs.jproteome.4c00125] [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: 02/21/2024] [Revised: 07/26/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
In human proteomics, substantial efforts are ongoing to leverage large collections of mass spectrometry (MS) fragment ion spectra into extensive spectral libraries (SL) as a resource for data independent acquisition (DIA) analysis. Currently, such initiatives in equine research are still missing. Here we present a large-scale equine SL, comprising 6394 canonical proteins and 89,329 unique peptides, based on data dependent acquisition analysis of 75 tissue and body fluid samples from horses. The SL enabled large-scale DIA-MS based quantification of the same samples to generate a quantitative equine protein distribution atlas to infer dominant proteins in different organs and body fluids. Data mining revealed 163 proteins uniquely identified in a specific type of tissue or body fluid, serving as a starting point to determine tissue-specific or tissue-type-specific proteins. We showcase the SL by highlighting proteome dynamics in equine synovial fluid samples during experimental lipopolysaccharide-induced arthritis. A fuzzy c-means cluster analysis pinpointed SERPINB1, ATRN, NGAL, LTF, MMP1, and LBP as putative biomarkers for joint inflammation. This SL provides an extendable resource for future equine studies employing DIA-MS.
Collapse
Affiliation(s)
- Louise Bundgaard
- Section
of Medicine and Surgery, Department of Veterinary Clinical Sciences, University of Copenhagen, 2630 Taastrup, Denmark
- Department
of Biotechnology and Biomedicine, Technical
University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Filip Årman
- Division
of Infection Medicine Proteomics, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Emma Åhrman
- Division
of Infection Medicine Proteomics, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Marie Walters
- Section
of Medicine and Surgery, Department of Veterinary Clinical Sciences, University of Copenhagen, 2630 Taastrup, Denmark
| | - Ulrich auf dem Keller
- Department
of Biotechnology and Biomedicine, Technical
University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Johan Malmström
- Division
of Infection Medicine Proteomics, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Stine Jacobsen
- Section
of Medicine and Surgery, Department of Veterinary Clinical Sciences, University of Copenhagen, 2630 Taastrup, Denmark
| |
Collapse
|
6
|
Hsiao Y, Zhang H, Li GX, Deng Y, Yu F, Valipour Kahrood H, Steele JR, Schittenhelm RB, Nesvizhskii AI. Analysis and Visualization of Quantitative Proteomics Data Using FragPipe-Analyst. J Proteome Res 2024; 23:4303-4315. [PMID: 39254081 DOI: 10.1021/acs.jproteome.4c00294] [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: 09/11/2024]
Abstract
The FragPipe computational proteomics platform is gaining widespread popularity among the proteomics research community because of its fast processing speed and user-friendly graphical interface. Although FragPipe produces well-formatted output tables that are ready for analysis, there is still a need for an easy-to-use and user-friendly downstream statistical analysis and visualization tool. FragPipe-Analyst addresses this need by providing an R shiny web server to assist FragPipe users in conducting downstream analyses of the resulting quantitative proteomics data. It supports major quantification workflows, including label-free quantification, tandem mass tags, and data-independent acquisition. FragPipe-Analyst offers a range of useful functionalities, such as various missing value imputation options, data quality control, unsupervised clustering, differential expression (DE) analysis using Limma, and gene ontology and pathway enrichment analysis using Enrichr. To support advanced analysis and customized visualizations, we also developed FragPipeAnalystR, an R package encompassing all FragPipe-Analyst functionalities that is extended to support site-specific analysis of post-translational modifications (PTMs). FragPipe-Analyst and FragPipeAnalystR are both open-source and freely available.
Collapse
Affiliation(s)
- Yi Hsiao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Haijian Zhang
- Monash Proteomics & Metabolomics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Ginny Xiaohe Li
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yamei Deng
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hossein Valipour Kahrood
- Monash Proteomics & Metabolomics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Monash Genomics & Bioinformatics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Joel R Steele
- Monash Proteomics & Metabolomics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Alexey I Nesvizhskii
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
7
|
Pelaz SG, Flores-Hernández R, Vujic T, Schvartz D, Álvarez-Vázquez A, Ding Y, García-Vicente L, Belloso A, Talaverón R, Sánchez JC, Tabernero A. A proteomic approach supports the clinical relevance of TAT-Cx43 266-283 in glioblastoma. Transl Res 2024; 272:95-110. [PMID: 38876188 DOI: 10.1016/j.trsl.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/18/2024] [Accepted: 06/01/2024] [Indexed: 06/16/2024]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary brain cancer. The Src inhibitor, TAT-Cx43266-283, exerts antitumor effects in in vitro and in vivo models of GBM. Because addressing the mechanism of action is essential to translate these results to a clinical setting, in this study we carried out an unbiased proteomic approach. Data-independent acquisition mass spectrometry proteomics allowed the identification of 190 proteins whose abundance was modified by TAT-Cx43266-283. Our results were consistent with the inhibition of Src as the mechanism of action of TAT-Cx43266-283 and unveiled antitumor effectors, such as p120 catenin. Changes in the abundance of several proteins suggested that TAT-Cx43266-283 may also impact the brain microenvironment. Importantly, the proteins whose abundance was reduced by TAT-Cx43266-283 correlated with an improved GBM patient survival in clinical datasets and none of the proteins whose abundance was increased by TAT-Cx43266-283 correlated with shorter survival, supporting its use in clinical trials.
Collapse
Affiliation(s)
- Sara G Pelaz
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain.
| | - Raquel Flores-Hernández
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Tatjana Vujic
- Department of Medicine, University of Geneva, 1211, Geneva, Switzerland; University Center of Legal Medicine, Lausanne-Geneva, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne Geneva, Switzerland
| | - Domitille Schvartz
- Department of Medicine, University of Geneva, 1211, Geneva, Switzerland; University of Geneva, Faculty of Medicine, Proteomics Core Facility, Geneva, Switzerland
| | - Andrea Álvarez-Vázquez
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Yuxin Ding
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Laura García-Vicente
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Aitana Belloso
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Rocío Talaverón
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | | | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain.
| |
Collapse
|
8
|
Sevim Bayrak C, Forst CV, Jones DR, Gresham DJ, Pushalkar S, Wu S, Vogel C, Mahal LK, Ghedin E, Ross T, García-Sastre A, Zhang B. Patient subtyping analysis of baseline multi-omic data reveals distinct pre-immune states associated with antibody response to seasonal influenza vaccination. Clin Immunol 2024; 266:110333. [PMID: 39089348 PMCID: PMC11340208 DOI: 10.1016/j.clim.2024.110333] [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: 06/18/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/03/2024]
Abstract
Understanding the molecular mechanisms underpinning diverse vaccination responses is critical for developing efficient vaccines. Molecular subtyping can offer insights into heterogeneous nature of responses and aid in vaccine design. We analyzed multi-omic data from 62 haemagglutinin seasonal influenza vaccine recipients (2019-2020), including transcriptomics, proteomics, glycomics, and metabolomics data collected pre-vaccination. We performed a subtyping analysis on the integrated data revealing five subtypes with distinct molecular signatures. These subtypes differed in the expression of pre-existing adaptive or innate immunity signatures, which were linked to significant variation in baseline immunoglobulin A (IgA) and hemagglutination inhibition (HAI) titer levels. It is worth noting that these differences persisted through day 28 post-vaccination, indicating the effect of initial immune state on vaccination response. These findings highlight the significance of interpersonal variation in baseline immune status as a crucial factor in determining the effectiveness of seasonal vaccines. Ultimately, incorporating molecular profiling could enable personalized vaccine optimization.
Collapse
Affiliation(s)
- Cigdem Sevim Bayrak
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mt Sinai, New York, NY, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Christian V Forst
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mt Sinai, New York, NY, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Microbiology, Icahn School of Medicine at Mt Sinai, New York, NY, USA
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, NY, New York, USA
| | - David J Gresham
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Smruti Pushalkar
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Shaohuan Wu
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Christine Vogel
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Ted Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mt Sinai, New York, NY, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Meddeb M, Koleini N, Binek A, Keykhaei M, Darehgazani R, Kwon S, Aboaf C, Margulies KB, Bedi KC, Lehar M, Sharma K, Hahn VS, Van Eyk JE, Drachenberg CI, Kass DA. Myocardial ultrastructure of human heart failure with preserved ejection fraction. NATURE CARDIOVASCULAR RESEARCH 2024; 3:907-914. [PMID: 39196036 PMCID: PMC11498130 DOI: 10.1038/s44161-024-00516-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 06/26/2024] [Indexed: 08/29/2024]
Abstract
Over half of patients with heart failure have a preserved ejection fraction (>50%, called HFpEF), a syndrome with substantial morbidity/mortality and few effective therapies1. Its dominant comorbidity is now obesity, which worsens disease and prognosis1-3. Myocardial data from patients with morbid obesity and HFpEF show depressed myocyte calcium-stimulated tension4 and disrupted gene expression of mitochondrial and lipid metabolic pathways5,6, abnormalities shared by human HF with a reduced EF but less so in HFpEF without severe obesity. The impact of severe obesity on human HFpEF myocardial ultrastructure remains unexplored. Here we assessed the myocardial ultrastructure in septal biopsies from patients with HFpEF using transmission electron microscopy. We observed sarcomere disruption and sarcolysis, mitochondrial swelling with cristae separation and dissolution and lipid droplet accumulation that was more prominent in the most obese patients with HFpEF and not dependent on comorbid diabetes. Myocardial proteomics revealed associated reduction in fatty acid uptake, processing and oxidation and mitochondrial respiration proteins, particularly in very obese patients with HFpEF.
Collapse
Grants
- R01 HL149891 NHLBI NIH HHS
- HL166565-01 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL007227 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL149891 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32 HL007227 NHLBI NIH HHS
- R35 HL166565 NHLBI NIH HHS
- 20SRG35490443 American Heart Association (American Heart Association, Inc.)
- 23POST1026402 American Heart Association (American Heart Association, Inc.)
- R35 HL135827 NHLBI NIH HHS
- HL155346 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL166565 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL135827 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL155346 NHLBI NIH HHS
- K23HL166770 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K23 HL166770 NHLBI NIH HHS
- 16SFRN28620000 American Heart Association (American Heart Association, Inc.)
- L30 HL138884 NHLBI NIH HHS
Collapse
Affiliation(s)
- Mariam Meddeb
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Navid Koleini
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Aleksandra Binek
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mohammad Keykhaei
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Reyhane Darehgazani
- Department of Biological Sciences, University of Maryland, Baltimore, MD, USA
| | - Seoyoung Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Celia Aboaf
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ken C Bedi
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamed Lehar
- Department of Anesthesia, Johns Hopkins University, Baltimore, MD, USA
| | - Kavita Sharma
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Virginia S Hahn
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
10
|
Mansoor H, Lee IXY, Lin MTY, Ang HP, Xue YC, Krishaa L, Patil M, Koh SK, Tan HC, Zhou L, Liu YC. Topical and oral peroxisome proliferator-activated receptor-α agonist ameliorates diabetic corneal neuropathy. Sci Rep 2024; 14:13435. [PMID: 38862650 PMCID: PMC11167005 DOI: 10.1038/s41598-024-64451-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 06/10/2024] [Indexed: 06/13/2024] Open
Abstract
Diabetic corneal neuropathy (DCN) is a common diabetic ocular complication with limited treatment options. In this study, we investigated the effects of topical and oral fenofibrate, a peroxisome proliferator-activated receptor-α agonist, on the amelioration of DCN using diabetic mice (n = 120). Ocular surface assessments, corneal nerve and cell imaging analysis, tear proteomics and its associated biological pathways, immuno-histochemistry and western blot on PPARα expression, were studied before and 12 weeks after treatment. At 12 weeks, PPARα expression markedly restored after topical and oral fenofibrate. Topical fenofibrate significantly improved corneal nerve fibre density (CNFD) and tortuosity coefficient. Likewise, oral fenofibrate significantly improved CNFD. Both topical and oral forms significantly improved corneal sensitivity. Additionally, topical and oral fenofibrate significantly alleviated diabetic keratopathy, with fenofibrate eye drops demonstrating earlier therapeutic effects. Both topical and oral fenofibrate significantly increased corneal β-III tubulin expression. Topical fenofibrate reduced neuroinflammation by significantly increasing the levels of nerve growth factor and substance P. It also significantly increased β-III-tubulin and reduced CDC42 mRNA expression in trigeminal ganglions. Proteomic analysis showed that neurotrophin signalling and anti-inflammation reactions were significantly up-regulated after fenofibrate treatment, whether applied topically or orally. This study concluded that both topical and oral fenofibrate ameliorate DCN, while topical fenofibrate significantly reduces neuroinflammation.
Collapse
Affiliation(s)
| | - Isabelle Xin Yu Lee
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, 11 Third Hospital Ave, Singapore, 168751, Singapore
| | - Molly Tzu-Yu Lin
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, 11 Third Hospital Ave, Singapore, 168751, Singapore
| | - Heng Pei Ang
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, 11 Third Hospital Ave, Singapore, 168751, Singapore
| | - Yao Cong Xue
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, 11 Third Hospital Ave, Singapore, 168751, Singapore
| | - L Krishaa
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, 11 Third Hospital Ave, Singapore, 168751, Singapore
| | - Moushmi Patil
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, 11 Third Hospital Ave, Singapore, 168751, Singapore
| | - Siew-Kwan Koh
- Ocular Proteomic Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Hong Chang Tan
- Department of Endocrinology, Singapore General Hospital, Singapore, Singapore
| | - Lei Zhou
- Department of Applied Biology and Chemical Technology, School of Optometry, Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Pak Shek Kok, Hong Kong
| | - Yu-Chi Liu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, 11 Third Hospital Ave, Singapore, 168751, Singapore.
- Cornea and Refractive Surgery Group, Singapore Eye Research Institute, Singapore, Singapore.
- Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore, Singapore.
- Eye-Academic Clinical Program, Singapore Graduate Medical School, Duke-National University, Singapore, Singapore.
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
11
|
Alfahel L, Gschwendtberger T, Kozareva V, Dumas L, Gibbs R, Kertser A, Baruch K, Zaccai S, Kahn J, Thau-Habermann N, Eggenschwiler R, Sterneckert J, Hermann A, Sundararaman N, Vaibhav V, Van Eyk JE, Rafuse VF, Fraenkel E, Cantz T, Petri S, Israelson A. Targeting low levels of MIF expression as a potential therapeutic strategy for ALS. Cell Rep Med 2024; 5:101546. [PMID: 38703766 PMCID: PMC11148722 DOI: 10.1016/j.xcrm.2024.101546] [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: 05/17/2023] [Revised: 11/03/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by motor neuron (MN) loss. We previously discovered that macrophage migration inhibitory factor (MIF), whose levels are extremely low in spinal MNs, inhibits mutant SOD1 misfolding and toxicity. In this study, we show that a single peripheral injection of adeno-associated virus (AAV) delivering MIF into adult SOD1G37R mice significantly improves their motor function, delays disease progression, and extends survival. Moreover, MIF treatment reduces neuroinflammation and misfolded SOD1 accumulation, rescues MNs, and corrects dysregulated pathways as observed by proteomics and transcriptomics. Furthermore, we reveal low MIF levels in human induced pluripotent stem cell-derived MNs from familial ALS patients with different genetic mutations, as well as in post mortem tissues of sporadic ALS patients. Our findings indicate that peripheral MIF administration may provide a potential therapeutic mechanism for modulating misfolded SOD1 in vivo and disease outcome in ALS patients.
Collapse
Affiliation(s)
- Leenor Alfahel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel; The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel
| | - Thomas Gschwendtberger
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; Center for Systems Neuroscience, Hannover Medical School, 30625 Hannover, Germany
| | - Velina Kozareva
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Laura Dumas
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada; Brain Repair Centre, Life Sciences Research Institute, Halifax, Nova Scotia B3H 4R2, Canada
| | - Rachel Gibbs
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada; Brain Repair Centre, Life Sciences Research Institute, Halifax, Nova Scotia B3H 4R2, Canada
| | | | - Kuti Baruch
- ImmunoBrain Checkpoint Ltd., Ness Ziona 7404905, Israel
| | - Shir Zaccai
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel; The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel
| | - Joy Kahn
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel; The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel
| | | | - Reto Eggenschwiler
- Gastroenterology, Hepatology and Endocrinology Department, Hannover Medical School, 30625 Hannover, Germany; Translational Hepatology and Stem Cell Biology, REBIRTH - Research Center for Translational Regenerative Medicine and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Jared Sterneckert
- Center for Regenerative Therapies Dresden, Technical University Dresden, 01307 Dresden, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section, "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, 18147 Rostock, Germany; Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
| | - Niveda Sundararaman
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vineet Vaibhav
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Victor F Rafuse
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada; Brain Repair Centre, Life Sciences Research Institute, Halifax, Nova Scotia B3H 4R2, Canada
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tobias Cantz
- Gastroenterology, Hepatology and Endocrinology Department, Hannover Medical School, 30625 Hannover, Germany; Translational Hepatology and Stem Cell Biology, REBIRTH - Research Center for Translational Regenerative Medicine and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; Max Planck Institute for Molecular Biomedicine, Cell and Developmental Biology, 48149 Münster, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany; Center for Systems Neuroscience, Hannover Medical School, 30625 Hannover, Germany
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel; The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel.
| |
Collapse
|
12
|
Marchesin V, Monnier L, Blattmann P, Chevillard F, Kuntz C, Forny C, Kamper J, Studer R, Bossu A, Ertel EA, Nayler O, Brotschi C, Williams JT, Gatfield J. A uniquely efficacious type of CFTR corrector with complementary mode of action. SCIENCE ADVANCES 2024; 10:eadk1814. [PMID: 38427726 PMCID: PMC11801377 DOI: 10.1126/sciadv.adk1814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/26/2024] [Indexed: 03/03/2024]
Abstract
Three distinct pharmacological corrector types (I, II, III) with different binding sites and additive behavior only partially rescue the F508del-cystic fibrosis transmembrane conductance regulator (CFTR) folding and trafficking defect observed in cystic fibrosis. We describe uniquely effective, macrocyclic CFTR correctors that were additive to the known corrector types, exerting a complementary "type IV" corrector mechanism. Macrocycles achieved wild-type-like folding efficiency of F508del-CFTR at the endoplasmic reticulum and normalized CFTR currents in reconstituted patient-derived bronchial epithelium. Using photo-activatable macrocycles, docking studies and site-directed mutagenesis a highly probable binding site and pose for type IV correctors was identified in a cavity between lasso helix-1 (Lh1) and transmembrane helix-1 of membrane spanning domain (MSD)-1, distinct from the known corrector binding sites. Since only F508del-CFTR fragments spanning from Lh1 until MSD2 responded to type IV correctors, these likely promote cotranslational assembly of Lh1, MSD1, and MSD2. Previously corrector-resistant CFTR folding mutants were also robustly rescued, suggesting substantial therapeutic potential for type IV correctors.
Collapse
Affiliation(s)
| | - Lucile Monnier
- Idorsia Pharmaceuticals Ltd., 4123 Allschwil, Switzerland
| | | | | | | | - Camille Forny
- Idorsia Pharmaceuticals Ltd., 4123 Allschwil, Switzerland
| | - Judith Kamper
- Idorsia Pharmaceuticals Ltd., 4123 Allschwil, Switzerland
| | - Rolf Studer
- Idorsia Pharmaceuticals Ltd., 4123 Allschwil, Switzerland
| | | | - Eric A. Ertel
- Idorsia Pharmaceuticals Ltd., 4123 Allschwil, Switzerland
| | - Oliver Nayler
- Idorsia Pharmaceuticals Ltd., 4123 Allschwil, Switzerland
| | | | | | - John Gatfield
- Idorsia Pharmaceuticals Ltd., 4123 Allschwil, Switzerland
| |
Collapse
|
13
|
Pushalkar S, Wu S, Maity S, Pressler M, Rendleman J, Vitrinel B, Jeffery L, Abdelhadi R, Chen M, Ross T, Carlock M, Choi H, Vogel C. Complex changes in serum protein levels in COVID-19 convalescents. Sci Rep 2024; 14:4479. [PMID: 38396092 PMCID: PMC10891133 DOI: 10.1038/s41598-024-54534-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
The COVID-19 pandemic, triggered by severe acute respiratory syndrome coronavirus 2, has affected millions of people worldwide. Much research has been dedicated to our understanding of COVID-19 disease heterogeneity and severity, but less is known about recovery associated changes. To address this gap in knowledge, we quantified the proteome from serum samples from 29 COVID-19 convalescents and 29 age-, race-, and sex-matched healthy controls. Samples were acquired within the first months of the pandemic. Many proteins from pathways known to change during acute COVID-19 illness, such as from the complement cascade, coagulation system, inflammation and adaptive immune system, had returned to levels seen in healthy controls. In comparison, we identified 22 and 15 proteins with significantly elevated and lowered levels, respectively, amongst COVID-19 convalescents compared to healthy controls. Some of the changes were similar to those observed for the acute phase of the disease, i.e. elevated levels of proteins from hemolysis, the adaptive immune systems, and inflammation. In contrast, some alterations opposed those in the acute phase, e.g. elevated levels of CETP and APOA1 which function in lipid/cholesterol metabolism, and decreased levels of proteins from the complement cascade (e.g. C1R, C1S, and VWF), the coagulation system (e.g. THBS1 and VWF), and the regulation of the actin cytoskeleton (e.g. PFN1 and CFL1) amongst COVID-19 convalescents. We speculate that some of these shifts might originate from a transient decrease in platelet counts upon recovery from the disease. Finally, we observed race-specific changes, e.g. with respect to immunoglobulins and proteins related to cholesterol metabolism.
Collapse
Affiliation(s)
- Smruti Pushalkar
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| | - Shaohuan Wu
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Shuvadeep Maity
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
- Birla Institute of Technology and Science-Pilani (BITS Pilani), Hyderabad, India
| | - Matthew Pressler
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Justin Rendleman
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Burcu Vitrinel
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Lauren Jeffery
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Ryah Abdelhadi
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Mechi Chen
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Ted Ross
- Cleveland Clinic Florida Research & Innovation Center, Port St. Lucie, FL, USA
| | - Michael Carlock
- Cleveland Clinic Florida Research & Innovation Center, Port St. Lucie, FL, USA
| | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christine Vogel
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| |
Collapse
|
14
|
Osipov A, Nikolic O, Gertych A, Parker S, Hendifar A, Singh P, Filippova D, Dagliyan G, Ferrone CR, Zheng L, Moore JH, Tourtellotte W, Van Eyk JE, Theodorescu D. The Molecular Twin artificial-intelligence platform integrates multi-omic data to predict outcomes for pancreatic adenocarcinoma patients. NATURE CANCER 2024; 5:299-314. [PMID: 38253803 PMCID: PMC10899109 DOI: 10.1038/s43018-023-00697-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/30/2023] [Indexed: 01/24/2024]
Abstract
Contemporary analyses focused on a limited number of clinical and molecular biomarkers have been unable to accurately predict clinical outcomes in pancreatic ductal adenocarcinoma. Here we describe a precision medicine platform known as the Molecular Twin consisting of advanced machine-learning models and use it to analyze a dataset of 6,363 clinical and multi-omic molecular features from patients with resected pancreatic ductal adenocarcinoma to accurately predict disease survival (DS). We show that a full multi-omic model predicts DS with the highest accuracy and that plasma protein is the top single-omic predictor of DS. A parsimonious model learning only 589 multi-omic features demonstrated similar predictive performance as the full multi-omic model. Our platform enables discovery of parsimonious biomarker panels and performance assessment of outcome prediction models learning from resource-intensive panels. This approach has considerable potential to impact clinical care and democratize precision cancer medicine worldwide.
Collapse
Affiliation(s)
- Arsen Osipov
- Department of Medicine (Medical Oncology), Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Oncology, Pancreatic Cancer Precision Medicine Center of Excellence, Johns Hopkins University, Baltimore, MD, USA
| | | | - Arkadiusz Gertych
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sarah Parker
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrew Hendifar
- Department of Medicine (Medical Oncology), Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Grant Dagliyan
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cristina R Ferrone
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lei Zheng
- Department of Oncology, Pancreatic Cancer Precision Medicine Center of Excellence, Johns Hopkins University, Baltimore, MD, USA
| | - Jason H Moore
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Warren Tourtellotte
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer E Van Eyk
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dan Theodorescu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Bayrak CS, Forst C, Jones DR, Gresham D, Pushalkar S, Wu S, Vogel C, Mahal L, Ghedin E, Ross T, García-Sastre A, Zhang B. Patient Subtyping Analysis of Baseline Multi-omic Data Reveals Distinct Pre-immune States Predictive of Vaccination Responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576213. [PMID: 38328256 PMCID: PMC10849502 DOI: 10.1101/2024.01.18.576213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Understanding the molecular mechanisms that underpin diverse vaccination responses is a critical step toward developing efficient vaccines. Molecular subtyping approaches can offer valuable insights into the heterogeneous nature of responses and aid in the design of more effective vaccines. In order to explore the molecular signatures associated with the vaccine response, we analyzed baseline transcriptomics data from paired samples of whole blood, proteomics and glycomics data from serum, and metabolomics data from urine, obtained from influenza vaccine recipients (2019-2020 season) prior to vaccination. After integrating the data using a network-based model, we performed a subtyping analysis. The integration of multiple data modalities from 62 samples resulted in five baseline molecular subtypes with distinct molecular signatures. These baseline subtypes differed in the expression of pre-existing adaptive or innate immunity signatures, which were linked to significant variation across subtypes in baseline immunoglobulin A (IgA) and hemagglutination inhibition (HAI) titer levels. It is worth noting that these significant differences persisted through day 28 post-vaccination, indicating the effect of initial immune state on vaccination response. These findings highlight the significance of interpersonal variation in baseline immune status as a crucial factor in determining vaccine response and efficacy. Ultimately, incorporating molecular profiling could enable personalized vaccine optimization.
Collapse
|
16
|
Zila N, Eichhoff OM, Steiner I, Mohr T, Bileck A, Cheng PF, Leitner A, Gillet L, Sajic T, Goetze S, Friedrich B, Bortel P, Strobl J, Reitermaier R, Hogan SA, Martínez Gómez JM, Staeger R, Tuchmann F, Peters S, Stary G, Kuttke M, Elbe-Bürger A, Hoeller C, Kunstfeld R, Weninger W, Wollscheid B, Dummer R, French LE, Gerner C, Aebersold R, Levesque MP, Paulitschke V. Proteomic Profiling of Advanced Melanoma Patients to Predict Therapeutic Response to Anti-PD-1 Therapy. Clin Cancer Res 2024; 30:159-175. [PMID: 37861398 DOI: 10.1158/1078-0432.ccr-23-0562] [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: 03/01/2023] [Revised: 07/17/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023]
Abstract
PURPOSE Despite high clinical need, there are no biomarkers that accurately predict the response of patients with metastatic melanoma to anti-PD-1 therapy. EXPERIMENTAL DESIGN In this multicenter study, we applied protein depletion and enrichment methods prior to various proteomic techniques to analyze a serum discovery cohort (n = 56) and three independent serum validation cohorts (n = 80, n = 12, n = 17). Further validation analyses by literature and survival analysis followed. RESULTS We identified several significantly regulated proteins as well as biological processes such as neutrophil degranulation, cell-substrate adhesion, and extracellular matrix organization. Analysis of the three independent serum validation cohorts confirmed the significant differences between responders (R) and nonresponders (NR) observed in the initial discovery cohort. In addition, literature-based validation highlighted 30 markers overlapping with previously published signatures. Survival analysis using the TCGA database showed that overexpression of 17 of the markers we identified correlated with lower overall survival in patients with melanoma. CONCLUSIONS Ultimately, this multilayered serum analysis led to a potential marker signature with 10 key markers significantly altered in at least two independent serum cohorts: CRP, LYVE1, SAA2, C1RL, CFHR3, LBP, LDHB, S100A8, S100A9, and SAA1, which will serve as the basis for further investigation. In addition to patient serum, we analyzed primary melanoma tumor cells from NR and found a potential marker signature with four key markers: LAMC1, PXDN, SERPINE1, and VCAN.
Collapse
Affiliation(s)
- Nina Zila
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Ossia M Eichhoff
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Irene Steiner
- Center for Medical Data Science, Institute of Medical Statistics, Medical University of Vienna, Vienna, Austria
| | - Thomas Mohr
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Phil F Cheng
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alexander Leitner
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Ludovic Gillet
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Tatjana Sajic
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Sandra Goetze
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Zurich, Switzerland
| | - Betty Friedrich
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Patricia Bortel
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - René Reitermaier
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sabrina A Hogan
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Julia M Martínez Gómez
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ramon Staeger
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Felix Tuchmann
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sophie Peters
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Mario Kuttke
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | | | - Christoph Hoeller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Rainer Kunstfeld
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lars E French
- Department of Dermatology and Allergy University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Verena Paulitschke
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Plattner C, Lamberti G, Blattmann P, Kirchmair A, Rieder D, Loncova Z, Sturm G, Scheidl S, Ijsselsteijn M, Fotakis G, Noureen A, Lisandrelli R, Böck N, Nemati N, Krogsdam A, Daum S, Finotello F, Somarakis A, Schäfer A, Wilflingseder D, Gonzalez Acera M, Öfner D, Huber LA, Clevers H, Becker C, Farin HF, Greten FR, Aebersold R, de Miranda NF, Trajanoski Z. Functional and spatial proteomics profiling reveals intra- and intercellular signaling crosstalk in colorectal cancer. iScience 2023; 26:108399. [PMID: 38047086 PMCID: PMC10692669 DOI: 10.1016/j.isci.2023.108399] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/21/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
Precision oncology approaches for patients with colorectal cancer (CRC) continue to lag behind other solid cancers. Functional precision oncology-a strategy that is based on perturbing primary tumor cells from cancer patients-could provide a road forward to personalize treatment. We extend this paradigm to measuring proteome activity landscapes by acquiring quantitative phosphoproteomic data from patient-derived organoids (PDOs). We show that kinase inhibitors induce inhibitor- and patient-specific off-target effects and pathway crosstalk. Reconstruction of the kinase networks revealed that the signaling rewiring is modestly affected by mutations. We show non-genetic heterogeneity of the PDOs and upregulation of stemness and differentiation genes by kinase inhibitors. Using imaging mass-cytometry-based profiling of the primary tumors, we characterize the tumor microenvironment (TME) and determine spatial heterocellular crosstalk and tumor-immune cell interactions. Collectively, we provide a framework for inferring tumor cell intrinsic signaling and external signaling from the TME to inform precision (immuno-) oncology in CRC.
Collapse
Affiliation(s)
- Christina Plattner
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Giorgia Lamberti
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Peter Blattmann
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8092 Zurich, Switzerland
| | - Alexander Kirchmair
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dietmar Rieder
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Zuzana Loncova
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Gregor Sturm
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stefan Scheidl
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Marieke Ijsselsteijn
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Georgios Fotakis
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Asma Noureen
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Rebecca Lisandrelli
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Nina Böck
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Niloofar Nemati
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Anne Krogsdam
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Sophia Daum
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Francesca Finotello
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Antonios Somarakis
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Alexander Schäfer
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8092 Zurich, Switzerland
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Miguel Gonzalez Acera
- Department of Medicine 1, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Dietmar Öfner
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Lukas A. Huber
- Biocenter, Institute of Cell Biology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Hans Clevers
- Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Henner F. Farin
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, a partnership with DKFZ Heidelberg, Frankfurt/Mainz, Germany
| | - Florian R. Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, a partnership with DKFZ Heidelberg, Frankfurt/Mainz, Germany
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8092 Zurich, Switzerland
| | - Noel F.C.C. de Miranda
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
18
|
Verma N, Khare D, Poe AJ, Amador C, Ghiam S, Fealy A, Ebrahimi S, Shadrokh O, Song XY, Santiskulvong C, Mastali M, Parker S, Stotland A, Van Eyk JE, Ljubimov AV, Saghizadeh M. MicroRNA and Protein Cargos of Human Limbal Epithelial Cell-Derived Exosomes and Their Regulatory Roles in Limbal Stromal Cells of Diabetic and Non-Diabetic Corneas. Cells 2023; 12:2524. [PMID: 37947602 PMCID: PMC10649916 DOI: 10.3390/cells12212524] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/08/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023] Open
Abstract
Epithelial and stromal/mesenchymal limbal stem cells contribute to corneal homeostasis and cell renewal. Extracellular vesicles (EVs), including exosomes (Exos), can be paracrine mediators of intercellular communication. Previously, we described cargos and regulatory roles of limbal stromal cell (LSC)-derived Exos in non-diabetic (N) and diabetic (DM) limbal epithelial cells (LECs). Presently, we quantify the miRNA and proteome profiles of human LEC-derived Exos and their regulatory roles in N- and DM-LSC. We revealed some miRNA and protein differences in DM vs. N-LEC-derived Exos' cargos, including proteins involved in Exo biogenesis and packaging that may affect Exo production and ultimately cellular crosstalk and corneal function. Treatment by N-Exos, but not by DM-Exos, enhanced wound healing in cultured N-LSCs and increased proliferation rates in N and DM LSCs vs. corresponding untreated (control) cells. N-Exos-treated LSCs reduced the keratocyte markers ALDH3A1 and lumican and increased the MSC markers CD73, CD90, and CD105 vs. control LSCs. These being opposite to the changes quantified in wounded LSCs. Overall, N-LEC Exos have a more pronounced effect on LSC wound healing, proliferation, and stem cell marker expression than DM-LEC Exos. This suggests that regulatory miRNA and protein cargo differences in DM- vs. N-LEC-derived Exos could contribute to the disease state.
Collapse
Affiliation(s)
- Nagendra Verma
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Drirh Khare
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Adam J. Poe
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Cynthia Amador
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sean Ghiam
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
- Sackler School of Medicine, New York State/American Program of Tel Aviv University, Tel Aviv 6997801, Israel
| | - Andrew Fealy
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shaghaiegh Ebrahimi
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Odelia Shadrokh
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xue-Ying Song
- Genomics Core, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (X.-Y.S.); (C.S.)
| | - Chintda Santiskulvong
- Genomics Core, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (X.-Y.S.); (C.S.)
| | - Mitra Mastali
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (M.M.); (S.P.); (A.S.); (J.E.V.E.)
| | - Sarah Parker
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (M.M.); (S.P.); (A.S.); (J.E.V.E.)
| | - Aleksandr Stotland
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (M.M.); (S.P.); (A.S.); (J.E.V.E.)
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (M.M.); (S.P.); (A.S.); (J.E.V.E.)
| | - Alexander V. Ljubimov
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
| | - Mehrnoosh Saghizadeh
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, 8700 Beverly Boulevard, AHSP-A8104, Los Angeles, CA 90048, USA; (N.V.); (D.K.); (C.A.); (A.F.); (S.E.); (O.S.); (A.V.L.)
- Departments of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
| |
Collapse
|
19
|
Yu F, Teo GC, Kong AT, Fröhlich K, Li GX, Demichev V, Nesvizhskii AI. Analysis of DIA proteomics data using MSFragger-DIA and FragPipe computational platform. Nat Commun 2023; 14:4154. [PMID: 37438352 PMCID: PMC10338508 DOI: 10.1038/s41467-023-39869-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
Liquid chromatography (LC) coupled with data-independent acquisition (DIA) mass spectrometry (MS) has been increasingly used in quantitative proteomics studies. Here, we present a fast and sensitive approach for direct peptide identification from DIA data, MSFragger-DIA, which leverages the unmatched speed of the fragment ion indexing-based search engine MSFragger. Different from most existing methods, MSFragger-DIA conducts a database search of the DIA tandem mass (MS/MS) spectra prior to spectral feature detection and peak tracing across the LC dimension. To streamline the analysis of DIA data and enable easy reproducibility, we integrate MSFragger-DIA into the FragPipe computational platform for seamless support of peptide identification and spectral library building from DIA, data-dependent acquisition (DDA), or both data types combined. We compare MSFragger-DIA with other DIA tools, such as DIA-Umpire based workflow in FragPipe, Spectronaut, DIA-NN library-free, and MaxDIA. We demonstrate the fast, sensitive, and accurate performance of MSFragger-DIA across a variety of sample types and data acquisition schemes, including single-cell proteomics, phosphoproteomics, and large-scale tumor proteome profiling studies.
Collapse
Affiliation(s)
- Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| | - Guo Ci Teo
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Andy T Kong
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Klemens Fröhlich
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Ginny Xiaohe Li
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Vadim Demichev
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
20
|
Ammar C, Schessner JP, Willems S, Michaelis AC, Mann M. Accurate Label-Free Quantification by directLFQ to Compare Unlimited Numbers of Proteomes. Mol Cell Proteomics 2023; 22:100581. [PMID: 37225017 PMCID: PMC10315922 DOI: 10.1016/j.mcpro.2023.100581] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023] Open
Abstract
Recent advances in mass spectrometry-based proteomics enable the acquisition of increasingly large datasets within relatively short times, which exposes bottlenecks in the bioinformatics pipeline. Although peptide identification is already scalable, most label-free quantification (LFQ) algorithms scale quadratic or cubic with the sample numbers, which may even preclude the analysis of large-scale data. Here we introduce directLFQ, a ratio-based approach for sample normalization and the calculation of protein intensities. It estimates quantities via aligning samples and ion traces by shifting them on top of each other in logarithmic space. Importantly, directLFQ scales linearly with the number of samples, allowing analyses of large studies to finish in minutes instead of days or months. We quantify 10,000 proteomes in 10 min and 100,000 proteomes in less than 2 h, a 1000-fold faster than some implementations of the popular LFQ algorithm MaxLFQ. In-depth characterization of directLFQ reveals excellent normalization properties and benchmark results, comparing favorably to MaxLFQ for both data-dependent acquisition and data-independent acquisition. In addition, directLFQ provides normalized peptide intensity estimates for peptide-level comparisons. It is an important part of an overall quantitative proteomic pipeline that also needs to include high sensitive statistical analysis leading to proteoform resolution. Available as an open-source Python package and a graphical user interface with a one-click installer, it can be used in the AlphaPept ecosystem as well as downstream of most common computational proteomics pipelines.
Collapse
Affiliation(s)
- Constantin Ammar
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Julia Patricia Schessner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Sander Willems
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - André C Michaelis
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
21
|
Bokor BJ, Gorhe D, Jovanovic M, Rosenberger G. Network-centric analysis of co-fractionated protein complex profiles using SECAT. STAR Protoc 2023; 4:102293. [PMID: 37182203 PMCID: PMC10199247 DOI: 10.1016/j.xpro.2023.102293] [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: 01/06/2023] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 05/16/2023] Open
Abstract
The Size-Exclusion Chromatography Analysis Toolkit (SECAT) elucidates protein complex dynamics using co-fractionated bottom-up mass spectrometry (CF-MS) data. Here, we present a protocol for the network-centric analysis and interpretation of CF-MS profiles using SECAT. We describe the technical steps for preprocessing, scoring, semi-supervised machine learning, and quantification, including common pitfalls and their solutions. We further provide guidance for data export, visualization, and the interpretation of SECAT results to discover dysregulated proteins and interactions, supporting new hypotheses and biological insights. For complete details on the use and execution of this protocol, please refer to Rosenberger et al. (2020).1.
Collapse
Affiliation(s)
- Benjamin J Bokor
- Department of Biological Sciences, Columbia University, New York City, NY 10027, USA.
| | - Darvesh Gorhe
- Department of Biological Sciences, Columbia University, New York City, NY 10027, USA
| | - Marko Jovanovic
- Department of Biological Sciences, Columbia University, New York City, NY 10027, USA
| | - George Rosenberger
- Department of Systems Biology, Columbia University, New York City, NY 10032, USA.
| |
Collapse
|
22
|
Allgoewer K, Wu S, Choi H, Vogel C. Re-mining serum proteomics data reveals extensive post-translational modifications upon Zika and dengue infection. Mol Omics 2023; 19:308-320. [PMID: 36810580 PMCID: PMC10175154 DOI: 10.1039/d2mo00258b] [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] [Indexed: 02/17/2023]
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are two closely related flaviviruses with similar symptoms. However, due to the implications of ZIKV infections for pregnancy outcomes, understanding differences in their molecular impact on the host is of high interest. Viral infections change the host proteome, including post-translational modifications. As modifications are diverse and of low abundance, they typically require additional sample processing which is not feasible for large cohort studies. Therefore, we tested the potential of next-generation proteomics data in its ability to prioritize specific modifications for later analysis. We re-mined published mass spectra from 122 serum samples from ZIKV and DENV patients for the presence of phosphorylated, methylated, oxidized, glycosylated/glycated, sulfated, and carboxylated peptides. We identified 246 modified peptides with significantly differential abundance in ZIKV and DENV patients. Amongst these, methionine-oxidized peptides from apolipoproteins and glycosylated peptides from immunoglobulin proteins were more abundant in ZIKV patient serum and generate hypotheses on the potential roles of the modification in the infection. The results demonstrate how data-independent acquisition techniques can help prioritize future analyses of peptide modifications.
Collapse
Affiliation(s)
- Kristina Allgoewer
- New York University, Department of Biology, Center for Genomics and Systems Biology, New York, NY, USA.
- Humboldt University, Department of Biology, Berlin, Germany
| | - Shaohuan Wu
- New York University, Department of Biology, Center for Genomics and Systems Biology, New York, NY, USA.
| | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University, Singapore, Singapore
| | - Christine Vogel
- New York University, Department of Biology, Center for Genomics and Systems Biology, New York, NY, USA.
| |
Collapse
|
23
|
Norby FL, Nakamura K, Fu Q, Venkatraman V, Sundararaman N, Mastali M, Reinier K, Salvucci A, Jui J, Van Eyk JE, Chugh SS. A panel of blood biomarkers unique to sudden cardiac arrest. Heart Rhythm 2023; 20:414-422. [PMID: 36521734 PMCID: PMC9974970 DOI: 10.1016/j.hrthm.2022.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/17/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND The identification of circulating biomarkers specific for sudden cardiac arrest (SCA) could enhance risk prediction. Of particular interest are biomarkers specific to SCA, independent of coronary artery disease (CAD). OBJECTIVE The purpose of this study was to identify biomarkers of SCA obtained close to the SCA event. METHODS Twenty cases (survivors of SCA) and 40 age- and sex-matched controls were compared, with a replication analysis of 29 cases matched to 57 controls. A secondary analysis compared 20 SCA cases to 20 controls with CAD. Blood samples were obtained from SCA survivors at a median of 11 months after the SCA event. Proteins were analyzed on a mass spectrometer using data-independent acquisition; a subset of cytokines were analyzed using immunoassays; and 1153 lipids (13 classes) were analyzed. A false discovery rate P value of <.05 identified associated proteins. RESULTS Patients had a mean age of 58 years (range 25-87 years), and 70% were male. A total of 26 protein biomarkers associated with SCA when cases were compared with controls, of which 20 differentiated SCA from CAD. The replication analysis identified 8 of 26 biomarkers, of which 6 were not overlapping with CAD. The top identified biological processes involved the extracellular matrix, coagulation cascades, and platelet activation. Lipids in the lysophosphatidylcholine class were implicated in SCA through the CAD pathway. CONCLUSION We identified a panel of novel blood biomarkers specifically associated with SCA, including several that may be involved outside the CAD pathway. These biomarkers could have mechanistic significance and the potential to enhance clinical prediction of SCA.
Collapse
Affiliation(s)
- Faye L Norby
- Center for Cardiac Arrest Prevention, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, California
| | - Kotoka Nakamura
- Center for Cardiac Arrest Prevention, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, California
| | - Qin Fu
- Advanced Clinical Biosystems Research Institute at Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute at Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute at Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Mitra Mastali
- Advanced Clinical Biosystems Research Institute at Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kyndaron Reinier
- Center for Cardiac Arrest Prevention, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, California
| | | | - Jonathan Jui
- Oregon Health and Science University, Portland, Oregon
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute at Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Sumeet S Chugh
- Center for Cardiac Arrest Prevention, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, California.
| |
Collapse
|
24
|
Sundararaman N, Bhat A, Venkatraman V, Binek A, Dwight Z, Ariyasinghe NR, Escopete S, Joung SY, Cheng S, Parker SJ, Fert-Bober J, Van Eyk JE. BIRCH: An Automated Workflow for Evaluation, Correction, and Visualization of Batch Effect in Bottom-Up Mass Spectrometry-Based Proteomics Data. J Proteome Res 2023; 22:471-481. [PMID: 36695565 DOI: 10.1021/acs.jproteome.2c00671] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Recent surges in large-scale mass spectrometry (MS)-based proteomics studies demand a concurrent rise in methods to facilitate reliable and reproducible data analysis. Quantification of proteins in MS analysis can be affected by variations in technical factors such as sample preparation and data acquisition conditions leading to batch effects, which adds to noise in the data set. This may in turn affect the effectiveness of any biological conclusions derived from the data. Here we present Batch-effect Identification, Representation, and Correction of Heterogeneous data (BIRCH), a workflow for analysis and correction of batch effect through an automated, versatile, and easy to use web-based tool with the goal of eliminating technical variation. BIRCH also supports diagnosis of the data to check for the presence of batch effects, feasibility of batch correction, and imputation to deal with missing values in the data set. To illustrate the relevance of the tool, we explore two case studies, including an iPSC-derived cell study and a Covid vaccine study to show different context-specific use cases. Ultimately this tool can be used as an extremely powerful approach for eliminating technical bias while retaining biological bias, toward understanding disease mechanisms and potential therapeutics.
Collapse
Affiliation(s)
- Niveda Sundararaman
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Archana Bhat
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Vidya Venkatraman
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Aleksandra Binek
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Zachary Dwight
- Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Nethika R Ariyasinghe
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Sean Escopete
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Sandy Y Joung
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Sarah J Parker
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Justyna Fert-Bober
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| |
Collapse
|
25
|
Robinson AE, Binek A, Ramani K, Sundararaman N, Barbier-Torres L, Murray B, Venkatraman V, Kreimer S, Ardle AM, Noureddin M, Fernández-Ramos D, Lopitz-Otsoa F, Gutiérrez de Juan V, Millet O, Mato JM, Lu SC, Van Eyk JE. Hyperphosphorylation of hepatic proteome characterizes nonalcoholic fatty liver disease in S-adenosylmethionine deficiency. iScience 2023; 26:105987. [PMID: 36756374 PMCID: PMC9900401 DOI: 10.1016/j.isci.2023.105987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/15/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Methionine adenosyltransferase 1a (MAT1A) is responsible for hepatic S-adenosyl-L-methionine (SAMe) biosynthesis. Mat1a -/- mice have hepatic SAMe depletion, develop nonalcoholic steatohepatitis (NASH) which is reversed with SAMe administration. We examined temporal alterations in the proteome/phosphoproteome in pre-disease and NASH Mat1a -/- mice, effects of SAMe administration, and compared to human nonalcoholic fatty liver disease (NAFLD). Mitochondrial and peroxisomal lipid metabolism proteins were altered in pre-disease mice and persisted in NASH Mat1a -/- mice, which exhibited more progressive alterations in cytoplasmic ribosomes, ER, and nuclear proteins. A common mechanism found in both pre-disease and NASH livers was a hyperphosphorylation signature consistent with casein kinase 2α (CK2α) and AKT1 activation, which was normalized by SAMe administration. This was mimicked in human NAFLD with a metabolomic signature (M-subtype) resembling Mat1a -/- mice. In conclusion, we have identified a common proteome/phosphoproteome signature between Mat1a -/- mice and human NAFLD M-subtype that may have pathophysiological and therapeutic implications.
Collapse
Affiliation(s)
- Aaron E. Robinson
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Aleksandra Binek
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Komal Ramani
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
| | - Ben Murray
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Simion Kreimer
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Angela Mc Ardle
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Mazen Noureddin
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
| | - David Fernández-Ramos
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Fernando Lopitz-Otsoa
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Virginia Gutiérrez de Juan
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Oscar Millet
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - José M. Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
- Corresponding author
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
- Corresponding author
| |
Collapse
|
26
|
Matlock AD, Vaibhav V, Holewinski R, Venkatraman V, Dardov V, Manalo DM, Shelley B, Ornelas L, Banuelos M, Mandefro B, Escalante-Chong R, Li J, Finkbeiner S, Fraenkel E, Rothstein J, Thompson L, Sareen D, Svendsen CN, Van Eyk JE. NeuroLINCS Proteomics: Defining human-derived iPSC proteomes and protein signatures of pluripotency. Sci Data 2023; 10:24. [PMID: 36631473 PMCID: PMC9834231 DOI: 10.1038/s41597-022-01687-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 09/07/2022] [Indexed: 01/13/2023] Open
Abstract
The National Institute of Health (NIH) Library of integrated network-based cellular signatures (LINCS) program is premised on the generation of a publicly available data resource of cell-based biochemical responses or "signatures" to genetic or environmental perturbations. NeuroLINCS uses human inducible pluripotent stem cells (hiPSCs), derived from patients and healthy controls, and differentiated into motor neuron cell cultures. This multi-laboratory effort strives to establish i) robust multi-omic workflows for hiPSC and differentiated neuronal cultures, ii) public annotated data sets and iii) relevant and targetable biological pathways of spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS). Here, we focus on the proteomics and the quality of the developed workflow of hiPSC lines from 6 individuals, though epigenomics and transcriptomics data are also publicly available. Known and commonly used markers representing 73 proteins were reproducibly quantified with consistent expression levels across all hiPSC lines. Data quality assessments, data levels and metadata of all 6 genetically diverse human iPSCs analysed by DIA-MS are parsable and available as a high-quality resource to the public.
Collapse
Affiliation(s)
- Andrea D Matlock
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Vineet Vaibhav
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ronald Holewinski
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Vidya Venkatraman
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Victoria Dardov
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Danica-Mae Manalo
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Brandon Shelley
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Loren Ornelas
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Maria Banuelos
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Berhan Mandefro
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | | | - Jonathan Li
- NeuroLINCS, Department of Biological Engineering, MIT, Cambridge, MA, 02142, USA
| | - Steve Finkbeiner
- NeuroLINCS, Gladstone Institute of Neurological Disease and the Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Ernest Fraenkel
- NeuroLINCS, Department of Biological Engineering, MIT, Cambridge, MA, 02142, USA
| | - Jeffrey Rothstein
- NeuroLINCS, Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Leslie Thompson
- NeuroLINCS, Departments of Psychiatry and Human Behaviour, Neurobiology and Behaviour and UCI MIND, University of California Irvine, Irvine, CA, 92697, USA
| | - Dhruv Sareen
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Clive N Svendsen
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Jennifer E Van Eyk
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
27
|
Pamies D, Vujić T, Schvartz D, Boccard J, Repond C, Nunes C, Rudaz S, Sanchez JC, González-Ruiz V, Zurich MG. Digoxin Induces Human Astrocyte Reaction In Vitro. Mol Neurobiol 2023; 60:84-97. [PMID: 36223047 PMCID: PMC9758102 DOI: 10.1007/s12035-022-03057-1] [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: 06/22/2022] [Accepted: 09/21/2022] [Indexed: 12/30/2022]
Abstract
Astrocyte reaction is a complex cellular process involving astrocytes in response to various types of CNS injury and a marker of neurotoxicity. It has been abundantly studied in rodents but relatively poorly in human cells due to limited access to the brain. Astrocytes play important roles in cerebral energy metabolism and are also key players in neuroinflammation. Astroglial metabolic and inflammatory changes have been reported with age, leading to the hypothesis that mitochondrial metabolism and inflammatory responses are interconnected. However, the relationship between energy metabolism and astrocyte reactivity in the context of neurotoxicity is not known. We hypothesized that changes in energy metabolism of astrocytes will be coupled to their activation by xenobiotics. Astrocyte reaction and associated energy metabolic changes were assessed by immunostaining, gene expression, proteomics, metabolomics, and extracellular flux analyses after 24 h of exposure of human ReN-derived astrocytes to digoxin (1-10 µM) or TNFα (30 ng/ml) used as a positive control. Strong astrocytic reaction was observed, accompanied by increased glycolysis at low concentrations of digoxin (0.1 and 0.5 µM) and after TNFα exposure, suggesting that increased glycolysis may be a common feature of reactive astrocytes, independent of the triggering molecule. In conclusion, whether astrocyte activation is triggered by cytokines or a xenobiotic, it is strongly tied to energy metabolism in human ReN-derived astrocytes. Increased glycolysis might be considered as an endpoint to detect astrocyte activation by potentially neurotoxic compounds in vitro. Finally, ReN-derived astrocytes may help to decipher mechanisms of neurotoxicity in ascertaining the ability of chemicals to directly target astrocytes.
Collapse
Affiliation(s)
- David Pamies
- Department of Biological Sciences, University of Lausanne, Lausanne, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Tatjana Vujić
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Domitille Schvartz
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Julien Boccard
- Translational Biomarker Group, Department of Medicine, University of Geneva, Geneva, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Cendrine Repond
- Department of Biological Sciences, University of Lausanne, Lausanne, Switzerland
| | - Carolina Nunes
- Department of Biological Sciences, University of Lausanne, Lausanne, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Serge Rudaz
- Translational Biomarker Group, Department of Medicine, University of Geneva, Geneva, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Víctor González-Ruiz
- Translational Biomarker Group, Department of Medicine, University of Geneva, Geneva, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Marie-Gabrielle Zurich
- Department of Biological Sciences, University of Lausanne, Lausanne, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| |
Collapse
|
28
|
Vujić T, Schvartz D, Furlani IL, Meister I, González-Ruiz V, Rudaz S, Sanchez JC. Oxidative Stress and Extracellular Matrix Remodeling Are Signature Pathways of Extracellular Vesicles Released upon Morphine Exposure on Human Brain Microvascular Endothelial Cells. Cells 2022; 11:cells11233926. [PMID: 36497184 PMCID: PMC9741159 DOI: 10.3390/cells11233926] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/25/2022] [Accepted: 11/01/2022] [Indexed: 12/09/2022] Open
Abstract
Morphine, a commonly used antinociceptive drug in hospitals, is known to cross the blood-brain barrier (BBB) by first passing through brain endothelial cells. Despite its pain-relieving effect, morphine also has detrimental effects, such as the potential induction of redox imbalance in the brain. However, there is still insufficient evidence of these effects on the brain, particularly on the brain endothelial cells and the extracellular vesicles that they naturally release. Indeed, extracellular vesicles (EVs) are nanosized bioparticles produced by almost all cell types and are currently thought to reflect the physiological state of their parent cells. These vesicles have emerged as a promising source of biomarkers by indicating the functional or dysfunctional state of their parent cells and, thus, allowing a better understanding of the biological processes involved in an adverse state. However, there is very little information on the morphine effect on human brain microvascular endothelial cells (HBMECs), and even less on their released EVs. Therefore, the current study aimed at unraveling the detrimental mechanisms of morphine exposure (at 1, 10, 25, 50 and 100 µM) for 24 h on human brain microvascular endothelial cells as well as on their associated EVs. Isolation of EVs was carried out using an affinity-based method. Several orthogonal techniques (NTA, western blotting and proteomics analysis) were used to validate the EVs enrichment, quality and concentration. Data-independent mass spectrometry (DIA-MS)-based proteomics was applied in order to analyze the proteome modulations induced by morphine on HBMECs and EVs. We were able to quantify almost 5500 proteins in HBMECs and 1500 proteins in EVs, of which 256 and 148, respectively, were found to be differentially expressed in at least one condition. Pathway enrichment analysis revealed that the "cell adhesion and extracellular matrix remodeling" process and the "HIF1 pathway", a pathway related to oxidative stress responses, were significantly modulated upon morphine exposure in HBMECs and EVs. Altogether, the combination of proteomics and bioinformatics findings highlighted shared pathways between HBMECs exposed to morphine and their released EVs. These results put forward molecular signatures of morphine-induced toxicity in HBMECs that were also carried by EVs. Therefore, EVs could potentially be regarded as a useful tool to investigate brain endothelial cells dysfunction, and to a different extent, the BBB dysfunction in patient circulation using these "signature pathways".
Collapse
Affiliation(s)
- Tatjana Vujić
- Department of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | | | - Izadora Liranço Furlani
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Department of Chemistry, Federal University of São Carlos, São Carlos 13565-904, Brazil
| | - Isabel Meister
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Swiss Centre for Applied Human Toxicology, 4055 Basel, Switzerland
| | - Víctor González-Ruiz
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Swiss Centre for Applied Human Toxicology, 4055 Basel, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Swiss Centre for Applied Human Toxicology, 4055 Basel, Switzerland
| | - Jean-Charles Sanchez
- Department of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Correspondence: ; Tel.: +41-22-379-54-86
| |
Collapse
|
29
|
Wu S, Pushalkar S, Maity S, Pressler M, Rendleman J, Vitrinel B, Carlock M, Ross T, Choi H, Vogel C. Proteomic Signatures of the Serological Response to Influenza Vaccination in a Large Human Cohort Study. Viruses 2022; 14:v14112479. [PMID: 36366577 PMCID: PMC9696600 DOI: 10.3390/v14112479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
The serological response to the influenza virus vaccine is highly heterogeneous for reasons that are not entirely clear. While the impact of demographic factors such as age, body mass index (BMI), sex, prior vaccination and titer levels are known to impact seroconversion, they only explain a fraction of the response. To identify signatures of the vaccine response, we analyzed 273 protein levels from 138 serum samples of influenza vaccine recipients (2019-2020 season). We found that levels of proteins functioning in cholesterol transport were positively associated with seroconversion, likely linking to the known impact of BMI. When adjusting seroconversion for the demographic factors, we identified additional, unexpected signatures: proteins regulating actin cytoskeleton dynamics were significantly elevated in participants with high adjusted seroconversion. Viral strain specific analysis showed that this trend was largely driven by the H3N2 strain. Further, we identified complex associations between adjusted seroconversion and other factors: levels of proteins of the complement system associated positively with adjusted seroconversion in younger participants, while they were associated negatively in the older population. We observed the opposite trends for proteins of high density lipoprotein remodeling, transcription, and hemostasis. In sum, careful integrative modeling can extract new signatures of seroconversion from highly variable data that suggest links between the humoral response as well as immune cell communication and migration.
Collapse
Affiliation(s)
- Shaohuan Wu
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- Correspondence: (S.W.); (C.V.)
| | - Smruti Pushalkar
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Shuvadeep Maity
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- Birla Institute of Technology and Science (BITS)-Pilani (Hyderabad Campus), Hyderabad 500078, India
| | - Matthew Pressler
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Justin Rendleman
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Burcu Vitrinel
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Michael Carlock
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - Ted Ross
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Christine Vogel
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- Correspondence: (S.W.); (C.V.)
| |
Collapse
|
30
|
Mackmull MT, Nagel L, Sesterhenn F, Muntel J, Grossbach J, Stalder P, Bruderer R, Reiter L, van de Berg WDJ, de Souza N, Beyer A, Picotti P. Global, in situ analysis of the structural proteome in individuals with Parkinson's disease to identify a new class of biomarker. Nat Struct Mol Biol 2022; 29:978-989. [PMID: 36224378 DOI: 10.1038/s41594-022-00837-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/18/2022] [Indexed: 12/23/2022]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease for which robust biomarkers are needed. Because protein structure reflects function, we tested whether global, in situ analysis of protein structural changes provides insight into PD pathophysiology and could inform a new concept of structural disease biomarkers. Using limited proteolysis-mass spectrometry (LiP-MS), we identified 76 structurally altered proteins in cerebrospinal fluid (CSF) of individuals with PD relative to healthy donors. These proteins were enriched in processes misregulated in PD, and some proteins also showed structural changes in PD brain samples. CSF protein structural information outperformed abundance information in discriminating between healthy participants and those with PD and improved the discriminatory performance of CSF measures of the hallmark PD protein α-synuclein. We also present the first analysis of inter-individual variability of a structural proteome in healthy individuals, identifying biophysical features of variable protein regions. Although independent validation is needed, our data suggest that global analyses of the human structural proteome will guide the development of novel structural biomarkers of disease and enable hypothesis generation about underlying disease processes.
Collapse
Affiliation(s)
- Marie-Therese Mackmull
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Luise Nagel
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Fabian Sesterhenn
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | - Jan Grossbach
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Patrick Stalder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | | | - Wilma D J van de Berg
- Amsterdam UMC location Vrije Universiteit Amsterdam, Section Clinical Neuroanatomy and Biobanking, Department Anatomy and Neurosciences, Amsterdam, the Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Natalie de Souza
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.,Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Andreas Beyer
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany. .,Faculty of Medicine and University Hospital of Cologne, and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany. .,Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| | - Paola Picotti
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
31
|
Yang Y, Qiao L. Data-independent acquisition proteomics methods for analyzing post-translational modifications. Proteomics 2022; 23:e2200046. [PMID: 36036492 DOI: 10.1002/pmic.202200046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 08/20/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Abstract
Protein post-translational modifications (PTMs) increase the functional diversity of the cellular proteome. Accurate and high throughput identification and quantification of protein PTMs is a key task in proteomics research. Recent advancements in data-independent acquisition (DIA) mass spectrometry (MS) technology have achieved deep coverage and accurate quantification of proteins and PTMs. This review provides an overview of DIA data processing methods that cover three aspects of PTMs analysis, i.e., detection of PTMs, site localization, and characterization of complex modification moieties, such as glycosylation. In addition, a survey of deep learning methods that boost DIA-based PTMs analysis is presented, including in silico spectral library generation, as well as feature scoring and error rate control. The limitations and future directions of DIA methods for PTMs analysis are also discussed. Novel data analysis methods will take advantage of advanced MS instrumentation techniques to empower DIA MS for in-depth and accurate PTMs measurements. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yi Yang
- Department of Chemistry, and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
| | - Liang Qiao
- Department of Chemistry, and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
| |
Collapse
|
32
|
Lancaster SM, Lee-McMullen B, Abbott CW, Quijada JV, Hornburg D, Park H, Perelman D, Peterson DJ, Tang M, Robinson A, Ahadi S, Contrepois K, Hung CJ, Ashland M, McLaughlin T, Boonyanit A, Horning A, Sonnenburg JL, Snyder MP. Global, distinctive, and personal changes in molecular and microbial profiles by specific fibers in humans. Cell Host Microbe 2022; 30:848-862.e7. [PMID: 35483363 PMCID: PMC9187607 DOI: 10.1016/j.chom.2022.03.036] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/19/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022]
Abstract
Dietary fibers act through the microbiome to improve cardiovascular health and prevent metabolic disorders and cancer. To understand the health benefits of dietary fiber supplementation, we investigated two popular purified fibers, arabinoxylan (AX) and long-chain inulin (LCI), and a mixture of five fibers. We present multiomic signatures of metabolomics, lipidomics, proteomics, metagenomics, a cytokine panel, and clinical measurements on healthy and insulin-resistant participants. Each fiber is associated with fiber-dependent biochemical and microbial responses. AX consumption associates with a significant reduction in LDL and an increase in bile acids, contributing to its observed cholesterol reduction. LCI is associated with an increase in Bifidobacterium. However, at the highest LCI dose, there is increased inflammation and elevation in the liver enzyme alanine aminotransferase. This study yields insights into the effects of fiber supplementation and the mechanisms behind fiber-induced cholesterol reduction, and it shows effects of individual, purified fibers on the microbiome.
Collapse
Affiliation(s)
- Samuel M Lancaster
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Brittany Lee-McMullen
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Charles Wilbur Abbott
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jeniffer V Quijada
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Daniel Hornburg
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Heyjun Park
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dalia Perelman
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dylan J Peterson
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michael Tang
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Aaron Robinson
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Sara Ahadi
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Chia-Jui Hung
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Melanie Ashland
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Tracey McLaughlin
- Division of Endocrinology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Anna Boonyanit
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Aaron Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Justin L Sonnenburg
- Department of Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
33
|
Grossbach J, Gillet L, Clément‐Ziza M, Schmalohr CL, Schubert OT, Schütter M, Mawer JSP, Barnes CA, Bludau I, Weith M, Tessarz P, Graef M, Aebersold R, Beyer A. The impact of genomic variation on protein phosphorylation states and regulatory networks. Mol Syst Biol 2022; 18:e10712. [PMID: 35574625 PMCID: PMC9109056 DOI: 10.15252/msb.202110712] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 12/11/2022] Open
Abstract
Genomic variation impacts on cellular networks by affecting the abundance (e.g., protein levels) and the functional states (e.g., protein phosphorylation) of their components. Previous work has focused on the former, while in this context, the functional states of proteins have largely remained neglected. Here, we generated high-quality transcriptome, proteome, and phosphoproteome data for a panel of 112 genomically well-defined yeast strains. Genetic effects on transcripts were generally transmitted to the protein layer, but specific gene groups, such as ribosomal proteins, showed diverging effects on protein levels compared with RNA levels. Phosphorylation states proved crucial to unravel genetic effects on signaling networks. Correspondingly, genetic variants that cause phosphorylation changes were mostly different from those causing abundance changes in the respective proteins. Underscoring their relevance for cell physiology, phosphorylation traits were more strongly correlated with cell physiological traits such as chemical compound resistance or cell morphology, compared with transcript or protein abundance. This study demonstrates how molecular networks mediate the effects of genomic variants to cellular traits and highlights the particular importance of protein phosphorylation.
Collapse
Affiliation(s)
- Jan Grossbach
- Excellence Cluster on Cellular Stress Responses in Aging Associated DiseasesUniversity of CologneCologneGermany
| | - Ludovic Gillet
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Mathieu Clément‐Ziza
- Center for Molecular Medicine Cologne (CMMC)Medical Faculty, University of CologneCologneGermany
- Lesaffre InternationalMarcq‐en‐BarœulFrance
| | - Corinna L Schmalohr
- Excellence Cluster on Cellular Stress Responses in Aging Associated DiseasesUniversity of CologneCologneGermany
| | - Olga T Schubert
- Department of Human GeneticsUniversity of California, Los AngelesLos AngelesCAUSA
| | | | | | | | - Isabell Bludau
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Matthias Weith
- Excellence Cluster on Cellular Stress Responses in Aging Associated DiseasesUniversity of CologneCologneGermany
| | - Peter Tessarz
- Excellence Cluster on Cellular Stress Responses in Aging Associated DiseasesUniversity of CologneCologneGermany
- Max Planck Institute for Biology of AgeingCologneGermany
| | - Martin Graef
- Excellence Cluster on Cellular Stress Responses in Aging Associated DiseasesUniversity of CologneCologneGermany
- Max Planck Institute for Biology of AgeingCologneGermany
| | - Ruedi Aebersold
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
- Faculty of ScienceUniversity of ZurichZurichSwitzerland
| | - Andreas Beyer
- Excellence Cluster on Cellular Stress Responses in Aging Associated DiseasesUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)Medical Faculty, University of CologneCologneGermany
- Institute for GeneticsFaculty of Mathematics and Natural SciencesUniversity of CologneCologneGermany
| |
Collapse
|
34
|
Reymond S, Vujić T, Schvartz D, Sanchez JC. Morphine-induced modulation of Nrf2-antioxidant response element signaling pathway in primary human brain microvascular endothelial cells. Sci Rep 2022; 12:4588. [PMID: 35301408 PMCID: PMC8931063 DOI: 10.1038/s41598-022-08712-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 03/07/2022] [Indexed: 11/09/2022] Open
Abstract
Morphine is one of the most potent opioid analgesic used for pain treatment. Morphine action in the central nervous system requires crossing the blood-brain barrier. Due to the controversial relationship between morphine and oxidative stress, the potential pro- or antioxidant effects of morphine in the blood-brain barrier is important to be understood, as oxidative stress could cause its disruption and predispose to neurodegenerative diseases. However, investigation is scarce in human brain endothelial cells. Therefore, the present study evaluated the impact of morphine exposure at three different concentrations (1, 10 and 100 µM) for 24 h and 48 h on primary human brain microvascular endothelial cells. A quantitative data-independent acquisition mass spectrometry strategy was used to analyze proteome modulations. Almost 3000 proteins were quantified of which 217 were reported to be significantly regulated in at least one condition versus untreated control. Pathway enrichment analysis unveiled dysregulation of the Nrf2 pathway involved in oxidative stress response. Seahorse assay underlined mitochondria dysfunctions, which were supported by significant expression modulations of relevant mitochondrial proteins. In conclusion, our study revealed the dysregulation of the Nrf2 pathway and mitochondria dysfunctions after morphine exposure, highlighting a potential redox imbalance in human brain endothelial cells.
Collapse
Affiliation(s)
- Sandrine Reymond
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Tatjana Vujić
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Domitille Schvartz
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Swiss Center for Applied Human Toxicology, Geneva, Switzerland.
| |
Collapse
|
35
|
Baxi EG, Thompson T, Li J, Kaye JA, Lim RG, Wu J, Ramamoorthy D, Lima L, Vaibhav V, Matlock A, Frank A, Coyne AN, Landin B, Ornelas L, Mosmiller E, Thrower S, Farr SM, Panther L, Gomez E, Galvez E, Perez D, Meepe I, Lei S, Mandefro B, Trost H, Pinedo L, Banuelos MG, Liu C, Moran R, Garcia V, Workman M, Ho R, Wyman S, Roggenbuck J, Harms MB, Stocksdale J, Miramontes R, Wang K, Venkatraman V, Holewenski R, Sundararaman N, Pandey R, Manalo DM, Donde A, Huynh N, Adam M, Wassie BT, Vertudes E, Amirani N, Raja K, Thomas R, Hayes L, Lenail A, Cerezo A, Luppino S, Farrar A, Pothier L, Prina C, Morgan T, Jamil A, Heintzman S, Jockel-Balsarotti J, Karanja E, Markway J, McCallum M, Joslin B, Alibazoglu D, Kolb S, Ajroud-Driss S, Baloh R, Heitzman D, Miller T, Glass JD, Patel-Murray NL, Yu H, Sinani E, Vigneswaran P, Sherman AV, Ahmad O, Roy P, Beavers JC, Zeiler S, Krakauer JW, Agurto C, Cecchi G, Bellard M, Raghav Y, Sachs K, Ehrenberger T, Bruce E, Cudkowicz ME, Maragakis N, Norel R, Van Eyk JE, Finkbeiner S, Berry J, Sareen D, Thompson LM, Fraenkel E, Svendsen CN, et alBaxi EG, Thompson T, Li J, Kaye JA, Lim RG, Wu J, Ramamoorthy D, Lima L, Vaibhav V, Matlock A, Frank A, Coyne AN, Landin B, Ornelas L, Mosmiller E, Thrower S, Farr SM, Panther L, Gomez E, Galvez E, Perez D, Meepe I, Lei S, Mandefro B, Trost H, Pinedo L, Banuelos MG, Liu C, Moran R, Garcia V, Workman M, Ho R, Wyman S, Roggenbuck J, Harms MB, Stocksdale J, Miramontes R, Wang K, Venkatraman V, Holewenski R, Sundararaman N, Pandey R, Manalo DM, Donde A, Huynh N, Adam M, Wassie BT, Vertudes E, Amirani N, Raja K, Thomas R, Hayes L, Lenail A, Cerezo A, Luppino S, Farrar A, Pothier L, Prina C, Morgan T, Jamil A, Heintzman S, Jockel-Balsarotti J, Karanja E, Markway J, McCallum M, Joslin B, Alibazoglu D, Kolb S, Ajroud-Driss S, Baloh R, Heitzman D, Miller T, Glass JD, Patel-Murray NL, Yu H, Sinani E, Vigneswaran P, Sherman AV, Ahmad O, Roy P, Beavers JC, Zeiler S, Krakauer JW, Agurto C, Cecchi G, Bellard M, Raghav Y, Sachs K, Ehrenberger T, Bruce E, Cudkowicz ME, Maragakis N, Norel R, Van Eyk JE, Finkbeiner S, Berry J, Sareen D, Thompson LM, Fraenkel E, Svendsen CN, Rothstein JD. Answer ALS, a large-scale resource for sporadic and familial ALS combining clinical and multi-omics data from induced pluripotent cell lines. Nat Neurosci 2022; 25:226-237. [PMID: 35115730 PMCID: PMC8825283 DOI: 10.1038/s41593-021-01006-0] [Show More Authors] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022]
Abstract
Answer ALS is a biological and clinical resource of patient-derived, induced pluripotent stem (iPS) cell lines, multi-omic data derived from iPS neurons and longitudinal clinical and smartphone data from over 1,000 patients with ALS. This resource provides population-level biological and clinical data that may be employed to identify clinical-molecular-biochemical subtypes of amyotrophic lateral sclerosis (ALS). A unique smartphone-based system was employed to collect deep clinical data, including fine motor activity, speech, breathing and linguistics/cognition. The iPS spinal neurons were blood derived from each patient and these cells underwent multi-omic analytics including whole-genome sequencing, RNA transcriptomics, ATAC-sequencing and proteomics. The intent of these data is for the generation of integrated clinical and biological signatures using bioinformatics, statistics and computational biology to establish patterns that may lead to a better understanding of the underlying mechanisms of disease, including subgroup identification. A web portal for open-source sharing of all data was developed for widespread community-based data analytics.
Collapse
Affiliation(s)
- Emily G Baxi
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Jonathan Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Julia A Kaye
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Ryan G Lim
- UCI MIND, University of California, Irvine, CA, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Divya Ramamoorthy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Leandro Lima
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Vineet Vaibhav
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrea Matlock
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aaron Frank
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alyssa N Coyne
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barry Landin
- Computational Biology Center, IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Loren Ornelas
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Elizabeth Mosmiller
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sara Thrower
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Lindsey Panther
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emilda Gomez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Erick Galvez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel Perez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Imara Meepe
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Lei
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Berhan Mandefro
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hannah Trost
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Louis Pinedo
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maria G Banuelos
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Chunyan Liu
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ruby Moran
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Veronica Garcia
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael Workman
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Richie Ho
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stacia Wyman
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Matthew B Harms
- Department of Neurology and Genetics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jennifer Stocksdale
- Department of Psychiatry and Human Behavior and Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
| | | | - Keona Wang
- Department of Psychiatry and Human Behavior and Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ronald Holewenski
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rakhi Pandey
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Danica-Mae Manalo
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aneesh Donde
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nhan Huynh
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Miriam Adam
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brook T Wassie
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Edward Vertudes
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Naufa Amirani
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Krishna Raja
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Reuben Thomas
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Lindsey Hayes
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alex Lenail
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aianna Cerezo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Luppino
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alanna Farrar
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lindsay Pothier
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carolyn Prina
- Department of Neurology and Genetics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Arish Jamil
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Sarah Heintzman
- Department of Neurology and Genetics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | - Jesse Markway
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Molly McCallum
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Ben Joslin
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Deniz Alibazoglu
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Stephen Kolb
- Department of Neurology and Genetics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Robert Baloh
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Tim Miller
- Department of Neurology, Washington University, St. Louis, MO, USA
| | | | | | - Hong Yu
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ervin Sinani
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Prasha Vigneswaran
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander V Sherman
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Omar Ahmad
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Promit Roy
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jay C Beavers
- Microsoft Research, Microsoft Corporation, Redmond, WA, USA
| | - Steven Zeiler
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John W Krakauer
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carla Agurto
- Computational Biology Center, IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Guillermo Cecchi
- Computational Biology Center, IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Mary Bellard
- Microsoft University Relations, Microsoft Corporation, Redmond, WA, USA
| | - Yogindra Raghav
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Karen Sachs
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tobias Ehrenberger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Elizabeth Bruce
- Microsoft University Relations, Microsoft Corporation, Redmond, WA, USA
| | - Merit E Cudkowicz
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholas Maragakis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raquel Norel
- Computational Biology Center, IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - James Berry
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dhruv Sareen
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Leslie M Thompson
- UCI MIND, University of California, Irvine, CA, USA
- Department of Biological Chemistry, University of California, Irvine, CA, USA
- Department of Psychiatry and Human Behavior and Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Clive N Svendsen
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jeffrey D Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
36
|
Ramani K, Robinson AE, Berlind J, Fan W, Abeynayake A, Binek A, Barbier-Torres L, Noureddin M, Nissen NN, Yildirim Z, Erbay E, Mato JM, Van Eyk JE, Lu SC. S-adenosylmethionine inhibits la ribonucleoprotein domain family member 1 in murine liver and human liver cancer cells. Hepatology 2022; 75:280-296. [PMID: 34449924 PMCID: PMC8766892 DOI: 10.1002/hep.32130] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/22/2021] [Accepted: 08/09/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIMS Methionine adenosyltransferase 1A (MAT1A) is responsible for S-adenosylmethionine (SAMe) biosynthesis in the liver. Mice lacking Mat1a have hepatic SAMe depletion and develop NASH and HCC spontaneously. Several kinases are activated in Mat1a knockout (KO) mice livers. However, characterizing the phospho-proteome and determining whether they contribute to liver pathology remain open for study. Our study aimed to provide this knowledge. APPROACH AND RESULTS We performed phospho-proteomics in Mat1a KO mice livers with and without SAMe treatment to identify SAMe-dependent changes that may contribute to liver pathology. Our studies used Mat1a KO mice at different ages treated with and without SAMe, cell lines, in vitro translation and kinase assays, and human liver specimens. We found that the most striking change was hyperphosphorylation and increased content of La-related protein 1 (LARP1), which, in the unphosphorylated form, negatively regulates translation of 5'-terminal oligopyrimidine (TOP)-containing mRNAs. Consistently, multiple TOP proteins are induced in KO livers. Translation of TOP mRNAs ribosomal protein S3 and ribosomal protein L18 was enhanced by LARP1 overexpression in liver cancer cells. We identified LARP1-T449 as a SAMe-sensitive phospho-site of cyclin-dependent kinase 2 (CDK2). Knocking down CDK2 lowered LARP1 phosphorylation and prevented LARP1-overexpression-mediated increase in translation. LARP1-T449 phosphorylation induced global translation, cell growth, migration, invasion, and expression of oncogenic TOP-ribosomal proteins in HCC cells. LARP1 expression is increased in human NASH and HCC. CONCLUSIONS Our results reveal a SAMe-sensitive mechanism of LARP1 phosphorylation that may be involved in the progression of NASH to HCC.
Collapse
Affiliation(s)
- Komal Ramani
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Aaron E. Robinson
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
- Smidt Heart Institute and Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Joshua Berlind
- Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033
| | - Wei Fan
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Aushinie Abeynayake
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Aleksandra Binek
- Smidt Heart Institute and Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Mazen Noureddin
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Nicholas N. Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Zehra Yildirim
- Department of Cardiology, Department of Biomedical Sciences and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ebru Erbay
- Department of Cardiology, Department of Biomedical Sciences and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - José M. Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology, Derio, Bizkaia 48160, Spain
| | - Jennifer E. Van Eyk
- Smidt Heart Institute and Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
37
|
Comparative assessment and novel strategy on methods for imputing proteomics data. Sci Rep 2022; 12:1067. [PMID: 35058491 PMCID: PMC8776850 DOI: 10.1038/s41598-022-04938-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/04/2022] [Indexed: 11/09/2022] Open
Abstract
Missing values are a major issue in quantitative proteomics analysis. While many methods have been developed for imputing missing values in high-throughput proteomics data, a comparative assessment of imputation accuracy remains inconclusive, mainly because mechanisms contributing to true missing values are complex and existing evaluation methodologies are imperfect. Moreover, few studies have provided an outlook of future methodological development. We first re-evaluate the performance of eight representative methods targeting three typical missing mechanisms. These methods are compared on both simulated and masked missing values embedded within real proteomics datasets, and performance is evaluated using three quantitative measures. We then introduce fused regularization matrix factorization, a low-rank global matrix factorization framework, capable of integrating local similarity derived from additional data types. We also explore a biologically-inspired latent variable modeling strategy—convex analysis of mixtures—for missing value imputation and present preliminary experimental results. While some winners emerged from our comparative assessment, the evaluation is intrinsically imperfect because performance is evaluated indirectly on artificial missing or masked values not authentic missing values. Nevertheless, we show that our fused regularization matrix factorization provides a novel incorporation of external and local information, and the exploratory implementation of convex analysis of mixtures presents a biologically plausible new approach.
Collapse
|
38
|
Ho AS, Robinson A, Shon W, Laury A, Raedschelders K, Venkatraman V, Holewinski R, Zhang Y, Shiao SL, Chen MM, Mallen-St Clair J, Lin DC, Zumsteg ZS, Van Eyk JE. Comparative Proteomic Analysis of HPV(+) Oropharyngeal Squamous Cell Carcinoma Recurrence. J Proteome Res 2021; 21:200-208. [PMID: 34846153 DOI: 10.1021/acs.jproteome.1c00757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Deintensification therapy for human papillomavirus-related oropharyngeal squamous cell carcinoma (HPV(+) OPSCC) is under active investigation. An adaptive treatment approach based on molecular stratification could identify high-risk patients predisposed to recurrence and better select for appropriate treatment regimens. Collectively, 40 HPV(+) OPSCC FFPE samples (20 disease-free, 20 recurrent) were surveyed using mass spectrometry-based proteomic analysis via data-independent acquisition to obtain fold change and false discovery differences. Ten-year overall survival was 100.0 and 27.7% for HPV(+) disease-free and recurrent cohorts, respectively. Of 1414 quantified proteins, 77 demonstrated significant differential expression. Top enriched functional pathways included those involved in programmed cell death (73 proteins, p = 7.43 × 10-30), apoptosis (73 proteins, p = 5.56 × 10-9), β-catenin independent WNT signaling (47 proteins, p = 1.45 × 10-15), and Rho GTPase signaling (69 proteins, p = 1.09 × 10-5). PFN1 (p = 1.0 × 10-3), RAD23B (p = 2.9 × 10-4), LDHB (p = 1.0 × 10-3), and HINT1 (p = 3.8 × 10-3) pathways were significantly downregulated in the recurrent cohort. On functional validation via immunohistochemistry (IHC) staining, 46.9% (PFN1), 71.9% (RAD23B), 59.4% (LDHB), and 84.4% (HINT1) of cases were corroborated with mass spectrometry findings. Development of a multilateral molecular signature incorporating these targets may characterize high-risk disease, predict treatment response, and augment current management paradigms in head and neck cancer.
Collapse
|
39
|
Li J, Lim RG, Kaye JA, Dardov V, Coyne AN, Wu J, Milani P, Cheng A, Thompson TG, Ornelas L, Frank A, Adam M, Banuelos MG, Casale M, Cox V, Escalante-Chong R, Daigle JG, Gomez E, Hayes L, Holewenski R, Lei S, Lenail A, Lima L, Mandefro B, Matlock A, Panther L, Patel-Murray NL, Pham J, Ramamoorthy D, Sachs K, Shelley B, Stocksdale J, Trost H, Wilhelm M, Venkatraman V, Wassie BT, Wyman S, Yang S, Van Eyk JE, Lloyd TE, Finkbeiner S, Fraenkel E, Rothstein JD, Sareen D, Svendsen CN, Thompson LM. An integrated multi-omic analysis of iPSC-derived motor neurons from C9ORF72 ALS patients. iScience 2021; 24:103221. [PMID: 34746695 PMCID: PMC8554488 DOI: 10.1016/j.isci.2021.103221] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/29/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are challenging for systems biology because of the lack of reliable animal models or patient samples at early disease stages. Induced pluripotent stem cells (iPSCs) could address these challenges. We investigated DNA, RNA, epigenetics, and proteins in iPSC-derived motor neurons from patients with ALS carrying hexanucleotide expansions in C9ORF72. Using integrative computational methods combining all omics datasets, we identified novel and known dysregulated pathways. We used a C9ORF72 Drosophila model to distinguish pathways contributing to disease phenotypes from compensatory ones and confirmed alterations in some pathways in postmortem spinal cord tissue of patients with ALS. A different differentiation protocol was used to derive a separate set of C9ORF72 and control motor neurons. Many individual -omics differed by protocol, but some core dysregulated pathways were consistent. This strategy of analyzing patient-specific neurons provides disease-related outcomes with small numbers of heterogeneous lines and reduces variation from single-omics to elucidate network-based signatures.
Collapse
Affiliation(s)
- Jonathan Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ryan G. Lim
- UCI MIND, University of California, Irvine, CA 92697, USA
| | - Julia A. Kaye
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Victoria Dardov
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alyssa N. Coyne
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA
| | - Pamela Milani
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andrew Cheng
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | | | - Loren Ornelas
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Aaron Frank
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Miriam Adam
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Maria G. Banuelos
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Malcolm Casale
- UCI MIND, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Veerle Cox
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Renan Escalante-Chong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - J. Gavin Daigle
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Emilda Gomez
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Lindsey Hayes
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Ronald Holewenski
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Lei
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Alex Lenail
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leandro Lima
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Berhan Mandefro
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Andrea Matlock
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lindsay Panther
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | | | - Jacqueline Pham
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Divya Ramamoorthy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Karen Sachs
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brandon Shelley
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Jennifer Stocksdale
- UCI MIND, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Hannah Trost
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Mark Wilhelm
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Brook T. Wassie
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stacia Wyman
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697, USA
| | - Stephanie Yang
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | | | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thomas E. Lloyd
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, University of California, San Francisco, San Francisco, CA 94158, USA
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeffrey D. Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Dhruv Sareen
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Clive N. Svendsen
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Leslie M. Thompson
- UCI MIND, University of California, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697, USA
| |
Collapse
|
40
|
Porritt RA, Binek A, Paschold L, Rivas MN, McArdle A, Yonker LM, Alter G, Chandnani HK, Lopez M, Fasano A, Van Eyk JE, Binder M, Arditi M. The autoimmune signature of hyperinflammatory multisystem inflammatory syndrome in children. J Clin Invest 2021; 131:e151520. [PMID: 34437303 PMCID: PMC8516454 DOI: 10.1172/jci151520] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) manifests as a severe and uncontrolled inflammatory response with multiorgan involvement, occurring weeks after SARS-CoV-2 infection. Here, we utilized proteomics, RNA sequencing, autoantibody arrays, and B cell receptor (BCR) repertoire analysis to characterize MIS-C immunopathogenesis and identify factors contributing to severe manifestations and intensive care unit admission. Inflammation markers, humoral immune responses, neutrophil activation, and complement and coagulation pathways were highly enriched in MIS-C patient serum, with a more hyperinflammatory profile in severe than in mild MIS-C cases. We identified a strong autoimmune signature in MIS-C, with autoantibodies targeted to both ubiquitously expressed and tissue-specific antigens, suggesting autoantigen release and excessive antigenic drive may result from systemic tissue damage. We further identified a cluster of patients with enhanced neutrophil responses as well as high anti-Spike IgG and autoantibody titers. BCR sequencing of these patients identified a strong imprint of antigenic drive with substantial BCR sequence connectivity and usage of autoimmunity-associated immunoglobulin heavy chain variable region (IGHV) genes. This cluster was linked to a TRBV11-2 expanded T cell receptor (TCR) repertoire, consistent with previous studies indicating a superantigen-driven pathogenic process. Overall, we identify a combination of pathogenic pathways that culminate in MIS-C and may inform treatment.
Collapse
Affiliation(s)
- Rebecca A. Porritt
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences and
| | - Aleksandra Binek
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Lisa Paschold
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Magali Noval Rivas
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences and
| | - Angela McArdle
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Lael M. Yonker
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center and Department of Pediatrics, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Galit Alter
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center and Department of Pediatrics, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MIT, MGH and Harvard, Cambridge, Massachusetts, USA
| | | | - Merrick Lopez
- Department of Pediatrics, Loma Linda University Hospital, California, USA
| | - Alessio Fasano
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center and Department of Pediatrics, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Moshe Arditi
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences and
- Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
41
|
Tatjana V, Domitille S, Jean-Charles S. Paraquat-induced cholesterol biosynthesis proteins dysregulation in human brain microvascular endothelial cells. Sci Rep 2021; 11:18137. [PMID: 34518572 PMCID: PMC8438088 DOI: 10.1038/s41598-021-97175-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/23/2021] [Indexed: 12/25/2022] Open
Abstract
Despite Paraquat (PQ) being banned in several countries, it is still one of the most commonly used herbicides in agriculture. This compound is known to induce damaging effects on human and animal brain cells by generating Reactive Oxygen Species (ROS). However, there is few evidence of PQ effect on Human Brain Microvascular Endothelial Cells (HBMECs), one of the major component of the Blood–Brain Barrier (BBB). The present study aimed at unraveling biological mechanisms associated to the exposure of 1, 10 and 100 µM of PQ for 24 h on HBMECs. High-throughput mass spectrometry-based proteomics using data-independent acquisition (DIA) was applied. Biological pathway enrichment and cellular assays such as mitochondrial respiration and cholesterol level were performed to verify proteomics results. A total of 3753 proteins were quantified out of which 419 were significantly modulated by paraquat exposure. Biological pathway enrichment revealed the ubiquinone metabolism, a pathway directly linked to mitochondrial complex I proteins, confirming the well-known mechanism of PQ inducing oxidative stress. Additionally, this study also described the cholesterol biosynthesis modulation on HBMECs not yet described. In conclusion, our data indicate the toxic effect of PQ on HBMECs by downregulating proteins involved in mitochondrial complex I and cholesterol pathways.
Collapse
Affiliation(s)
- Vujić Tatjana
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Schvartz Domitille
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Sanchez Jean-Charles
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Swiss Center for Applied Human Toxicology, Geneva, Switzerland.
| |
Collapse
|
42
|
Bahmani M, O’Lone CE, Juhász A, Nye-Wood M, Dunn H, Edwards IB, Colgrave ML. Application of Mass Spectrometry-Based Proteomics to Barley Research. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:8591-8609. [PMID: 34319719 PMCID: PMC8389776 DOI: 10.1021/acs.jafc.1c01871] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Barley (Hordeum vulgare) is the fourth most cultivated crop in the world in terms of production volume, and it is also the most important raw material of the malting and brewing industries. Barley belongs to the grass (Poaceae) family and plays an important role in food security and food safety for both humans and livestock. With the global population set to reach 9.7 billion by 2050, but with less available and/or suitable land for agriculture, the use of biotechnology tools in breeding programs are of considerable importance in the quest to meet the growing food gap. Proteomics as a member of the "omics" technologies has become popular for the investigation of proteins in cereal crops and particularly barley and its related products such as malt and beer. This technology has been applied to study how proteins in barley respond to adverse environmental conditions including abiotic and/or biotic stresses, how they are impacted during food processing including malting and brewing, and the presence of proteins implicated in celiac disease. Moreover, proteomics can be used in the future to inform breeding programs that aim to enhance the nutritional value and broaden the application of this crop in new food and beverage products. Mass spectrometry analysis is a valuable tool that, along with genomics and transcriptomics, can inform plant breeding strategies that aim to produce superior barley varieties. In this review, recent studies employing both qualitative and quantitative mass spectrometry approaches are explored with a focus on their application in cultivation, manufacturing, processing, quality, and the safety of barley and its related products.
Collapse
Affiliation(s)
- Mahya Bahmani
- Australian
Research Council Centre of Excellence for Innovations in Peptide and
Protein Science, Edith Cowan University, School of Science, 270 Joondalup
Drive, Joondalup, Western
Australia 6027, Australia
| | - Clare E. O’Lone
- Australian
Research Council Centre of Excellence for Innovations in Peptide and
Protein Science, Edith Cowan University, School of Science, 270 Joondalup
Drive, Joondalup, Western
Australia 6027, Australia
| | - Angéla Juhász
- Australian
Research Council Centre of Excellence for Innovations in Peptide and
Protein Science, Edith Cowan University, School of Science, 270 Joondalup
Drive, Joondalup, Western
Australia 6027, Australia
| | - Mitchell Nye-Wood
- Australian
Research Council Centre of Excellence for Innovations in Peptide and
Protein Science, Edith Cowan University, School of Science, 270 Joondalup
Drive, Joondalup, Western
Australia 6027, Australia
| | - Hugh Dunn
- Australian
Research Council Centre of Excellence for Innovations in Peptide and
Protein Science, Edith Cowan University, School of Science, 270 Joondalup
Drive, Joondalup, Western
Australia 6027, Australia
| | - Ian B. Edwards
- Edstar
Genetics Pty Ltd, SABC - Loneragan Building, Murdoch University, 90 South Street, Murdoch, Western Australia 6150, Australia
| | - Michelle L. Colgrave
- Australian
Research Council Centre of Excellence for Innovations in Peptide and
Protein Science, Edith Cowan University, School of Science, 270 Joondalup
Drive, Joondalup, Western
Australia 6027, Australia
- CSIRO
Agriculture and Food, 306 Carmody Road, St. Lucia, Queensland 4067, Australia
- Phone: +61-7-3214-2697. . Fax: +61-7-3214-2900
| |
Collapse
|
43
|
Yan J, Zhai H, Zhu L, Sa S, Ding X. obaDIA: one-step biological analysis pipeline for data-independent acquisition and other quantitative proteomics data. Bioinformatics 2021; 37:2066-2067. [PMID: 33270834 DOI: 10.1093/bioinformatics/btaa893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/26/2020] [Accepted: 10/02/2020] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Data mining and data quality evaluation are indispensable constituents of quantitative proteomics, but few integrated tools available. RESULTS We introduced obaDIA, a one-step pipeline to generate visualizable and comprehensive results for quantitative proteomics data. obaDIA supports fragment-level, peptide-level and protein-level abundance matrices from DIA technique, as well as protein-level abundance matrices from other quantitative proteomic techniques. The result contains abundance matrix statistics, differential expression analysis, protein functional annotation and enrichment analysis. Additionally, enrichment strategies which use total proteins or expressed proteins as background are optional, and HTML based interactive visualization for differentially expressed proteins in the KEGG pathway is offered, which helps biological significance mining. In short, obaDIA is an automatic tool for bioinformatics analysis for quantitative proteomics. AVAILABILITY AND IMPLEMENTATION obaDIA is freely available from https://github.com/yjthu/obaDIA.git. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jun Yan
- Department of Crop Genomics and Bioinformatics, College of Agronomy and Biotechnology, China Agricultural University, Beijing 100094, China.,Department of Bioinformatics, Beijing Mingde Zhengkang Medical Research Co., Ltd., Beijing 102206, China
| | - Hongning Zhai
- Department of Bioinformatics, Beijing Mingde Zhengkang Medical Research Co., Ltd., Beijing 102206, China
| | - Ling Zhu
- Department of College English, Inner Mongolia Medical University, Huhehot, Inner Mongolia 010110, China
| | - Sasha Sa
- Department of Bioinformatics, Beijing Mingde Zhengkang Medical Research Co., Ltd., Beijing 102206, China
| | - Xiaojun Ding
- Department of Bioinformatics, Beijing Mingde Zhengkang Medical Research Co., Ltd., Beijing 102206, China
| |
Collapse
|
44
|
Pei W, Chen J, Wu W, Wei W, Yu Y, Feng Y. Comparison of the rabbit and human corneal endothelial proteomes regarding proliferative capacity. Exp Eye Res 2021; 209:108629. [PMID: 34029595 DOI: 10.1016/j.exer.2021.108629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/29/2021] [Accepted: 05/16/2021] [Indexed: 02/05/2023]
Abstract
The shortage of human donor corneas has raised important concerns about engineering of corneal endothelial cells (CECs) for clinical use. However, due to the limited proliferative capacity of human CECs, driving them into proliferation and regeneration may be difficult. Unlike human CECs, rabbit CECs have a marked proliferative capacity. To clarify the potential reason for this difference, we analysed the proteomes of four human corneal endothelium samples and four rabbit corneal endothelium samples with quantitative label-free proteomics and downstream analysis. We discovered that vitamin and selenocompound metabolism and some signaling pathways such as NF-kappa B signaling pathway differed between the samples. Moreover, TGFβ, PITX2 and keratocan were distinctively expressed in rabbit samples, which might be associated with active proliferation in rabbit CECs. This study illustrates the proteomic differences between human and rabbit CECs and might promote CEC engineering strategies.
Collapse
Affiliation(s)
- Wendi Pei
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China
| | - Jun Chen
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenyu Wu
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Wei Wei
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Yang Yu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Yun Feng
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
45
|
Čuklina J, Lee CH, Williams EG, Sajic T, Collins BC, Rodríguez Martínez M, Sharma VS, Wendt F, Goetze S, Keele GR, Wollscheid B, Aebersold R, Pedrioli PGA. Diagnostics and correction of batch effects in large-scale proteomic studies: a tutorial. Mol Syst Biol 2021; 17:e10240. [PMID: 34432947 PMCID: PMC8447595 DOI: 10.15252/msb.202110240] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 07/16/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022] Open
Abstract
Advancements in mass spectrometry-based proteomics have enabled experiments encompassing hundreds of samples. While these large sample sets deliver much-needed statistical power, handling them introduces technical variability known as batch effects. Here, we present a step-by-step protocol for the assessment, normalization, and batch correction of proteomic data. We review established methodologies from related fields and describe solutions specific to proteomic challenges, such as ion intensity drift and missing values in quantitative feature matrices. Finally, we compile a set of techniques that enable control of batch effect adjustment quality. We provide an R package, "proBatch", containing functions required for each step of the protocol. We demonstrate the utility of this methodology on five proteomic datasets each encompassing hundreds of samples and consisting of multiple experimental designs. In conclusion, we provide guidelines and tools to make the extraction of true biological signal from large proteomic studies more robust and transparent, ultimately facilitating reliable and reproducible research in clinical proteomics and systems biology.
Collapse
Affiliation(s)
- Jelena Čuklina
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
- PhD Program in Systems BiologyUniversity of Zurich and ETH ZurichZurichSwitzerland
- IBM Research EuropeRüschlikonSwitzerland
| | - Chloe H Lee
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Evan G Williams
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgLuxembourgLuxembourg
| | - Tatjana Sajic
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Ben C Collins
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
- Queen’s University BelfastBelfastUK
| | | | - Varun S Sharma
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Fabian Wendt
- Department of Health Sciences and TechnologyInstitute of Translational MedicineETH ZurichZurichSwitzerland
| | - Sandra Goetze
- Department of Health Sciences and TechnologyInstitute of Translational MedicineETH ZurichZurichSwitzerland
- ETH ZürichPHRT‐CPACZürichSwitzerland
- SIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | | | - Bernd Wollscheid
- Department of Health Sciences and TechnologyInstitute of Translational MedicineETH ZurichZurichSwitzerland
- ETH ZürichPHRT‐CPACZürichSwitzerland
- SIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Ruedi Aebersold
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
- Faculty of ScienceUniversity of ZurichZurichSwitzerland
| | - Patrick G A Pedrioli
- Department of BiologyInstitute of Molecular Systems BiologyETH ZurichZurichSwitzerland
- Department of Health Sciences and TechnologyInstitute of Translational MedicineETH ZurichZurichSwitzerland
- ETH ZürichPHRT‐CPACZürichSwitzerland
- SIB Swiss Institute of BioinformaticsLausanneSwitzerland
| |
Collapse
|
46
|
Kim J, Yeon A, Parker SJ, Shahid M, Thiombane A, Cho E, You S, Emam H, Kim DG, Kim M. Alendronate-induced Perturbation of the Bone Proteome and Microenvironmental Pathophysiology. Int J Med Sci 2021; 18:3261-3270. [PMID: 34400895 PMCID: PMC8364444 DOI: 10.7150/ijms.61552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/11/2021] [Indexed: 11/05/2022] Open
Abstract
Objectives: Bisphosphonates (BPs) are powerful inhibitors of osteoclastogenesis and are used to prevent osteoporotic bone loss and reduce the risk of osteoporotic fracture in patients suffering from postmenopausal osteoporosis. Patients with breast cancer or gynecological malignancies being treated with BPs or those receiving bone-targeted therapy for metastatic prostate cancer are at increased risk of bisphosphonate-related osteonecrosis of the jaw (BRONJ). Although BPs markedly ameliorate osteoporosis, their adverse effects largely limit the clinical application of these drugs. This study focused on providing a deeper understanding of one of the most popular BPs, the alendronate (ALN)-induced perturbation of the bone proteome and microenvironmental pathophysiology. Methods: To understand the molecular mechanisms underlying ALN-induced side-effects, an unbiased and global proteomics approach combined with big data bioinformatics was applied. This was followed by biochemical and functional analyses to determine the clinicopathological mechanisms affected by ALN. Results: The findings from this proteomics study suggest that the RIPK3/Wnt/GSK3/β-catenin signaling pathway is significantly perturbed upon ALN treatment, resulting in abnormal angiogenesis, inflammation, anabolism, remodeling, and mineralization in bone cells in an in vitro cell culture system. Conclusion: Our investigation into potential key signaling mechanisms in response to ALN provides a rational basis for suppressing BP-induced adverse effect and presents various therapeutic strategies.
Collapse
Affiliation(s)
- Jayoung Kim
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, University of California Los Angeles, CA, USA
| | - Austin Yeon
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sarah J. Parker
- Smidt Heart Institute, Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Muhammad Shahid
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aissatou Thiombane
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eunho Cho
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hany Emam
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Do-Gyoon Kim
- Division of Oral Surgery, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Minjung Kim
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| |
Collapse
|
47
|
Kammers K, Taub MA, Mathias RA, Yanek LR, Kanchan K, Venkatraman V, Sundararaman N, Martin J, Liu S, Hoyle D, Raedschelders K, Holewinski R, Parker S, Dardov V, Faraday N, Becker DM, Cheng L, Wang ZZ, Leek JT, Van Eyk JE, Becker LC. Gene and protein expression in human megakaryocytes derived from induced pluripotent stem cells. J Thromb Haemost 2021; 19:1783-1799. [PMID: 33829634 DOI: 10.1111/jth.15334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/25/2021] [Accepted: 02/19/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND There is interest in deriving megakaryocytes (MKs) from pluripotent stem cells (iPSC) for biological studies. We previously found that genomic structural integrity and genotype concordance is maintained in iPSC-derived MKs. OBJECTIVE To establish a comprehensive dataset of genes and proteins expressed in iPSC-derived MKs. METHODS iPSCs were reprogrammed from peripheral blood mononuclear cells (MNCs) and MKs were derived from the iPSCs in 194 healthy European American and African American subjects. mRNA was isolated and gene expression measured by RNA sequencing. Protein expression was measured in 62 of the subjects using mass spectrometry. RESULTS AND CONCLUSIONS MKs expressed genes and proteins known to be important in MK and platelet function and demonstrated good agreement with previous studies in human MKs derived from CD34+ progenitor cells. The percent of cells expressing the MK markers CD41 and CD42a was consistent in biological replicates, but variable across subjects, suggesting that unidentified subject-specific factors determine differentiation of MKs from iPSCs. Gene and protein sets important in platelet function were associated with increasing expression of CD41/42a, while those related to more basic cellular functions were associated with lower CD41/42a expression. There was differential gene expression by the sex and race (but not age) of the subject. Numerous genes and proteins were highly expressed in MKs but not known to play a role in MK or platelet function; these represent excellent candidates for future study of hematopoiesis, platelet formation, and/or platelet function.
Collapse
Affiliation(s)
- Kai Kammers
- Division of Biostatistics and Bioinformatics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Rasika A Mathias
- The GeneSTAR Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lisa R Yanek
- The GeneSTAR Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kanika Kanchan
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, Barbra Streisand Woman's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, Barbra Streisand Woman's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Joshua Martin
- The GeneSTAR Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Senquan Liu
- Division of Hematology and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dixie Hoyle
- Division of Hematology and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Koen Raedschelders
- Advanced Clinical Biosystems Research Institute, Barbra Streisand Woman's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ronald Holewinski
- Advanced Clinical Biosystems Research Institute, Barbra Streisand Woman's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sarah Parker
- Advanced Clinical Biosystems Research Institute, Barbra Streisand Woman's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Victoria Dardov
- Advanced Clinical Biosystems Research Institute, Barbra Streisand Woman's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nauder Faraday
- The GeneSTAR Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Diane M Becker
- The GeneSTAR Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linzhao Cheng
- Division of Hematology and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zack Z Wang
- Division of Hematology and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey T Leek
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Barbra Streisand Woman's Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Lewis C Becker
- The GeneSTAR Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Narayanaswamy P, Teo G, Ow JR, Lau A, Kaldis P, Tate S, Choi H. MetaboKit: a comprehensive data extraction tool for untargeted metabolomics. Mol Omics 2021; 16:436-447. [PMID: 32519713 DOI: 10.1039/d0mo00030b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have developed MetaboKit, a comprehensive software package for compound identification and relative quantification in mass spectrometry-based untargeted metabolomics analysis. In data dependent acquisition (DDA) analysis, MetaboKit constructs a customized spectral library with compound identities from reference spectral libraries, adducts, dimers, in-source fragments (ISF), MS/MS fragmentation spectra, and more importantly the retention time information unique to the chromatography system used in the experiment. Using the customized library, the software performs targeted peak integration for precursor ions in DDA analysis and for precursor and product ions in data independent acquisition (DIA) analysis. With its stringent identification algorithm requiring matches by both MS and MS/MS data, MetaboKit provides identification results with significantly greater specificity than the competing software packages without loss in sensitivity. The proposed MS/MS-based screening of ISFs also reduces the chance of unverifiable identification of ISFs considerably. MetaboKit's quantification module produced peak area values highly correlated with known concentrations in a DIA analysis of the metabolite standards at both MS1 and MS2 levels. Moreover, the analysis of Cdk1Liv-/- mouse livers showed that MetaboKit can identify a wide range of lipid species and their ISFs, and quantitatively reconstitute the well-characterized fatty liver phenotype in these mice. In DIA data, the MS1-level and MS2-level peak area data produced similar fold change estimates in the differential abundance analysis, and the MS2-level peak area data allowed for quantitative comparisons in compounds whose precursor ion chromatogram was too noisy for peak integration.
Collapse
Affiliation(s)
| | - Guoshou Teo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Jin Rong Ow
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | | | - Philipp Kaldis
- Institute of Molecular and Cell Biology, A*STAR, Singapore and Department of Clinical Sciences, Lund University, Sweden
| | | | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore. and Institute of Molecular and Cell Biology, A*STAR, Singapore
| |
Collapse
|
49
|
Dabke K, Kreimer S, Jones MR, Parker SJ. A Simple Optimization Workflow to Enable Precise and Accurate Imputation of Missing Values in Proteomic Data Sets. J Proteome Res 2021; 20:3214-3229. [PMID: 33939434 DOI: 10.1021/acs.jproteome.1c00070] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Missing values in proteomic data sets have real consequences on downstream data analysis and reproducibility. Although several imputation methods exist to handle missing values, no single imputation method is best suited for a diverse range of data sets, and no clear strategy exists for evaluating imputation methods for clinical DIA-MS data sets, especially at different levels of protein quantification. To navigate through the different imputation strategies available in the literature, we have established a strategy to assess imputation methods on clinical label-free DIA-MS data sets. We used three DIA-MS data sets with real missing values to evaluate eight imputation methods with multiple parameters at different levels of protein quantification: a dilution series data set, a small pilot data set, and a clinical proteomic data set comparing paired tumor and stroma tissue. We found that imputation methods based on local structures within the data, like local least-squares (LLS) and random forest (RF), worked well in our dilution series data set, whereas imputation methods based on global structures within the data, like BPCA, performed well in the other two data sets. We also found that imputation at the most basic protein quantification level-fragment level-improved accuracy and the number of proteins quantified. With this analytical framework, we quickly and cost-effectively evaluated different imputation methods using two smaller complementary data sets to narrow down to the larger proteomic data set's most accurate methods. This acquisition strategy allowed us to provide reproducible evidence of the accuracy of the imputation method, even in the absence of a ground truth. Overall, this study indicates that the most suitable imputation method relies on the overall structure of the data set and provides an example of an analytic framework that may assist in identifying the most appropriate imputation strategies for the differential analysis of proteins.
Collapse
Affiliation(s)
- Kruttika Dabke
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States.,Graduate Program in Biomedical Sciences, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Simion Kreimer
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Departments of Cardiology and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Michelle R Jones
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Sarah J Parker
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Departments of Cardiology and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| |
Collapse
|
50
|
Tan YM, Gao Y, Teo G, Koh HW, Tai ES, Khoo CM, Choi KP, Zhou L, Choi H. Plasma Metabolome and Lipidome Associations with Type 2 Diabetes and Diabetic Nephropathy. Metabolites 2021; 11:metabo11040228. [PMID: 33918080 PMCID: PMC8069978 DOI: 10.3390/metabo11040228] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
We conducted untargeted metabolomics analysis of plasma samples from a cross-sectional case–control study with 30 healthy controls, 30 patients with diabetes mellitus and normal renal function (DM-N), and 30 early diabetic nephropathy (DKD) patients using liquid chromatography-mass spectrometry (LC-MS). We employed two different modes of MS acquisition on a high-resolution MS instrument for identification and semi-quantification, and analyzed data using an advanced multivariate method for prioritizing differentially abundant metabolites. We obtained semi-quantification data for 1088 unique compounds (~55% lipids), excluding compounds that may be either exogenous compounds or treated as medication. Supervised classification analysis over a confounding-free partial correlation network shows that prostaglandins, phospholipids, nucleotides, sugars, and glycans are elevated in the DM-N and DKD patients, whereas glutamine, phenylacetylglutamine, 3-indoxyl sulfate, acetylphenylalanine, xanthine, dimethyluric acid, and asymmetric dimethylarginine are increased in DKD compared to DM-N. The data recapitulate the well-established plasma metabolome changes associated with DM-N and suggest uremic solutes and oxidative stress markers as the compounds indicating early renal function decline in DM patients.
Collapse
Affiliation(s)
- Yan Ming Tan
- Department of Statistics and Applied Probability, Faculty of Science, National University of Singapore, Singapore 117546, Singapore; (Y.M.T.); (K.P.C.)
| | - Yan Gao
- Singapore Eye Research Institute, The Academia, 20 College Road, Singapore 169856, Singapore;
| | - Guoshou Teo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (G.T.); (H.W.L.K.); (E.S.T.); (C.M.K.)
| | - Hiromi W.L. Koh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (G.T.); (H.W.L.K.); (E.S.T.); (C.M.K.)
| | - E Shyong Tai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (G.T.); (H.W.L.K.); (E.S.T.); (C.M.K.)
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (G.T.); (H.W.L.K.); (E.S.T.); (C.M.K.)
| | - Kwok Pui Choi
- Department of Statistics and Applied Probability, Faculty of Science, National University of Singapore, Singapore 117546, Singapore; (Y.M.T.); (K.P.C.)
| | - Lei Zhou
- Singapore Eye Research Institute, The Academia, 20 College Road, Singapore 169856, Singapore;
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Research Program, Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
- Correspondence: (L.Z.); (H.C.)
| | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (G.T.); (H.W.L.K.); (E.S.T.); (C.M.K.)
- Correspondence: (L.Z.); (H.C.)
| |
Collapse
|