1
|
Neirinck J, Buysse M, De Vriendt C, Hofmans M, Bonroy C. The role of immunophenotyping in common variable immunodeficiency: a narrative review. Crit Rev Clin Lab Sci 2025; 62:65-84. [PMID: 39364936 DOI: 10.1080/10408363.2024.2404842] [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: 06/24/2024] [Revised: 08/06/2024] [Accepted: 09/12/2024] [Indexed: 10/05/2024]
Abstract
Common variable immunodeficiency (CVID) is a heterogeneous primary immunodeficiency (PID) characterized by an impaired immunoglobulin production, in association with an increased susceptibility to infections and a diversity of clinical manifestations. This narrative review summarizes immunophenotypic abnormalities in CVID patients and their relevance for diagnosis and disease classification. A comprehensive search across four databases - PubMED, Web of Science, EMBASE and Google Scholar - yielded 170 relevant studies published between 1988 and April 31, 2023. Over the past decades, the role of immunophenotyping in CVID diagnosis has become evident by identifying "hallmark" immunophenotypic aberrancies in patient subsets, with some now integrated in the consensus diagnostic criteria. Furthermore, the role of immunophenotyping in subclassifying CVID in relation to clinical presentation and prognosis has been extensively studied. Certain immunophenotypic patterns consistently correlate with clinical manifestations and/or subsets of CVID, particularly those associated with noninfectious complications (i.e. low switched memory B cells, shifts in follicular helper T cell subsets, low naïve CD4+ T cells, low regulatory T cells, and expansion of CD21low B cells, often associated with autoimmunity and/or splenomegaly). Also, efforts to associate subset levels of innate immune cells, such as Natural Killer (NK) cells, invariant (i)NKT cells, innate lymphoid cells (ILCs), and dendritic cells (DCs) to CVID complications are evident albeit in a lesser degree. However, inconsistencies regarding the role of flow cytometry in classification and prognosis persist, reflecting the disease complexity, but probably also cohort variations and methodological differences between published studies. This underscores the need for collaborative efforts to integrate emerging concepts, such as standardized flow cytometry and computational tools, for a more precise CVID classification approach. Additionally, recent studies suggest a potential value of (epi)genetic-based molecular assays to this effort.
Collapse
Affiliation(s)
- Jana Neirinck
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Malicorne Buysse
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ciel De Vriendt
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Mattias Hofmans
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
2
|
Neirinck J, Buysse M, Brdickova N, Perez-Andres M, De Vriendt C, Kerre T, Haerynck F, Bossuyt X, van Dongen JJM, Orfao A, Hofmans M, Bonroy C, Kalina T. The EuroFlow PIDOT external quality assurance scheme: enhancing laboratory performance evaluation in immunophenotyping of rare lymphoid immunodeficiencies. Clin Chem Lab Med 2025; 63:621-635. [PMID: 39423371 DOI: 10.1515/cclm-2024-0749] [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: 06/26/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES The development of External Quality Assessment Schemes (EQAS) for clinical flow cytometry (FCM) is challenging in the context of rare (immunological) diseases. Here, we introduce a novel EQAS monitoring the primary immunodeficiency Orientation Tube (PIDOT), developed by EuroFlow, in both a 'wet' and 'dry' format. This EQAS provides feedback on the quality of individual laboratories (i.e., accuracy, reproducibility and result interpretation), while eliminating the need for sample distribution. METHODS In the wet format, marker staining intensities (MedFIs) within landmark cell populations in PIDOT analysis performed on locally collected healthy control (HC) samples, were compared to EQAS targets. In the dry format, participants analyzed centrally distributed PIDOT flow cytometry data (n=10). RESULTS We report the results of six EQAS rounds across 20 laboratories in 11 countries. The wet format (212 HC samples) demonstrated consistent technical performance among laboratories (median %rCV on MedFIs=34.5 %; average failure rate 17.3 %) and showed improvement upon repeated participation. The dry format demonstrated effective proficiency of participants in cell count enumeration (range %rCVs 3.1-7.1 % for the major lymphoid subsets), and in identifying lymphoid abnormalities (79.3 % alignment with reference). CONCLUSIONS The PIDOT-EQAS allows laboratories, adhering to the standardized EuroFlow approach, to monitor interlaboratory variations without the need for sample distribution, and provides them educational support to recognize rare clinically relevant immunophenotypic patterns of primary immunodeficiencies (PID). This EQAS contributes to quality improvement of PID diagnostics and can serve as an example for future flow cytometry EQAS in the context of rare diseases.
Collapse
Affiliation(s)
- Jana Neirinck
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Malicorne Buysse
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Naděžda Brdickova
- CLIP Cytometry, Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Martín Perez-Andres
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Ciel De Vriendt
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Tessa Kerre
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Pediatric Pulmonology and Immunology and PID Research Laboratory, University Hospital Ghent, Ghent, Belgium
| | - Xavier Bossuyt
- Department of Laboratory Medicine, University Hospital Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jacques J M van Dongen
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Mattias Hofmans
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Tomas Kalina
- CLIP Cytometry, Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
3
|
Macchia I, La Sorsa V, Ciervo A, Ruspantini I, Negri D, Borghi M, De Angelis ML, Luciani F, Martina A, Taglieri S, Durastanti V, Altavista MC, Urbani F, Mancini F. T Cell Peptide Prediction, Immune Response, and Host-Pathogen Relationship in Vaccinated and Recovered from Mild COVID-19 Subjects. Biomolecules 2024; 14:1217. [PMID: 39456150 PMCID: PMC11505848 DOI: 10.3390/biom14101217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 remains a significant threat, particularly to vulnerable populations. The emergence of new variants necessitates the development of treatments and vaccines that induce both humoral and cellular immunity. This study aimed to identify potentially immunogenic SARS-CoV-2 peptides and to explore the intricate host-pathogen interactions involving peripheral immune responses, memory profiles, and various demographic, clinical, and lifestyle factors. Using in silico and experimental methods, we identified several CD8-restricted SARS-CoV-2 peptides that are either poorly studied or have previously unreported immunogenicity: fifteen from the Spike and three each from non-structural proteins Nsp1-2-3-16. A Spike peptide, LA-9, demonstrated a 57% response rate in ELISpot assays using PBMCs from 14 HLA-A*02:01 positive, vaccinated, and mild-COVID-19 recovered subjects, indicating its potential for diagnostics, research, and multi-epitope vaccine platforms. We also found that younger individuals, with fewer vaccine doses and longer intervals since infection, showed lower anti-Spike (ELISA) and anti-Wuhan neutralizing antibodies (pseudovirus assay), higher naïve T cells, and lower central memory, effector memory, and CD4hiCD8low T cells (flow cytometry) compared to older subjects. In our cohort, a higher prevalence of Vδ2-γδ and DN T cells, and fewer naïve CD8 T cells, seemed to correlate with strong cellular and lower anti-NP antibody responses and to associate with Omicron infection, absence of confusional state, and habitual sporting activity.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina La Sorsa
- Research Promotion and Coordination Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Alessandra Ciervo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Irene Ruspantini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Francesca Luciani
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Antonio Martina
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Silvia Taglieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina Durastanti
- Neurology Unit, San Filippo Neri Hospital, ASL RM1, 00135 Rome, Italy; (V.D.); (M.C.A.)
| | | | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Fabiola Mancini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| |
Collapse
|
4
|
Nemzow L, Boehringer T, Mayenburg J, Beaton-Green LA, Wilkins RC, Turner HC. Interlaboratory comparison of high-throughput protein biomarker assay quantifications for radiation exposure classification. PLoS One 2024; 19:e0301418. [PMID: 38683751 PMCID: PMC11057749 DOI: 10.1371/journal.pone.0301418] [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: 11/09/2023] [Accepted: 03/15/2024] [Indexed: 05/02/2024] Open
Abstract
In the event of a widespread radiological incident, thousands of individuals will require rapid assessment of exposure using validated biodosimetry assays to inform clinical triage. In this scenario, multiple biodosimetry laboratories may be necessary for large-volume sample processing. To meet this need, we have developed a high-throughput assay for the rapid measurement of intracellular protein biomarkers in human peripheral blood samples using an Imaging Flow Cytometry (IFC) platform. The objective of this work was to harmonize and validate the reproducibility of our blood biomarker assay for radiation exposure across three IFC instruments, two located at Columbia University (CU) and the third at Health Canada. The Center for Radiological Research (CRR) at CU served as the central laboratory and reference instrument, where samples were prepared in triplicate, labeled with two radiation responsive leukocyte biomarkers (BAX and phosphor-p53 (Ser37)), and distributed for simultaneous interrogation by each IFC. Initial tests showed that significantly different baseline biomarker measurements were generated on each instrument when using the same acquisition settings, suggesting that harmonization of signal intensities is necessary. Subsequent tests harmonized biomarker measurements after irradiation by modulating laser intensity using two reference materials: unstained samples and standardized rainbow beads. Both methods generated measurements on each instrument without significant differences between the new and references instruments, allowing for the use of one master template to quantify biomarker expression across multiple instruments. Deming regression analyses of 0-5 Gy dose-response curves showed overall good correlation of BAX and p53 values across new and reference instruments. While Bland-Altman analyses indicated low to moderate instrument biases, ROC Curve analyses ultimately show successful discrimination between exposed and unexposed samples on each instrument (AUC values > 0.85).
Collapse
Affiliation(s)
- Leah Nemzow
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Thomas Boehringer
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Jessica Mayenburg
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | | | - Ruth C. Wilkins
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Helen C. Turner
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York, United States of America
| |
Collapse
|
5
|
Rybakowska P, Alarcón-Riquelme ME, Marañón C. Approaching Mass Cytometry Translational Studies by Experimental and Data Curation Settings. Methods Mol Biol 2024; 2779:369-394. [PMID: 38526795 DOI: 10.1007/978-1-0716-3738-8_17] [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: 03/27/2024]
Abstract
Clinical studies are conducted to better understand the pathological mechanism of diseases and to find biomarkers associated with disease activity, drug response, or outcome prediction. Mass cytometry (MC) is a high-throughput single-cell technology that measures hundreds of cells per second with more than 40 markers per cell. Thus, it is a suitable tool for immune monitoring and biomarker discovery studies. Working in translational and clinical settings requires a careful experimental design to minimize, monitor, and correct the variations introduced during sample collection, preparation, acquisition, and analysis. In this review, we will focus on these important aspects of MC-related experiments and data curation in the context of translational clinical research projects.
Collapse
Affiliation(s)
- Paulina Rybakowska
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Marta E Alarcón-Riquelme
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Institute for Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Concepción Marañón
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain.
| |
Collapse
|
6
|
Ortega-Ferreira C, Soret P, Robin G, Speca S, Hubert S, Le Gall M, Desvaux E, Jendoubi M, Saint-Paul J, Chadli L, Chomel A, Berger S, Nony E, Neau B, Fould B, Licznar A, Levasseur F, Guerrier T, Elouej S, Courtade-Gaïani S, Provost N, Nguyen TQ, Verdier J, Launay D, De Ceuninck F. Antibody-mediated neutralization of galectin-3 as a strategy for the treatment of systemic sclerosis. Nat Commun 2023; 14:5291. [PMID: 37652913 PMCID: PMC10471779 DOI: 10.1038/s41467-023-41117-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune, inflammatory and fibrotic disease with limited treatment options. Developing new therapies is therefore crucial to address patient needs. To this end, we focused on galectin-3 (Gal-3), a lectin known to be associated with several pathological processes seen in SSc. Using RNA sequencing of whole-blood samples in a cross-sectional cohort of 249 patients with SSc, Gal-3 and its interactants defined a strong transcriptomic fingerprint associated with disease severity, pulmonary and cardiac malfunctions, neutrophilia and lymphopenia. We developed new Gal-3 neutralizing monoclonal antibodies (mAb), which were then evaluated in a mouse model of hypochlorous acid (HOCl)-induced SSc. We show that two of these antibodies, D11 and E07, reduced pathological skin thickening, lung and skin collagen deposition, pulmonary macrophage content, and plasma interleukin-5 and -6 levels. Moreover, E07 changed the transcriptional profiles of HOCl-treated mice, resulting in a gene expression pattern that resembled that of control mice. Similarly, pathological pathways engaged in patients with SSc were counteracted by E07 in mice. Collectively, these findings demonstrate the translational potential of Gal-3 blockade as a therapeutic option for SSc.
Collapse
Affiliation(s)
- Céline Ortega-Ferreira
- Servier R&D Center, Biomarker Assay Development, Translational Medicine, Gif-sur-Yvette, France
| | - Perrine Soret
- Servier R&D Center, Biomarker Biostatistics, Gif-sur-Yvette, France
| | | | - Silvia Speca
- U1286 INFINITE, Institute for Translational Research in Inflammation, Lille University, Gif-sur-Yvette, France
- Inserm, Lille, France
| | - Sandra Hubert
- Servier R&D Center, Neurosciences and Immuno-inflammation Therapeutic Area, Gif-sur-Yvette, France
| | | | - Emiko Desvaux
- Servier R&D Center, Neurosciences and Immuno-inflammation Therapeutic Area, Gif-sur-Yvette, France
| | - Manel Jendoubi
- U1286 INFINITE, Institute for Translational Research in Inflammation, Lille University, Gif-sur-Yvette, France
- Inserm, Lille, France
| | | | - Loubna Chadli
- Servier R&D Center, Clinical Biomarker Development, Translational Medicine, Gif-sur-Yvette, France
| | - Agnès Chomel
- Servier R&D Center, Protein Sciences, Gif-sur-Yvette, France
| | - Sylvie Berger
- Servier R&D Center, Structural Sciences, Gif-sur-Yvette, France
| | - Emmanuel Nony
- Servier R&D Center, Protein Sciences, Gif-sur-Yvette, France
| | - Béatrice Neau
- Servier R&D Center, Preclinical Biostatistics, Quantitative Pharmacology, Gif-sur-Yvette, France
| | - Benjamin Fould
- Servier R&D Center, Protein Sciences, Gif-sur-Yvette, France
| | - Anne Licznar
- Servier R&D Center, DMPK Department, Translational Medicine, Gif-sur-Yvette, France
| | - Franck Levasseur
- Servier R&D Center, DMPK Department, Translational Medicine, Gif-sur-Yvette, France
| | - Thomas Guerrier
- U1286 INFINITE, Institute for Translational Research in Inflammation, Lille University, Gif-sur-Yvette, France
- Inserm, Lille, France
| | - Sahar Elouej
- Servier R&D Center, Computational Medicine, Gif-sur-Yvette, France
| | | | - Nicolas Provost
- Servier R&D Center, Molecular Genomics, Gif-sur-Yvette, France
| | | | - Julien Verdier
- Servier R&D Center, Neurosciences and Immuno-inflammation Therapeutic Area, Gif-sur-Yvette, France
| | - David Launay
- U1286 INFINITE, Institute for Translational Research in Inflammation, Lille University, Gif-sur-Yvette, France
- Inserm, Lille, France
- Lille University Hospital, Department of Internal Medicine and Clinical Immunology, Reference Center for Rare Systemic Autoimmune Diseases, North and North-West France (CeRAINO), Lille, France
| | - Frédéric De Ceuninck
- Servier R&D Center, Neurosciences and Immuno-inflammation Therapeutic Area, Gif-sur-Yvette, France.
| |
Collapse
|
7
|
Macchia I, La Sorsa V, Urbani F, Moretti S, Antonucci C, Afferni C, Schiavoni G. Eosinophils as potential biomarkers in respiratory viral infections. Front Immunol 2023; 14:1170035. [PMID: 37483591 PMCID: PMC10358847 DOI: 10.3389/fimmu.2023.1170035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Eosinophils are bone marrow-derived granulocytes that, under homeostatic conditions, account for as much as 1-3% of peripheral blood leukocytes. During inflammation, eosinophils can rapidly expand and infiltrate inflamed tissues, guided by cytokines and alarmins (such as IL-33), adhesion molecules and chemokines. Eosinophils play a prominent role in allergic asthma and parasitic infections. Nonetheless, they participate in the immune response against respiratory viruses such as respiratory syncytial virus and influenza. Notably, respiratory viruses are associated with asthma exacerbation. Eosinophils release several molecules endowed with antiviral activity, including cationic proteins, RNases and reactive oxygen and nitrogen species. On the other hand, eosinophils release several cytokines involved in homeostasis maintenance and Th2-related inflammation. In the context of SARS-CoV-2 infection, emerging evidence indicates that eosinophils can represent possible blood-based biomarkers for diagnosis, prognosis, and severity prediction of disease. In particular, eosinopenia seems to be an indicator of severity among patients with COVID-19, whereas an increased eosinophil count is associated with a better prognosis, including a lower incidence of complications and mortality. In the present review, we provide an overview of the role and plasticity of eosinophils focusing on various respiratory viral infections and in the context of viral and allergic disease comorbidities. We will discuss the potential utility of eosinophils as prognostic/predictive immune biomarkers in emerging respiratory viral diseases, particularly COVID-19. Finally, we will revisit some of the relevant methods and tools that have contributed to the advances in the dissection of various eosinophil subsets in different pathological settings for future biomarker definition.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina La Sorsa
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Caterina Antonucci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Claudia Afferni
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
8
|
Gao J, Luo Y, Li H, Zhao Y, Zhao J, Han X, Han J, Lin H, Qian F. Deep Immunophenotyping of Human Whole Blood by Standardized Multi-parametric Flow Cytometry Analyses. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:309-328. [PMID: 37325713 PMCID: PMC10260734 DOI: 10.1007/s43657-022-00092-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 06/17/2023]
Abstract
Immunophenotyping is proving crucial to understanding the role of the immune system in health and disease. High-throughput flow cytometry has been used extensively to reveal changes in immune cell composition and function at the single-cell level. Here, we describe six optimized 11-color flow cytometry panels for deep immunophenotyping of human whole blood. A total of 51 surface antibodies, which are readily available and validated, were selected to identify the key immune cell populations and evaluate their functional state in a single assay. The gating strategies for effective flow cytometry data analysis are included in the protocol. To ensure data reproducibility, we provide detailed procedures in three parts, including (1) instrument characterization and detector gain optimization, (2) antibody titration and sample staining, and (3) data acquisition and quality checks. This standardized approach has been applied to a variety of donors for a better understanding of the complexity of the human immune system. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-022-00092-9.
Collapse
Affiliation(s)
- Jian Gao
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Yali Luo
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Helian Li
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Yiran Zhao
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Jialin Zhao
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Xuling Han
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Jingxuan Han
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Huiqin Lin
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Feng Qian
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center and School of Life Sciences, Fudan University, Shanghai, 200438 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438 China
- Institute of Immunophenome, International Human Phenome Institutes (Shanghai), Shanghai, 200433 China
| |
Collapse
|
9
|
Rybakowska P, Van Gassen S, Martorell Marugán J, Quintelier K, Saeys Y, Alarcón-Riquelme ME, Marañón C. Protocol for large scale whole blood immune monitoring by mass cytometry and Cyto Quality Pipeline. STAR Protoc 2022; 3:101697. [PMID: 36353363 PMCID: PMC9637821 DOI: 10.1016/j.xpro.2022.101697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Mass cytometry (MC) is a powerful large-scale immune monitoring technology. To maximize MC data quality, we present a protocol for whole blood analysis together with an R package, Cyto Quality Pipeline (CytoQP), which minimizes the experimental artifacts and batch effects to ensure data reproducibility. We describe the steps to stimulate, fix, and freeze blood samples before acquisition to make them suitable for retrospective studies. We then detail the use of barcoding and reference samples to facilitate multicenter and multi-batch experiments. For complete details on the use and execution of this protocol, please refer to Rybakowska et al. (2021a) and (2021b).
Collapse
Affiliation(s)
- Paulina Rybakowska
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/ Andalusian Regional Government, PTS, 18016 Granada, Spain.
| | - Sofie Van Gassen
- Department of Applied Mathematics, Computer Sciences and Statistics, Ghent University, 9000 Gent, Belgium; Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, 9000 Gent, Belgium
| | - Jordi Martorell Marugán
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/ Andalusian Regional Government, PTS, 18016 Granada, Spain; Department of Statistics and Operational Research, University of Granada, 18071 Granada, Spain; Data Science for Health Research Unit, Fondazione Bruno Kessler, 38123 Trento, Italy
| | - Katrien Quintelier
- Department of Applied Mathematics, Computer Sciences and Statistics, Ghent University, 9000 Gent, Belgium; Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, 9000 Gent, Belgium; Department of Pulmonary Diseases, Erasmus MC, 3015 Rotterdam, the Netherlands
| | - Yvan Saeys
- Department of Applied Mathematics, Computer Sciences and Statistics, Ghent University, 9000 Gent, Belgium; Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, 9000 Gent, Belgium
| | - Marta E Alarcón-Riquelme
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/ Andalusian Regional Government, PTS, 18016 Granada, Spain; Institute for Environmental Medicine, Karolinska Institute, 17177 Stockholm, Sweden
| | - Concepción Marañón
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/ Andalusian Regional Government, PTS, 18016 Granada, Spain.
| |
Collapse
|
10
|
Vetrano S, Bouma G, Benschop RJ, Birngruber T, Costanzo A, D’Haens GRAM, Frasca L, Hillenbrand R, Iversen L, Johansen C, Kaser A, Koenen HJPM, Noehammer C, Peyrin-Biroulet L, Raes J, Ricotti L, Rosenstiel P, Satagopam VP, Schreiber S, Vermeire S, Wollenberg A, Weidinger S, Ziemek D, Danese S. ImmUniverse Consortium: Multi-omics integrative approach in personalized medicine for immune-mediated inflammatory diseases. Front Immunol 2022; 13:1002629. [PMID: 36439150 PMCID: PMC9682955 DOI: 10.3389/fimmu.2022.1002629] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/26/2022] [Indexed: 09/22/2023] Open
Abstract
Immune mediated inflammatory diseases (IMIDs) are a heterogeneous group of debilitating, multifactorial and unrelated conditions featured by a dysregulated immune response leading to destructive chronic inflammation. The immune dysregulation can affect various organ systems: gut (e.g., inflammatory bowel disease), joints (e.g., rheumatoid arthritis), skin (e.g., psoriasis, atopic dermatitis), resulting in significant morbidity, reduced quality of life, increased risk for comorbidities, and premature death. As there are no reliable disease progression and therapy response biomarkers currently available, it is very hard to predict how the disease will develop and which treatments will be effective in a given patient. In addition, a considerable proportion of patients do not respond sufficiently to the treatment. ImmUniverse is a large collaborative consortium of 27 partners funded by the Innovative Medicine Initiative (IMI), which is sponsored by the European Union (Horizon 2020) and in-kind contributions of participating pharmaceutical companies within the European Federation of Pharmaceutical Industries and Associations (EFPIA). ImmUniverse aims to advance our understanding of the molecular mechanisms underlying two immune-mediated diseases, ulcerative colitis (UC) and atopic dermatitis (AD), by pursuing an integrative multi-omics approach. As a consequence of the heterogeneity among IMIDs patients, a comprehensive, evidence-based identification of novel biomarkers is necessary to enable appropriate patient stratification that would account for the inter-individual differences in disease severity, drug efficacy, side effects or prognosis. This would guide clinicians in the management of patients and represent a major step towards personalized medicine. ImmUniverse will combine the existing and novel advanced technologies, including multi-omics, to characterize both the tissue microenvironment and blood. This comprehensive, systems biology-oriented approach will allow for identification and validation of tissue and circulating biomarker signatures as well as mechanistic principles, which will provide information about disease severity and future disease progression. This truly makes the ImmUniverse Consortium an unparalleled approach.
Collapse
Affiliation(s)
- Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- IBD Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Gerben Bouma
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline R&D, United Kingdom
| | - Robert J. Benschop
- Immunology and Translation, Eli Lilly and Company, Indianapolis, IN, United States
| | - Thomas Birngruber
- Joanneum Research GmbH, HEALTH - Institute for Biomedicine and Health Sciences, Graz, Austria
| | - Antonio Costanzo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - G. R. A. M. D’Haens
- Department of Gastroenterology and Hepatology, Academisch Medisch Centrum Bij De Universiteit Van Amsterdam, Amsterdam, Netherlands
| | - Loredana Frasca
- Pharmacological Research and Experimental Therapy Unit, Istituto Superiore Di Sanità, Roma, Italy
| | | | - Lars Iversen
- Department of Dermatology, Aarhus Universitetshospital, Aarhus, Denmark
| | | | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Hans J. P. M. Koenen
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Stichting Radboud Universitair Medisch Centrum, Nijmegen, Netherlands
| | - Christa Noehammer
- Department of Health and Environment, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Centre Hospitalier Regional Universitaire Nancy, Nancy, France
| | - Jeroen Raes
- Department of Microbiology and Immunology, Vib Vzw, Gent, Belgium
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Pisa, Italy
- Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian Albrechts University and University Hospital, Kiel, Germany
| | - Venkata P. Satagopam
- Luxembourg Centre for Systems Biomedicine, House of Biomedicine II, University of Luxembourg, Belvaux, Luxembourg
| | - Stefan Schreiber
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Severine Vermeire
- Department of Gastroenterology and Hepatology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Andreas Wollenberg
- Department of Dermatology, Ludwig-Maximilian-University Munich, Munich, Germany
- Department of Dermatology, Free University Brussels, University Hospital Brussels, Brussels, Belgium
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Daniel Ziemek
- Worldwide Research and Development, Pfizer Pharma, Berlin, Germany
| | - Silvio Danese
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Department of Gastroenterology and Endoscopy IRCCS Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
11
|
Liu Y, Zhao H, Fu B, Jiang S, Wang J, Wan Y. Mapping Cell Phenomics with Multiparametric Flow Cytometry Assays. PHENOMICS (CHAM, SWITZERLAND) 2022; 2:272-281. [PMID: 36939758 PMCID: PMC9590532 DOI: 10.1007/s43657-021-00031-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 11/26/2022]
Abstract
Phenomics explores the complex interactions among genes, epigenetics, symbiotic microorganisms, diet, and environmental exposure based on the physical, chemical, and biological characteristics of individuals and groups. Increasingly efficient and comprehensive phenotyping techniques have been integrated into modern phenomics-related research. Multicolor flow cytometry technology provides more measurement parameters than conventional flow cytometry. Based on detailed descriptions of cell phenotypes, rare cell populations and cell subsets can be distinguished, new cell phenotypes can be discovered, and cell apoptosis characteristics can be detected, which will expand the potential of cell phenomics research. Based on the enhancements in multicolor flow cytometry hardware, software, reagents, and method design, the present review summarizes the recent advances and applications of multicolor flow cytometry in cell phenomics, illuminating the potential of applying phenomics in future studies.
Collapse
Affiliation(s)
- Yang Liu
- Biomedical Analysis Center, Army Medical University, Chongqing, 400038 China
- Chongqing Key Laboratory of Cytomics, Chongqing, 400038 China
| | - Haichu Zhao
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518055 China
| | - Boqiang Fu
- National Institute of Metrology, Beijing, 100029 China
| | - Shan Jiang
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518055 China
| | - Jing Wang
- National Institute of Metrology, Beijing, 100029 China
| | - Ying Wan
- Biomedical Analysis Center, Army Medical University, Chongqing, 400038 China
- Chongqing Key Laboratory of Cytomics, Chongqing, 400038 China
| |
Collapse
|
12
|
2021 White Paper on Recent Issues in Bioanalysis: ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/Oral & Multispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry ( Part 2 - Recommendations on Biomarkers/CDx Assays Development & Validation, Cytometry Validation & Innovation, Biotherapeutics PK LBA Regulated Bioanalysis, Critical Reagents & Positive Controls Generation). Bioanalysis 2022; 14:627-692. [PMID: 35578974 DOI: 10.4155/bio-2022-0080] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The 15th edition of the Workshop on Recent Issues in Bioanalysis (15th WRIB) was held on 27 September to 1 October 2021. Even with a last-minute move from in-person to virtual, an overwhelmingly high number of nearly 900 professionals representing pharma and biotech companies, contract research organizations (CROs), and multiple regulatory agencies still eagerly convened to actively discuss the most current topics of interest in bioanalysis. The 15th WRIB included three Main Workshops and seven Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on biomarker assay development and validation (BAV) (focused on clarifying the confusion created by the increased use of the term "context of use" [COU]); mass spectrometry of proteins (therapeutic, biomarker and transgene); state-of-the-art cytometry innovation and validation; and critical reagent and positive control generation were the special features of the 15th edition. This 2021 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop, and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2021 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 2) covers the recommendations on ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/Oral & Multispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry. Part 1A (Endogenous Compounds, Small Molecules, Complex Methods, Regulated Mass Spec of Large Molecules, Small Molecule, PoC), Part 1B (Regulatory Agencies' Inputs on Bioanalysis, Biomarkers, Immunogenicity, Gene & Cell Therapy and Vaccine) and Part 3 (TAb/NAb, Viral Vector CDx, Shedding Assays; CRISPR/Cas9 & CAR-T Immunogenicity; PCR & Vaccine Assay Performance; ADA Assay Comparability & Cut Point Appropriateness) are published in volume 14 of Bioanalysis, issues 9 and 11 (2022), respectively.
Collapse
|
13
|
Rodriguez-Esteban R, Duarte J, Teixeira PC, Richard F, Koltsova S, So WV. Prediction of standard cell types and functional markers from textual descriptions of flow cytometry gating definitions using machine learning. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2022; 102:220-227. [PMID: 35253974 DOI: 10.1002/cyto.b.22065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND A key step in clinical flow cytometry data analysis is gating, which involves the identification of cell populations. The process of gating produces a set of reportable results, which are typically described by gating definitions. The non-standardized, non-interpreted nature of gating definitions represents a hurdle for data interpretation and data sharing across and within organizations. Interpreting and standardizing gating definitions for subsequent analysis of gating results requires a curation effort from experts. Machine learning approaches have the potential to help in this process by predicting expert annotations associated with gating definitions. METHODS We created a gold-standard dataset by manually annotating thousands of gating definitions with cell type and functional marker annotations. We used this dataset to train and test a machine learning pipeline able to predict standard cell types and functional marker genes associated with gating definitions. RESULTS The machine learning pipeline predicted annotations with high accuracy for both cell types and functional marker genes. Accuracy was lower for gating definitions from assays belonging to laboratories from which limited or no prior data was available in the training. Manual error review ensured that resulting predicted annotations could be reused subsequently as additional gold-standard training data. CONCLUSIONS Machine learning methods are able to consistently predict annotations associated with gating definitions from flow cytometry assays. However, a hybrid automatic and manual annotation workflow would be recommended to achieve optimal results.
Collapse
Affiliation(s)
- Raul Rodriguez-Esteban
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - José Duarte
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Priscila C Teixeira
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Fabien Richard
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Svetlana Koltsova
- Curation Department, Rancho BioSciences LLC, San Diego, California, USA
| | - W Venus So
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center New York, New York, USA
| |
Collapse
|
14
|
Teruel M, Barturen G, Martínez-Bueno M, Castellini-Pérez O, Barroso-Gil M, Povedano E, Kerick M, Català-Moll F, Makowska Z, Buttgereit A, Pers JO, Marañón C, Ballestar E, Martin J, Carnero-Montoro E, Alarcón-Riquelme ME. Integrative epigenomics in Sjögren´s syndrome reveals novel pathways and a strong interaction between the HLA, autoantibodies and the interferon signature. Sci Rep 2021; 11:23292. [PMID: 34857786 PMCID: PMC8640069 DOI: 10.1038/s41598-021-01324-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Primary Sjögren's syndrome (SS) is a systemic autoimmune disease characterized by lymphocytic infiltration and damage of exocrine salivary and lacrimal glands. The etiology of SS is complex with environmental triggers and genetic factors involved. By conducting an integrated multi-omics study, we confirmed a vast coordinated hypomethylation and overexpression effects in IFN-related genes, what is known as the IFN signature. Stratified and conditional analyses suggest a strong interaction between SS-associated HLA genetic variation and the presence of Anti-Ro/SSA autoantibodies in driving the IFN epigenetic signature and determining SS. We report a novel epigenetic signature characterized by increased DNA methylation levels in a large number of genes enriched in pathways such as collagen metabolism and extracellular matrix organization. We identified potential new genetic variants associated with SS that might mediate their risk by altering DNA methylation or gene expression patterns, as well as disease-interacting genetic variants that exhibit regulatory function only in the SS population. Our study sheds new light on the interaction between genetics, autoantibody profiles, DNA methylation and gene expression in SS, and contributes to elucidate the genetic architecture of gene regulation in an autoimmune population.
Collapse
Affiliation(s)
- María Teruel
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Guillermo Barturen
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Manuel Martínez-Bueno
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Olivia Castellini-Pérez
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Miguel Barroso-Gil
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Elena Povedano
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Martin Kerick
- IPBLN-CSIC, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, 18016, Granada, Spain
| | - Francesc Català-Moll
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916, Badalona, Barcelona, Spain
- IDIBELL, Bellvitge Biomedical Research Institute 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Zuzanna Makowska
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | - Anne Buttgereit
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | | | - Concepción Marañón
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916, Badalona, Barcelona, Spain
- IDIBELL, Bellvitge Biomedical Research Institute 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Martin
- IPBLN-CSIC, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, 18016, Granada, Spain
| | - Elena Carnero-Montoro
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain.
| | - Marta E Alarcón-Riquelme
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain.
- Institute for Environmental Medicine, Karolinska Institutet, 171 67, Solna, Sweden.
| |
Collapse
|
15
|
Soret P, Le Dantec C, Desvaux E, Foulquier N, Chassagnol B, Hubert S, Jamin C, Barturen G, Desachy G, Devauchelle-Pensec V, Boudjeniba C, Cornec D, Saraux A, Jousse-Joulin S, Barbarroja N, Rodríguez-Pintó I, De Langhe E, Beretta L, Chizzolini C, Kovács L, Witte T, Bettacchioli E, Buttgereit A, Makowska Z, Lesche R, Borghi MO, Martin J, Courtade-Gaiani S, Xuereb L, Guedj M, Moingeon P, Alarcón-Riquelme ME, Laigle L, Pers JO. A new molecular classification to drive precision treatment strategies in primary Sjögren's syndrome. Nat Commun 2021; 12:3523. [PMID: 34112769 PMCID: PMC8192578 DOI: 10.1038/s41467-021-23472-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/30/2021] [Indexed: 02/08/2023] Open
Abstract
There is currently no approved treatment for primary Sjögren's syndrome, a disease that primarily affects adult women. The difficulty in developing effective therapies is -in part- because of the heterogeneity in the clinical manifestation and pathophysiology of the disease. Finding common molecular signatures among patient subgroups could improve our understanding of disease etiology, and facilitate the development of targeted therapeutics. Here, we report, in a cross-sectional cohort, a molecular classification scheme for Sjögren's syndrome patients based on the multi-omic profiling of whole blood samples from a European cohort of over 300 patients, and a similar number of age and gender-matched healthy volunteers. Using transcriptomic, genomic, epigenetic, cytokine expression and flow cytometry data, combined with clinical parameters, we identify four groups of patients with distinct patterns of immune dysregulation. The biomarkers we identify can be used by machine learning classifiers to sort future patients into subgroups, allowing the re-evaluation of response to treatments in clinical trials.
Collapse
Affiliation(s)
- Perrine Soret
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | | | - Emiko Desvaux
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France
| | | | - Bastien Chassagnol
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | - Sandra Hubert
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | - Christophe Jamin
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France
- CHU de Brest, Brest, France
| | - Guillermo Barturen
- Department of Medical Genomics, Center for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Guillaume Desachy
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | | | - Cheïma Boudjeniba
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | - Divi Cornec
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France
- CHU de Brest, Brest, France
| | - Alain Saraux
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France
- CHU de Brest, Brest, France
| | | | - Nuria Barbarroja
- Reina Sofia Hospital, Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), University of Cordoba, Cordoba, Spain
| | - Ignasi Rodríguez-Pintó
- Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Catalonia, Spain
| | - Ellen De Langhe
- Skeletal Biology and Engineering Research Center, KU Leuven and Division of Rheumatology, UZ Leuven, Belgium
| | - Lorenzo Beretta
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Carlo Chizzolini
- Immunology & Allergy, University Hospital and School of Medicine, Geneva, Switzerland
| | | | - Torsten Witte
- Klinik für Immunologie und Rheumatologie, Medical University Hannover, Hannover, Germany
| | | | - Anne Buttgereit
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | - Zuzanna Makowska
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | - Ralf Lesche
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | | | - Javier Martin
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Sophie Courtade-Gaiani
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | - Laura Xuereb
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | - Mickaël Guedj
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | - Philippe Moingeon
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | - Marta E Alarcón-Riquelme
- Department of Medical Genomics, Center for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Laurence Laigle
- Institut de Recherches Internationales Servier, Departments of Translational Medicine and Immuno-Inflammatory Diseases Research and Development, Suresnes, France
| | | |
Collapse
|
16
|
Barturen G, Babaei S, Català-Moll F, Martínez-Bueno M, Makowska Z, Martorell-Marugán J, Carmona-Sáez P, Toro-Domínguez D, Carnero-Montoro E, Teruel M, Kerick M, Acosta-Herrera M, Le Lann L, Jamin C, Rodríguez-Ubreva J, García-Gómez A, Kageyama J, Buttgereit A, Hayat S, Mueller J, Lesche R, Hernandez-Fuentes M, Juarez M, Rowley T, White I, Marañón C, Gomes Anjos T, Varela N, Aguilar-Quesada R, Garrancho FJ, López-Berrio A, Rodriguez Maresca M, Navarro-Linares H, Almeida I, Azevedo N, Brandão M, Campar A, Faria R, Farinha F, Marinho A, Neves E, Tavares A, Vasconcelos C, Trombetta E, Montanelli G, Vigone B, Alvarez-Errico D, Li T, Thiagaran D, Blanco Alonso R, Corrales Martínez A, Genre F, López Mejías R, Gonzalez-Gay MA, Remuzgo S, Ubilla Garcia B, Cervera R, Espinosa G, Rodríguez-Pintó I, De Langhe E, Cremer J, Lories R, Belz D, Hunzelmann N, Baerlecken N, Kniesch K, Witte T, Lehner M, Stummvoll G, Zauner M, Aguirre-Zamorano MA, Barbarroja N, Castro-Villegas MC, Collantes-Estevez E, de Ramon E, Díaz Quintero I, Escudero-Contreras A, Fernández Roldán MC, Jiménez Gómez Y, Jiménez Moleón I, Lopez-Pedrera R, Ortega-Castro R, Ortego N, Raya E, Artusi C, Gerosa M, Meroni PL, Schioppo T, De Groof A, Ducreux J, Lauwerys B, Maudoux AL, Cornec D, Devauchelle-Pensec V, Jousse-Joulin S, Jouve PE, Rouvière B, Saraux A, Simon Q, Alvarez M, et alBarturen G, Babaei S, Català-Moll F, Martínez-Bueno M, Makowska Z, Martorell-Marugán J, Carmona-Sáez P, Toro-Domínguez D, Carnero-Montoro E, Teruel M, Kerick M, Acosta-Herrera M, Le Lann L, Jamin C, Rodríguez-Ubreva J, García-Gómez A, Kageyama J, Buttgereit A, Hayat S, Mueller J, Lesche R, Hernandez-Fuentes M, Juarez M, Rowley T, White I, Marañón C, Gomes Anjos T, Varela N, Aguilar-Quesada R, Garrancho FJ, López-Berrio A, Rodriguez Maresca M, Navarro-Linares H, Almeida I, Azevedo N, Brandão M, Campar A, Faria R, Farinha F, Marinho A, Neves E, Tavares A, Vasconcelos C, Trombetta E, Montanelli G, Vigone B, Alvarez-Errico D, Li T, Thiagaran D, Blanco Alonso R, Corrales Martínez A, Genre F, López Mejías R, Gonzalez-Gay MA, Remuzgo S, Ubilla Garcia B, Cervera R, Espinosa G, Rodríguez-Pintó I, De Langhe E, Cremer J, Lories R, Belz D, Hunzelmann N, Baerlecken N, Kniesch K, Witte T, Lehner M, Stummvoll G, Zauner M, Aguirre-Zamorano MA, Barbarroja N, Castro-Villegas MC, Collantes-Estevez E, de Ramon E, Díaz Quintero I, Escudero-Contreras A, Fernández Roldán MC, Jiménez Gómez Y, Jiménez Moleón I, Lopez-Pedrera R, Ortega-Castro R, Ortego N, Raya E, Artusi C, Gerosa M, Meroni PL, Schioppo T, De Groof A, Ducreux J, Lauwerys B, Maudoux AL, Cornec D, Devauchelle-Pensec V, Jousse-Joulin S, Jouve PE, Rouvière B, Saraux A, Simon Q, Alvarez M, Chizzolini C, Dufour A, Wynar D, Balog A, Bocskai M, Deák M, Dulic S, Kádár G, Kovács L, Cheng Q, Gerl V, Hiepe F, Khodadadi L, Thiel S, de Rinaldis E, Rao S, Benschop RJ, Chamberlain C, Dow ER, Ioannou Y, Laigle L, Marovac J, Wojcik J, Renaudineau Y, Borghi MO, Frostegård J, Martín J, Beretta L, Ballestar E, McDonald F, Pers JO, Alarcón-Riquelme ME. Integrative Analysis Reveals a Molecular Stratification of Systemic Autoimmune Diseases. Arthritis Rheumatol 2021; 73:1073-1085. [PMID: 33497037 DOI: 10.1002/art.41610] [Show More Authors] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/01/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Clinical heterogeneity, a hallmark of systemic autoimmune diseases, impedes early diagnosis and effective treatment, issues that may be addressed if patients could be classified into groups defined by molecular pattern. This study was undertaken to identify molecular clusters for reclassifying systemic autoimmune diseases independently of clinical diagnosis. METHODS Unsupervised clustering of integrated whole blood transcriptome and methylome cross-sectional data on 955 patients with 7 systemic autoimmune diseases and 267 healthy controls was undertaken. In addition, an inception cohort was prospectively followed up for 6 or 14 months to validate the results and analyze whether or not cluster assignment changed over time. RESULTS Four clusters were identified and validated. Three were pathologic, representing "inflammatory," "lymphoid," and "interferon" patterns. Each included all diagnoses and was defined by genetic, clinical, serologic, and cellular features. A fourth cluster with no specific molecular pattern was associated with low disease activity and included healthy controls. A longitudinal and independent inception cohort showed a relapse-remission pattern, where patients remained in their pathologic cluster, moving only to the healthy one, thus showing that the molecular clusters remained stable over time and that single pathogenic molecular signatures characterized each individual patient. CONCLUSION Patients with systemic autoimmune diseases can be jointly stratified into 3 stable disease clusters with specific molecular patterns differentiating different molecular disease mechanisms. These results have important implications for future clinical trials and the study of nonresponse to therapy, marking a paradigm shift in our view of systemic autoimmune diseases.
Collapse
Affiliation(s)
- Guillermo Barturen
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | | | | | - Manuel Martínez-Bueno
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | | | - Jordi Martorell-Marugán
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | - Pedro Carmona-Sáez
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | - Daniel Toro-Domínguez
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | - Elena Carnero-Montoro
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | - María Teruel
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | - Martin Kerick
- Institute of Parasitology and Biomedicine "López Neyra", Spanish National Research Council, Granada, Spain
| | - Marialbert Acosta-Herrera
- Institute of Parasitology and Biomedicine "López Neyra", Spanish National Research Council, Granada, Spain
| | - Lucas Le Lann
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | - Christophe Jamin
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | | | | | | | | | | | | | | | | | | | | | | | - Concepción Marañón
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | - Tania Gomes Anjos
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | - Nieves Varela
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain
| | | | | | | | | | | | | | | | | | - Ana Campar
- Centro Hospitalar do Porto, Porto, Portugal
| | | | | | | | | | | | | | - Elena Trombetta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gaia Montanelli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Vigone
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Tianlu Li
- Bellvitge Biomedical Research Institute, Barcelona, Spain
| | | | - Ricardo Blanco Alonso
- Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain
| | | | - Fernanda Genre
- Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Raquel López Mejías
- Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Miguel A Gonzalez-Gay
- Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Sara Remuzgo
- Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Begoña Ubilla Garcia
- Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Ricard Cervera
- Hospital Clínic and Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Gerard Espinosa
- Hospital Clínic and Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Ignasi Rodríguez-Pintó
- Hospital Clínic and Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Ellen De Langhe
- Katholieke Universiteit Leuven and Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Jonathan Cremer
- Katholieke Universiteit Leuven and Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Rik Lories
- Katholieke Universiteit Leuven and Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Doreen Belz
- Klinikum der Universitaet zu Koeln, Cologne, Germany
| | | | | | | | | | | | | | | | | | - Nuria Barbarroja
- Reina Sofia University Hospital and University of Cordoba, Cordoba, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Aurélie De Groof
- Université Catholique de Louvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Julie Ducreux
- Université Catholique de Louvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bernard Lauwerys
- Université Catholique de Louvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Anne-Lise Maudoux
- Université Catholique de Louvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Divi Cornec
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | | | - Sandrine Jousse-Joulin
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | | | - Bénédicte Rouvière
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | - Alain Saraux
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | - Quentin Simon
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | | | | | | | | | | | | | | | | | | | | | - Qingyu Cheng
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Velia Gerl
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Falk Hiepe
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Silvia Thiel
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | - Laurence Laigle
- Institut de Recherches Internationales Servier, Suresnes, France
| | | | | | - Yves Renaudineau
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | | | | | - Javier Martín
- Institute of Parasitology and Biomedicine "López Neyra", Spanish National Research Council, Granada, Spain
| | - Lorenzo Beretta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | - Jacques-Olivier Pers
- Université de Brest, Centre Hospitalier Universitaire de Brest, INSERM, and Labex IGO, Brest, France
| | - Marta E Alarcón-Riquelme
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Granada, Spain, and Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Calabrese DR, Aminian E, Mallavia B, Liu F, Cleary SJ, Aguilar OA, Wang P, Singer JP, Hays SR, Golden JA, Kukreja J, Dugger D, Nakamura M, Lanier LL, Looney MR, Greenland JR. Natural killer cells activated through NKG2D mediate lung ischemia-reperfusion injury. J Clin Invest 2021; 131:137047. [PMID: 33290276 PMCID: PMC7852842 DOI: 10.1172/jci137047] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary ischemia-reperfusion injury (IRI) is a clinical syndrome of acute lung injury that occurs after lung transplantation or remote organ ischemia. IRI causes early mortality and has no effective therapies. While NK cells are innate lymphocytes capable of recognizing injured cells, their roles in acute lung injury are incompletely understood. Here, we demonstrated that NK cells were increased in frequency and cytotoxicity in 2 different IRI mouse models. We showed that NK cells trafficked to the lung tissue from peripheral reservoirs and were more mature within lung tissue. Acute lung ischemia-reperfusion injury was blunted in a NK cell-deficient mouse strain but restored with adoptive transfer of NK cells. Mechanistically, NK cell NKG2D receptor ligands were induced on lung endothelial and epithelial cells following IRI, and antibody-mediated NK cell depletion or NKG2D stress receptor blockade abrogated acute lung injury. In human lung tissue, NK cells were increased at sites of ischemia-reperfusion injury and activated NK cells were increased in prospectively collected human bronchoalveolar lavage in subjects with severe IRI. These data support a causal role for recipient peripheral NK cells in pulmonary IRI via NK cell NKG2D receptor ligation. Therapies targeting NK cells may hold promise in acute lung injury.
Collapse
Affiliation(s)
- Daniel R. Calabrese
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Emily Aminian
- Department of Medicine, University of California, San Francisco, California
| | - Benat Mallavia
- Department of Medicine, University of California, San Francisco, California
| | - Fengchun Liu
- Department of Medicine, University of California, San Francisco, California
| | - Simon J. Cleary
- Department of Medicine, University of California, San Francisco, California
| | - Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Ping Wang
- Department of Medicine, University of California, San Francisco, California
| | - Jonathan P. Singer
- Department of Medicine, University of California, San Francisco, California
| | - Steven R. Hays
- Department of Medicine, University of California, San Francisco, California
| | - Jeffrey A. Golden
- Department of Medicine, University of California, San Francisco, California
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California
| | - Daniel Dugger
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Mary Nakamura
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco, California
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| |
Collapse
|
18
|
Bossini-Castillo L, Villanueva-Martin G, Kerick M, Acosta-Herrera M, López-Isac E, Simeón CP, Ortego-Centeno N, Assassi S, Hunzelmann N, Gabrielli A, de Vries-Bouwstra JK, Allanore Y, Fonseca C, Denton CP, Radstake TR, Alarcón-Riquelme ME, Beretta L, Mayes MD, Martin J. Genomic Risk Score impact on susceptibility to systemic sclerosis. Ann Rheum Dis 2021; 80:118-127. [PMID: 33004331 DOI: 10.1136/annrheumdis-2020-218558] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Genomic Risk Scores (GRS) successfully demonstrated the ability of genetics to identify those individuals at high risk for complex traits including immune-mediated inflammatory diseases (IMIDs). We aimed to test the performance of GRS in the prediction of risk for systemic sclerosis (SSc) for the first time. METHODS Allelic effects were obtained from the largest SSc Genome-Wide Association Study (GWAS) to date (9 095 SSc and 17 584 healthy controls with European ancestry). The best-fitting GRS was identified under the additive model in an independent cohort that comprised 400 patients with SSc and 571 controls. Additionally, GRS for clinical subtypes (limited cutaneous SSc and diffuse cutaneous SSc) and serological subtypes (anti-topoisomerase positive (ATA+) and anti-centromere positive (ACA+)) were generated. We combined the estimated GRS with demographic and immunological parameters in a multivariate generalised linear model. RESULTS The best-fitting SSc GRS included 33 single nucleotide polymorphisms (SNPs) and discriminated between patients with SSc and controls (area under the receiver operating characteristic (ROC) curve (AUC)=0.673). Moreover, the GRS differentiated between SSc and other IMIDs, such as rheumatoid arthritis and Sjögren's syndrome. Finally, the combination of GRS with age and immune cell counts significantly increased the performance of the model (AUC=0.787). While the SSc GRS was not able to discriminate between ATA+ and ACA+ patients (AUC<0.5), the serological subtype GRS, which was based on the allelic effects observed for the comparison between ACA+ and ATA+ patients, reached an AUC=0.693. CONCLUSIONS GRS was successfully implemented in SSc. The model discriminated between patients with SSc and controls or other IMIDs, confirming the potential of GRS to support early and differential diagnosis for SSc.
Collapse
MESH Headings
- Adult
- Aged
- Antibodies, Antinuclear/immunology
- Arthritis, Rheumatoid/genetics
- Autoantibodies/immunology
- Case-Control Studies
- DNA Topoisomerases/immunology
- Female
- Genetic Predisposition to Disease
- Genome-Wide Association Study
- Humans
- Linear Models
- Lupus Erythematosus, Systemic/genetics
- Male
- Middle Aged
- Polymorphism, Single Nucleotide
- Risk Factors
- Scleroderma, Diffuse/genetics
- Scleroderma, Diffuse/immunology
- Scleroderma, Limited/genetics
- Scleroderma, Limited/immunology
- Scleroderma, Systemic/genetics
- Scleroderma, Systemic/immunology
- Sjogren's Syndrome/genetics
- White People
Collapse
Affiliation(s)
- Lara Bossini-Castillo
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, Granada, Andalucía, Spain
| | | | - Martin Kerick
- Instituto de Parasitologia y Biomedicina Lopez-Neyra, Granada, Andalucía, Spain
| | | | - Elena López-Isac
- Instituto de Parasitologia y Biomedicina Lopez-Neyra, Granada, Andalucía, Spain
| | - Carmen P Simeón
- Departament of Internal Medicine, Hospital Vall d'Hebron, Barcelona, Catalunya, Spain
| | - Norberto Ortego-Centeno
- Departament of Internal Medicine, Hospital Universitario San Cecilio, Granada, Andalucía, Spain
| | - Shervin Assassi
- Division of Rheumatology and Clinical Immunogenetics, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nicolas Hunzelmann
- Department of Dermatology, University of Cologne, Koln, Nordrhein-Westfalen, Germany
| | - Armando Gabrielli
- Istituto di Clinica Medica Generale, Ematologia ed Immunologia Clinica, Università Politecnica delle Marche, Ancona, Marche, Italy
| | | | - Yannick Allanore
- Rheumatology A Department, Hospital Cochin, Paris, Île-de-France, France
| | - Carmen Fonseca
- Centre for Rheumatology, Royal Free and University College Medical School, London, London, UK
| | - Christopher P Denton
- Centre for Rheumatology, Royal Free and University College Medical School, London, London, UK
| | - Timothy Rdj Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, Utrecht, Netherlands
| | - Marta Eugenia Alarcón-Riquelme
- Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
| | - Lorenzo Beretta
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, La Fondazione IRCCS Ca' Granda Ospedale Maggiore di Milano Policlinico, Milano, Lombardia, Italy
| | - Maureen D Mayes
- Division of Rheumatology and Clinical Immunogenetics, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Javier Martin
- Instituto de Parasitologia y Biomedicina Lopez-Neyra, Granada, Andalucía, Spain
| |
Collapse
|
19
|
Rybakowska P, Burbano C, Van Gassen S, Varela N, Aguilar-Quesada R, Saeys Y, Alarcón-Riquelme ME, Marañón C. Stabilization of Human Whole Blood Samples for Multicenter and Retrospective Immunophenotyping Studies. Cytometry A 2020; 99:524-537. [PMID: 33070416 DOI: 10.1002/cyto.a.24241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/14/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023]
Abstract
Whole blood is often collected for large-scale immune monitoring studies to track changes in cell frequencies and responses using flow (FC) or mass cytometry (MC). In order to preserve sample composition and phenotype, blood samples should be analyzed within 24 h after bleeding, restricting the recruitment, analysis protocols, as well as biobanking. Herein, we have evaluated two whole blood preservation protocols that allow rapid sample processing and long-term stability. Two fixation buffers were used, Phosphoflow Fix and Lyse (BD) and Proteomic Stabilizer (PROT) to fix and freeze whole blood samples for up to 6 months. After analysis by an 8-plex panel by FC and a 26-plex panel by MC, manual gating of circulating leukocyte populations and cytokines was performed. Additionally, we tested the stability of a single sample over a 13-months period using 45 consecutive aliquots and a 34-plex panel by MC. We observed high correlation and low bias toward any cell population when comparing fresh and 6 months frozen blood with FC and MC. This correlation was confirmed by hierarchical clustering. Low coefficients of variation (CV) across studied time points indicate good sample preservation for up to 6 months. Cytokine detection stability was confirmed by low CVs, with some differences between fresh and fixed conditions. Thirteen months regular follow-up of PROT samples showed remarkable sample stability. Whole blood can be preserved for phenotyping and cytokine-response studies provided the careful selection of a compatible antibody panel. However, possible changes in cell morphology, differences in antibody affinity, and changes in cytokine-positive cell frequencies when compared to fresh blood should be considered. Our setting constitutes a valuable tool for multicentric and retrospective studies. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Paulina Rybakowska
- Department of Medical Genomics, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, Spain
| | - Catalina Burbano
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Sofie Van Gassen
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium.,Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
| | - Nieves Varela
- Department of Medical Genomics, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, Spain
| | | | - Yvan Saeys
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium.,Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
| | - Marta E Alarcón-Riquelme
- Department of Medical Genomics, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, Spain.,Unit for Chronic Inflammatory Diseases, Institute for Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Concepción Marañón
- Department of Medical Genomics, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, Spain
| |
Collapse
|
20
|
Espasa A, Torrents S, Morales‐Indiano C, Rico LG, Bardina J, Ancochea A, Bistué‐Rovira À, Linio R, Raya M, Vergara S, Juncà J, Grifols J, Petriz J, Soria M, Sorigue M. Diagnostic performance of the ClearLLab 10C B cell tube. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:519-530. [DOI: 10.1002/cyto.b.21955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Andrea Espasa
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
| | | | - Cristian Morales‐Indiano
- Clinical Laboratory ICS‐Metropolitana Nord, Core‐Hematology Department Hospital Germans Trias i Pujol Badalona Spain
| | - Laura G. Rico
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | - Jorge Bardina
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | - Agueda Ancochea
- Banc de Sang i Teixits Hospital Germans Trias i Pujol Badalona Spain
| | - Àngel Bistué‐Rovira
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | - Rosa Linio
- Banc de Sang i Teixits Hospital Germans Trias i Pujol Badalona Spain
| | - Minerva Raya
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
| | - Sara Vergara
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
| | - Jordi Juncà
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | | | - Jordi Petriz
- Functional Cytomics, Josep Carreras Leukaemia Research Institute (IJC), Universitat Autònoma de Barcelona, Badalona Barcelona Spain
| | | | - Marc Sorigue
- Hematology Laboratory Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Functional cytomics‐IJC, Universitat Autònoma de Barcelona Badalona Spain
| |
Collapse
|
21
|
Beretta L, Barturen G, Vigone B, Bellocchi C, Hunzelmann N, De Langhe E, Cervera R, Gerosa M, Kovács L, Ortega Castro R, Almeida I, Cornec D, Chizzolini C, Pers JO, Makowska Z, Lesche R, Kerick M, Alarcón-Riquelme ME, Martin J. Genome-wide whole blood transcriptome profiling in a large European cohort of systemic sclerosis patients. Ann Rheum Dis 2020; 79:1218-1226. [PMID: 32561607 PMCID: PMC7456554 DOI: 10.1136/annrheumdis-2020-217116] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/30/2020] [Accepted: 05/14/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVES The analysis of annotated transcripts from genome-wide expression studies may help to understand the pathogenesis of complex diseases, such as systemic sclerosis (SSc). We performed a whole blood (WB) transcriptome analysis on RNA collected in the context of the European PRECISESADS project, aiming at characterising the pathways that differentiate SSc from controls and that are reproducible in geographically diverse populations. METHODS Samples from 162 patients and 252 controls were collected in RNA stabilisers. Cases and controls were divided into a discovery (n=79+163; Southern Europe) and validation cohort (n=83+89; Central-Western Europe). RNA sequencing was performed by an Illumina assay. Functional annotations of Reactome pathways were performed with the Functional Analysis of Individual Microarray Expression (FAIME) algorithm. In parallel, immunophenotyping of 28 circulating cell populations was performed. We tested the presence of differentially expressed genes/pathways and the correlation between absolute cell counts and RNA transcripts/FAIME scores in regression models. Results significant in both populations were considered as replicated. RESULTS Overall, 15 224 genes and 1277 functional pathways were available; of these, 99 and 225 were significant in both sets. Among replicated pathways, we found a deregulation in type-I interferon, Toll-like receptor cascade, tumour suppressor p53 protein function, platelet degranulation and activation. RNA transcripts or FAIME scores were jointly correlated with cell subtypes with strong geographical differences; neutrophils were the major determinant of gene expression in SSc-WB samples. CONCLUSIONS We discovered a set of differentially expressed genes/pathways validated in two independent sets of patients with SSc, highlighting a number of deregulated processes that have relevance for the pathogenesis of autoimmunity and SSc.
Collapse
Affiliation(s)
- Lorenzo Beretta
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Guillermo Barturen
- GENYO, Centre for Genomics and Oncological Research Pfizer, University of Granada, Andalusian Regional Government, PTS GRANADA, Granada, Spain
| | - Barbara Vigone
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Chiara Bellocchi
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Nicolas Hunzelmann
- Klinik und Poliklinik für Dermatologie und Venerologie, Uniklinik Köln, Köln, Germany
| | - Ellen De Langhe
- Division of Rheumatology, University Hospitals Leuven and Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Ricard Cervera
- Department of Autoimmune Diseases, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Gerosa
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - László Kovács
- Department of Rheumatology and Immunology, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Rafaela Ortega Castro
- Servicio de Reumatologia, Hospital Universitario Reina Sofía, Instituto Maimónides de Investigación Biomédica IMIBIC, Córdoba, Spain
| | - Isabel Almeida
- Serviço de Imunologia EX-CICAP, Centro Hospitalar e Universitário do Porto, Porto, Portugal
| | - Divi Cornec
- UMR1227, Lymphocytes B et Autoimmunité, Université de Brest, Inserm, Labex IGO, Brest, France
- Rheumatology Department, Cavale Blanche Hospital, Brest, France
| | - Carlo Chizzolini
- Immunology & Allergy, University Hospital and School of Medicine (HCUGE), Geneva, Switzerland
| | - Jacques-Olivier Pers
- UMR1227, Lymphocytes B et Autoimmunité, Université de Brest, Inserm, Labex IGO, Brest, France
| | | | | | - Martin Kerick
- Instituto de Parasitologia y Biomedicina Lopez-Neyra, Granada, Spain
| | - Marta Eugenia Alarcón-Riquelme
- GENYO, Centre for Genomics and Oncological Research Pfizer, University of Granada, Andalusian Regional Government, PTS GRANADA, Granada, Spain
| | - Javier Martin
- Instituto de Parasitologia y Biomedicina Lopez-Neyra, Granada, Spain
| |
Collapse
|
22
|
Standardization procedure for flow cytometry data harmonization in prospective multicenter studies. Sci Rep 2020; 10:11567. [PMID: 32665668 PMCID: PMC7360585 DOI: 10.1038/s41598-020-68468-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 06/22/2020] [Indexed: 01/26/2023] Open
Abstract
One of the most challenging objective for clinical cytometry in prospective multicenter immunomonitoring trials is to compare frequencies, absolute numbers of leukocyte populations and further the mean fluorescence intensities of cell markers, especially when the data are generated from different instruments. Here, we describe an innovative standardization workflow to compare all data to carry out any large-scale, prospective multicentric flow cytometry analysis whatever the duration, the number or type of instruments required for the realization of such projects.
Collapse
|
23
|
Multicentre Harmonisation of a Six-Colour Flow Cytometry Panel for Naïve/Memory T Cell Immunomonitoring. J Immunol Res 2020; 2020:1938704. [PMID: 32322591 PMCID: PMC7153001 DOI: 10.1155/2020/1938704] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/10/2020] [Accepted: 02/26/2020] [Indexed: 01/17/2023] Open
Abstract
Background Personalised medicine in oncology needs standardised immunological assays. Flow cytometry (FCM) methods represent an essential tool for immunomonitoring, and their harmonisation is crucial to obtain comparable data in multicentre clinical trials. The objective of this study was to design a harmonisation workflow able to address the most effective issues contributing to intra- and interoperator variabilities in a multicentre project. Methods The Italian National Institute of Health (Istituto Superiore di Sanità, ISS) managed a multiparametric flow cytometric panel harmonisation among thirteen operators belonging to five clinical and research centres of Lazio region (Italy). The panel was based on a backbone mixture of dried antibodies (anti-CD3, anti-CD4, anti-CD8, anti-CD45RA, and anti-CCR7) to detect naïve/memory T cells, recognised as potential prognostic/predictive immunological biomarkers in cancer immunotherapies. The coordinating centre distributed frozen peripheral blood mononuclear cells (PBMCs) and fresh whole blood (WB) samples from healthy donors, reagents, and Standard Operating Procedures (SOPs) to participants who performed experiments by their own equipment, in order to mimic a real-life scenario. Operators returned raw and locally analysed data to ISS for central analysis and statistical elaboration. Results Harmonised and reproducible results were obtained by sharing experimental set-up and procedures along with centralising data analysis, leading to a reduction of cross-centre variability for naïve/memory subset frequencies particularly in the whole blood setting. Conclusion Our experimental and analytical working process proved to be suitable for the harmonisation of FCM assays in a multicentre setting, where high-quality data are required to evaluate potential immunological markers, which may contribute to select better therapeutic options.
Collapse
|
24
|
Rybakowska P, Alarcón-Riquelme ME, Marañón C. Key steps and methods in the experimental design and data analysis of highly multi-parametric flow and mass cytometry. Comput Struct Biotechnol J 2020; 18:874-886. [PMID: 32322369 PMCID: PMC7163213 DOI: 10.1016/j.csbj.2020.03.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 01/05/2023] Open
Abstract
High-dimensional, single-cell cell technologies revolutionized the way to study biological systems, and polychromatic flow cytometry (FC) and mass cytometry (MC) are two of the drivers of this revolution. As up to 30-50 dimensions respectively can be measured per single-cell, they allow deep phenotyping combined with cellular functions studies, like cytokine production or protein phosphorylation. In parallel, the bioinformatics field develops algorithms that are able to process incoming data and extract the most useful and meaningful biological information. However, the success of automated analysis tools depends on the generation of high-quality data. In this review we present the most recent FC and MC computational approaches that are used to prepare, process and interpret high-content cytometry data. We also underscore proper experimental design as a key step for obtaining good quality data.
Collapse
Affiliation(s)
- Paulina Rybakowska
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Spain
| | - Marta E. Alarcón-Riquelme
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Spain
- Institute for Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Concepción Marañón
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Spain
| |
Collapse
|
25
|
Bourgoin P, Lediagon G, Arnoux I, Bernot D, Morange PE, Michelet P, Malergue F, Markarian T. Flow cytometry evaluation of infection-related biomarkers in febrile subjects in the emergency department. Future Microbiol 2020; 15:189-201. [PMID: 32065550 DOI: 10.2217/fmb-2019-0256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: In an Emergency Department (ED), the etiological identification of infected subjects is essential. 13 infection-related biomarkers were assessed using a new flow cytometry procedure. Materials & methods: If subjects presented with febrile symptoms at the ED, 13 biomarkers' levels, including CD64 on neutrophils (nCD64) and CD169 on monocytes (mCD169), were tested and compared with clinical records. Results: Among 50 subjects, 78% had bacterial infections and 8% had viral infections. nCD64 showed 82% sensitivity and 91% specificity for identifying subjects with bacterial infections. mCD169, HLA-ABC ratio and HLA-DR on monocytes had high values in subjects with viral infections. Conclusion: Biomarkers showed promising performances to improve the ED's infectious stratification.
Collapse
Affiliation(s)
- Pénélope Bourgoin
- Department of Research & Development, Beckman Coulter Life Sciences-Immunotech, 130 Avenue de Lattre de Tassigny, 13009 Marseille, France.,C2VN INSERM-INRA, Aix-Marseille University, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | - Guillaume Lediagon
- Adult Emergency Unit, La Timone Hospital, APHM, 264 Rue Saint Pierre, 13385 Marseille, France
| | - Isabelle Arnoux
- Hematology Laboratory, La Timone Hospital, APHM, 264 Rue Saint Pierre, 13385 Marseille, France
| | - Denis Bernot
- Hematology Laboratory, La Timone Hospital, APHM, 264 Rue Saint Pierre, 13385 Marseille, France
| | - Pierre-Emmanuel Morange
- C2VN INSERM-INRA, Aix-Marseille University, 27 Boulevard Jean Moulin, 13385 Marseille, France.,Hematology Laboratory, La Timone Hospital, APHM, 264 Rue Saint Pierre, 13385 Marseille, France
| | - Pierre Michelet
- Adult Emergency Unit, La Timone Hospital, APHM, 264 Rue Saint Pierre, 13385 Marseille, France
| | - Fabrice Malergue
- Department of Research & Development, Beckman Coulter Life Sciences-Immunotech, 130 Avenue de Lattre de Tassigny, 13009 Marseille, France
| | - Thibaut Markarian
- Adult Emergency Unit, La Timone Hospital, APHM, 264 Rue Saint Pierre, 13385 Marseille, France
| |
Collapse
|
26
|
Kalina T. Reproducibility of Flow Cytometry Through Standardization: Opportunities and Challenges. Cytometry A 2019; 97:137-147. [DOI: 10.1002/cyto.a.23901] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 09/04/2019] [Accepted: 09/11/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Tomas Kalina
- CLIP‐Childhood Leukemia Investigation Prague, Department of Pediatric Hematology and Oncology2nd Medical School, Charles University and University Hospital Motol Prague Czech Republic
| |
Collapse
|
27
|
Abstract
Autoimmune rheumatic diseases pose many problems that have, in general, already been solved in the field of cancer. The heterogeneity of each disease, the clinical similarities and differences between different autoimmune rheumatic diseases and the large number of patients that remain without a diagnosis underline the need to reclassify these diseases via new approaches. Knowledge about the molecular basis of systemic autoimmune diseases, along with the availability of bioinformatics tools capable of handling and integrating large volumes of various types of molecular data at once, offer the possibility of reclassifying these diseases. A new taxonomy could lead to the discovery of new biomarkers for patient stratification and prognosis. Most importantly, this taxonomy might enable important changes in clinical trial design to reach the expected outcomes or the design of molecularly targeted therapies. In this Review, we discuss the basis for a new molecular taxonomy for autoimmune rheumatic diseases. We highlight the evidence surrounding the idea that these diseases share molecular features related to their pathogenesis and development and discuss previous attempts to classify these diseases. We evaluate the tools available to analyse and combine different types of molecular data. Finally, we introduce PRECISESADS, a project aimed at reclassifying the systemic autoimmune diseases.
Collapse
|
28
|
Immunophenotyping Using Dried and Lyophilized Reagents. Methods Mol Biol 2019; 2032:69-79. [PMID: 31522413 DOI: 10.1007/978-1-4939-9650-6_4] [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: 01/11/2023]
Abstract
Antibody reagents that are used for flow cytometry immunophenotyping have traditionally been prepared by combining individual liquid antibody conjugates into mixtures. These cocktails have limited shelf-life, and their preparation is time-consuming and prone to laboratory error. Manufacturers of these reagents, in collaboration with several clinical and research centers, have made advances in constructing dried antibody cocktails which have addressed many of the problems inherent in preparing the liquid cocktails on the lab bench. This chapter discusses methods for the use of dried reagents.
Collapse
|
29
|
Pitoiset F, Cassard L, El Soufi K, Boselli L, Grivel J, Roux A, Klatzmann D, Chaput N, Rosenzwajg M. Deep phenotyping of immune cell populations by optimized and standardized flow cytometry analyses. Cytometry A 2018; 93:793-802. [DOI: 10.1002/cyto.a.23570] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 06/28/2018] [Accepted: 07/02/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Fabien Pitoiset
- Sorbonne Universités; UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3); F-75005 Paris France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B); Hôpital Pitié-Salpêtrière, AP-HP; F-75651 Paris France
| | - Lydie Cassard
- Laboratory of Immunomonitoring in Oncology; Gustave Roussy Cancer Campus, CNRS-UMS 3655 and INSERM-US23; Villejuif F-94805 France
| | - Karim El Soufi
- Sorbonne Universités; UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3); F-75005 Paris France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B); Hôpital Pitié-Salpêtrière, AP-HP; F-75651 Paris France
| | - Lisa Boselli
- Laboratory of Immunomonitoring in Oncology; Gustave Roussy Cancer Campus, CNRS-UMS 3655 and INSERM-US23; Villejuif F-94805 France
| | - Jonathan Grivel
- Laboratory of Immunomonitoring in Oncology; Gustave Roussy Cancer Campus, CNRS-UMS 3655 and INSERM-US23; Villejuif F-94805 France
| | - Alexandra Roux
- Sorbonne Universités; UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3); F-75005 Paris France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B); Hôpital Pitié-Salpêtrière, AP-HP; F-75651 Paris France
| | - David Klatzmann
- Sorbonne Universités; UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3); F-75005 Paris France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B); Hôpital Pitié-Salpêtrière, AP-HP; F-75651 Paris France
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology; Gustave Roussy Cancer Campus, CNRS-UMS 3655 and INSERM-US23; Villejuif F-94805 France
- Faculty of Pharmacy, University Paris-Sud; Chatenay-Malabry F-92296 France
| | - Michelle Rosenzwajg
- Sorbonne Universités; UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3); F-75005 Paris France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B); Hôpital Pitié-Salpêtrière, AP-HP; F-75651 Paris France
| |
Collapse
|
30
|
Schreiber K, Nocturne G, Cornec D, Daïen CI. Lymphocytes as Biomarkers of Therapeutic Response in Rheumatic Autoimmune Diseases, Is It a Realistic Goal? Clin Rev Allergy Immunol 2018; 53:277-290. [PMID: 28560621 DOI: 10.1007/s12016-017-8614-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Many therapies are available for patients with rheumatoid arthritis (RA) while biological therapies have limited effects in patients with systemic lupus erythematosus (SLE) and primary Sjögren's syndrome (pSS). In both cases, biomarkers predicting drug response would be very useful to guide clinicians in their choice. We performed a systematic review to evaluate the value of lymphocyte phenotyping as a marker of therapeutic response. Of the 1063 articles retrieved, 39 fulfilled inclusion criteria and were included in the present review (25 for RA, 10 for SLE, and 4 for pSS). Lymphocyte phenotyping was described as a biomarker of therapeutic response in many studies, but most results could not be confirmed by independent teams using multivariate analysis. The most consistent result might be the association between rituximab response and the levels of memory B cells before therapy, although some studies were controversial. Thus, lymphocyte phenotyping cannot yet be proposed as a biomarker of response in rheumatic autoimmune diseases. The lack of reproducibility between studies may be explained by technical issues influencing lymphocyte phenotyping so standardization procedures should be developed for future studies. The patients' characteristics vary between studies, and large population studies, including a wide range of patients' characteristics and biomarkers, are required to provide predictive models for clinical outcomes. The use of new flow cytometry techniques such as single-cell mass cytometry technology might also help finder reliable biomarkers in the future.
Collapse
Affiliation(s)
- Kristina Schreiber
- Rheumatology Department, Lapeyronie Hospital and Montpellier I University, Montpellier, France
| | | | | | - Claire I Daïen
- Rheumatology Department, Lapeyronie Hospital and Montpellier I University, Montpellier, France.
- Institute of Molecular Genetic, UMR 5535, CNRS, Montpellier, France.
| |
Collapse
|
31
|
Carvajal Alegria G, Gazeau P, Hillion S, Daïen CI, Cornec DYK. Could Lymphocyte Profiling be Useful to Diagnose Systemic Autoimmune Diseases? Clin Rev Allergy Immunol 2018; 53:219-236. [PMID: 28474288 DOI: 10.1007/s12016-017-8608-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Considering the implications of B, T, and natural killer (NK) cells in the pathophysiology of systemic autoimmune diseases, the assessment of their distribution in the blood could be helpful for physicians in the complex process of determining a precise diagnosis. In primary Sjögren's syndrome, transitional and active naive B cells are increased and memory B cells are decreased compared to healthy controls and other systemic diseases. However, their utility to improve the accuracy of classification criteria has not been proven. In early untreated rheumatoid arthritis, proportions of regulatory T cells are constantly reduced, but other patterns are difficult to determine given the heterogeneity of published studies. In systemic lupus erythematosus, the lack of studies using large cohorts of patients and the diversity of the possible pathological mechanisms involved are also important impediments. Nevertheless, transitional B cell and plasma cell proportions are increased in most of the studies, the CD4/CD8 ratio is decreased, and the number of NK cells is reduced. Despite the low number of studies, anomalies of lymphocyte subset distribution was also described in ANCA-associated vasculitis, systemic scleroderma, and myositis. For now, flow cytometric analysis of lymphocyte subsets has focused mainly on specific subpopulations and is more useful for basic and translational research than for diagnostics in clinical practice. However, new modern methods such as mass cytometry and bioinformatics analyses may offer the possibility to simultaneously account for the relative proportions of multiple lymphocyte subsets and define a global profile in homogeneous groups of patients. The years to come will certainly incorporate such global lymphocyte profiling in reclassification of systemic autoimmune diseases.
Collapse
Affiliation(s)
- Guillermo Carvajal Alegria
- Service de Rhumatologie, Hôpital de la Cavale Blanche, CHRU Brest, BP 824, 29609, Brest cedex, France.,INSERM U1227, European University of Brest, Brest, France
| | - Pierre Gazeau
- Service de Rhumatologie, Hôpital de la Cavale Blanche, CHRU Brest, BP 824, 29609, Brest cedex, France
| | - Sophie Hillion
- INSERM U1227, European University of Brest, Brest, France.,Laboratoire d'Immunologie et Immunothérapie, CHRU Morvan, Brest, France
| | - Claire I Daïen
- Rheumatology Department, Lapeyronie Hospital and Montpellier I University, Montpellier, France.,UMR5535, CNRS, Institute of molecular genetic, Montpellier, France
| | - Divi Y K Cornec
- Service de Rhumatologie, Hôpital de la Cavale Blanche, CHRU Brest, BP 824, 29609, Brest cedex, France. .,INSERM U1227, European University of Brest, Brest, France.
| |
Collapse
|
32
|
Pitoiset F, Barbié M, Monneret G, Braudeau C, Pochard P, Pellegrin I, Trauet J, Labalette M, Klatzmann D, Rosenzwajg M. A standardized flow cytometry procedure for the monitoring of regulatory T cells in clinical trials. CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 94:621-626. [PMID: 29316248 DOI: 10.1002/cyto.b.21622] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/21/2017] [Accepted: 01/03/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Quantification of regulatory T cells (Tregs) is crucial in immunomonitoring in clinical trials as this cell population has been shown to be involved in a wide range of diseases, including cancers, autoimmune diseases, infections, and allergies. Human Tregs are defined as CD4+ CD25+ CD127low FoxP3+ cells, and the standardization of their staining by flow cytometry is a challenge, especially in multicenter clinical trials, notably because of the intracellular location of FoxP3. METHOD A flow cytometry staining procedure was settled and standardized to measure human Tregs in peripheral whole blood using precoated dried antibodies in ready-to-use tubes. It was compared with reference methods and implemented and validated to be suitable with different cytometer platforms. RESULTS The standardized protocol developed with dried antibodies and reduced volumes of whole blood allows an optimal identification of Tregs. Compared with classical staining procedure, it reduces the number of steps required, in a very fast and simple technique. The accuracy of the method was confirmed by a multicenter comparison with different cytometer brands. CONCLUSIONS Our results highlight the reliability of this high-standard protocol that could become a reference method for the monitoring of Tregs in clinical trials. © 2018 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Fabien Pitoiset
- Sorbonne Université, UPMC Univ Paris 06, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris F-75005, France.,Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (I2B), AP-HP, Hôpital Pitié-Salpêtrière, Paris, F-75651, France
| | - Michèle Barbié
- Sorbonne Université, UPMC Univ Paris 06, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris F-75005, France.,Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (I2B), AP-HP, Hôpital Pitié-Salpêtrière, Paris, F-75651, France
| | - Guillaume Monneret
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Laboratoire d'Immunologie, Lyon F-69003, France
| | - Cécile Braudeau
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France.,Centre de Recherche en Transplantation et Immunologie (UMR1064), INSERM Université de Nantes, Nantes, France
| | - Pierre Pochard
- INSERM ERI29, EA2216, Université de Brest, Labex IGO, CHRU Morvan, Brest, France
| | - Isabelle Pellegrin
- Service d'Immunologie-Immunogénétique, Pôle de Biologie et Pathologie, Groupe Hospitalier Pellegrin, CHU Bordeaux, Bordeaux, France
| | - Jacques Trauet
- University of Lille, U995 - LIRIC-Lille Inflammation Research International Center, Lille F-59000, France.,INSERM, U995, Lille F-59000, France.,CHU LILLE, Institut d'Immunologie, Lille F-59000, France
| | - Myriam Labalette
- University of Lille, U995 - LIRIC-Lille Inflammation Research International Center, Lille F-59000, France.,INSERM, U995, Lille F-59000, France.,CHU LILLE, Institut d'Immunologie, Lille F-59000, France
| | - David Klatzmann
- Sorbonne Université, UPMC Univ Paris 06, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris F-75005, France.,Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (I2B), AP-HP, Hôpital Pitié-Salpêtrière, Paris, F-75651, France
| | - Michelle Rosenzwajg
- Sorbonne Université, UPMC Univ Paris 06, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris F-75005, France.,Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (I2B), AP-HP, Hôpital Pitié-Salpêtrière, Paris, F-75651, France
| |
Collapse
|
33
|
Barteneva NS, Vorobjev IA. Heterogeneity of Metazoan Cells and Beyond: To Integrative Analysis of Cellular Populations at Single-Cell Level. Methods Mol Biol 2018; 1745:3-23. [PMID: 29476460 DOI: 10.1007/978-1-4939-7680-5_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this paper, we review some of the recent advances in cellular heterogeneity and single-cell analysis methods. In modern research of cellular heterogeneity, there are four major approaches: analysis of pooled samples, single-cell analysis, high-throughput single-cell analysis, and lately integrated analysis of cellular population at a single-cell level. Recently developed high-throughput single-cell genetic analysis methods such as RNA-Seq require purification step and destruction of an analyzed cell often are providing a snapshot of the investigated cell without spatiotemporal context. Correlative analysis of multiparameter morphological, functional, and molecular information is important for differentiation of more uniform groups in the spectrum of different cell types. Simplified distributions (histograms and 2D plots) can underrepresent biologically significant subpopulations. Future directions may include the development of nondestructive methods for dissecting molecular events in intact cells, simultaneous correlative cellular analysis of phenotypic and molecular features by hybrid technologies such as imaging flow cytometry, and further progress in supervised and non-supervised statistical analysis algorithms.
Collapse
Affiliation(s)
- Natasha S Barteneva
- PCMM-Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.
- Department of Biology, School of Sciences and Technology, Nazarbayev University, Astana, Kazakhstan.
| | - Ivan A Vorobjev
- Department of Biology, School of Sciences and Technology, Nazarbayev University, Astana, Kazakhstan
| |
Collapse
|
34
|
Immunophenotyping As a New Tool for Classification and Monitoring of Systemic Autoimmune Diseases. Clin Rev Allergy Immunol 2017; 53:177-180. [DOI: 10.1007/s12016-017-8604-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
35
|
Roy NS, Wei Y, Kuklinski E, Asbell PA. The Growing Need for Validated Biomarkers and Endpoints for Dry Eye Clinical Research. Invest Ophthalmol Vis Sci 2017; 58:BIO1-BIO19. [PMID: 28475698 PMCID: PMC5455411 DOI: 10.1167/iovs.17-21709] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/04/2017] [Indexed: 11/30/2022] Open
Abstract
Purpose Biomarkers with minimally invasive and reproducible objective metrics provide the key to future paradigm shifts in understanding of the underlying causes of dry eye disease (DED) and approaches to treatment of DED. We review biomarkers and their validity in providing objective metrics for DED clinical research and patient care. Methods The English-language literature in PubMed primarily over the last decade was surveyed for studies related to identification of biomarkers of DED: (1) inflammation, (2) point-of-care, (3) ocular imaging, and (4) genetics. Relevant studies in each group were individually evaluated for (1) methodological and analytical details, (2) data and concordance with other similar studies, and (3) potential to serve as validated biomarkers with objective metrics. Results Significant work has been done to identify biomarkers for DED clinical trials and for patient care. Interstudy variation among studies dealing with the same biomarker type was high. This could be attributed to biologic variations and/or differences in processing, and data analysis. Correlation with other signs and symptoms of DED was not always clear or present. Conclusions Many of the biomarkers reviewed show the potential to serve as validated and objective metrics for clinical research and patient care in DED. Interstudy variation for a given biomarker emphasizes the need for detailed reporting of study methodology, including information on subject characteristics, quality control, processing, and analysis methods to optimize development of nonsubjective metrics. Biomarker development offers a rich opportunity to significantly move forward clinical research and patient care in DED. Overview DED is an unmet medical need - a chronic pain syndrome associated with variable vision that affects quality of life, is common with advancing age, interferes with the comfortable use of contact lenses, and can diminish results of eye surgeries, such as cataract extraction, LASIK, and glaucoma procedures. It is a worldwide medical challenge with a prevalence rate ranging from 8% to 50%. Many clinicians and researchers across the globe are searching for better answers to understand the mechanisms related to the development and chronicity of DED. Though there have been many clinical trials for DED, few new treatments have emerged over the last decade. Biomarkers may provide the needed breakthrough to propel our understanding of DED to the next level and the potential to realize our goal of truly personalized medicine based on scientific evidence. Clinical trials and research on DED have suffered from the lack of validated biomarkers and less than objective and reproducible endpoints. Current work on biomarkers has provided the groundwork to move forward. This review highlights primarily ocular biomarkers that have been investigated for use in DED, discusses the methodologic outcomes in providing objective metrics for clinical research, and suggests recommendations for further work.
Collapse
Affiliation(s)
- Neeta S. Roy
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Yi Wei
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Eric Kuklinski
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Penny A. Asbell
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| |
Collapse
|