51
|
Liang X, Li P, Jiang J, Xin J, Luo J, Li J, Chen P, Ren K, Zhou Q, Guo B, Zhou X, Chen J, He L, Yang H, Hu W, Ma S, Li B, Chen X, Shi D, Li J, Chinese Group on the Study of Severe Hepatitis B (COSSH). Transcriptomics unveils immune metabolic disruption and a novel biomarker of mortality in patients with HBV-related acute-on-chronic liver failure. JHEP Rep 2023; 5:100848. [PMID: 37583946 PMCID: PMC10424217 DOI: 10.1016/j.jhepr.2023.100848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 06/16/2023] [Accepted: 07/01/2023] [Indexed: 08/17/2023] Open
Abstract
Background & Aims HBV-related acute-on-chronic liver failure (HBV-ACLF) is a complex syndrome associated with high short-term mortality. This study aims to reveal the molecular basis and identify novel HBV-ACLF biomarkers. Methods Seventy patients with HBV-ACLF and different short-term (28 days) outcomes underwent transcriptome sequencing using peripheral blood mononuclear cells. Candidate biomarkers were confirmed in two external cohorts using ELISA. Results Cellular composition analysis with peripheral blood mononuclear cell transcriptomics showed that the proportions of monocytes, T cells and natural killer cells were significantly correlated with 28-day mortality. Significant metabolic dysregulation of carbohydrate, energy and amino acid metabolism was observed in ACLF non-survivors. V-set and immunoglobulin domain-containing 4 (VSIG4) was the most robust predictor of patient survival (adjusted p = 1.74 × 10-16; variable importance in the projection = 1.21; AUROC = 0.89) and was significantly correlated with pathways involved in the progression of ACLF, including inflammation, oxidative phosphorylation, tricarboxylic acid cycle and T-cell activation/differentiation. Plasma VSIG4 analysis externally validated its diagnostic value in ACLF (compared with chronic liver disease and healthy groups, AUROC = 0.983). The prognostic performance for 28-/90-day mortality (AUROCs = 0.769/0.767) was comparable to that of three commonly used scores (COSSH-ACLFs, 0.867/0.884; CLIF-C ACLFs, 0.840/0.835; MELD-Na, 0.710/0.737). Plasma VSIG4 level, as an independent predictor, could be used to improve the prognostic performance of clinical scores. Risk stratification based on VSIG4 expression levels (>122 μg/ml) identified patients with ACLF at a high risk of death. The generality of VSIG4 in other etiologies was validated. Conclusions This study reveals that immune-metabolism disorder underlies poor ACLF outcomes. VSIG4 may be helpful as a diagnostic and prognostic biomarker in clinical practice. Impact and implications Acute-on-chronic liver failure (ACLF) is a lethal clinical syndrome associated with high mortality. We found significant immune cell alterations and metabolic dysregulation that were linked to high mortality in patients with HBV-ACLF based on transcriptomics using peripheral blood mononuclear cells. We identified VSIG4 (V-set and immunoglobulin domain-containing 4) as a diagnostic and prognostic biomarker in ACLF, which could specifically identify patients with ACLF at a high risk of death.
Collapse
Affiliation(s)
- Xi Liang
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Peng Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiaojiao Xin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinjin Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiaqi Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Pengcheng Chen
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics and Information Engineering, Taizhou University, Taizhou, China
| | - Keke Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qian Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Beibei Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xingping Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiaxian Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lulu He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Hui Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wen Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shiwen Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Bingqi Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Chen
- Institute of Pharmaceutical Biotechnology and the First Affiliated Hospital, Department of Radiation Oncology, Zhejiang University School of Medicine, Hangzhou, China
- Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, China
| | - Dongyan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jun Li
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chinese Group on the Study of Severe Hepatitis B (COSSH)
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics and Information Engineering, Taizhou University, Taizhou, China
- Institute of Pharmaceutical Biotechnology and the First Affiliated Hospital, Department of Radiation Oncology, Zhejiang University School of Medicine, Hangzhou, China
- Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, China
| |
Collapse
|
52
|
Huang H, Li X, Zha D, Lin H, Yang L, Wang Y, Xu L, Wang L, Lei T, Zhou Z, Xiao YF, Xin HB, Fu M, Qian Y. SARS-CoV-2 E protein-induced THP-1 pyroptosis is reversed by Ruscogenin. Biochem Cell Biol 2023; 101:303-312. [PMID: 36927169 DOI: 10.1139/bcb-2022-0359] [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] [Indexed: 03/18/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an emerging pathogenic coronavirus, has been reported to cause excessive inflammation and dysfunction in multiple cells and organs, but the underlying mechanisms remain largely unknown. Here we showed exogenous addition of SARS-CoV-2 envelop protein (E protein) potently induced cell death in cultured cell lines, including THP-1 monocytic leukemia cells, endothelial cells, and bronchial epithelial cells, in a time- and concentration-dependent manner. SARS-CoV-2 E protein caused pyroptosis-like cell death in THP-1 and led to GSDMD cleavage. In addition, SARS-CoV-2 E protein upregulated the expression of multiple pro-inflammatory cytokines that may be attributed to activation of NF-κB, JNK and p38 signal pathways. Notably, we identified a natural compound, Ruscogenin, effectively reversed E protein-induced THP-1 death via inhibition of NLRP3 activation and GSDMD cleavage. In conclusion, these findings suggested that Ruscogenin may have beneficial effects on preventing SARS-CoV-2 E protein-induced cell death and might be a promising treatment for the complications of COVID-19.
Collapse
Affiliation(s)
- Houda Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Xiuzhen Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Duoduo Zha
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Hongru Lin
- Department of Scientific Research, Hainan General Hospital, Haikou, 570311, China
| | - Lingyi Yang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Yihan Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Luyan Xu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Linsiqi Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Tianhua Lei
- Shock/Trauma Research Center, Department of Biomedical Sciences, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Zhou Zhou
- Shock/Trauma Research Center, Department of Biomedical Sciences, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Yun-Fei Xiao
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Hong-Bo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Mingui Fu
- Shock/Trauma Research Center, Department of Biomedical Sciences, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Yisong Qian
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| |
Collapse
|
53
|
Tsioumpekou M, Krijgsman D, Leusen JHW, Olofsen PA. The Role of Cytokines in Neutrophil Development, Tissue Homing, Function and Plasticity in Health and Disease. Cells 2023; 12:1981. [PMID: 37566060 PMCID: PMC10417597 DOI: 10.3390/cells12151981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023] Open
Abstract
Neutrophils are crucial innate immune cells and comprise 50-70% of the white blood cell population under homeostatic conditions. Upon infection and in cancer, blood neutrophil numbers significantly increase because of the secretion of various chemo- and cytokines by, e.g., leukocytes, pericytes, fibroblasts and endothelial cells present in the inflamed tissue or in the tumor microenvironment (TME). The function of neutrophils in cancer has recently gained considerable attention, as they can exert both pro- and anti-tumorigenic functions, dependent on the cytokine milieu present in the TME. Here, we review the effect of cytokines on neutrophil development, tissue homing, function and plasticity in cancer and autoimmune diseases as well as under physiological conditions in the bone marrow, bloodstream and various organs like the spleen, kidney, liver, lung and lymph nodes. In addition, we address several promising therapeutic options, such as cytokine therapy, immunocytokines and immunotherapy, which aim to exploit the anti-tumorigenic potential of neutrophils in cancer treatment or block excessive neutrophil-mediated inflammation in autoimmune diseases.
Collapse
Affiliation(s)
- Maria Tsioumpekou
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (M.T.); (D.K.); (J.H.W.L.)
| | - Daniëlle Krijgsman
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (M.T.); (D.K.); (J.H.W.L.)
- Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jeanette H. W. Leusen
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (M.T.); (D.K.); (J.H.W.L.)
| | - Patricia A. Olofsen
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (M.T.); (D.K.); (J.H.W.L.)
| |
Collapse
|
54
|
Cambier S, Beretta F, Pörtner N, Metzemaekers M, de Carvalho AC, Martens E, Kaes J, Aelbrecht C, Jacobs C, Van Mol P, Wauters E, Meersseman P, Hermans G, Marques RE, Vanaudenaerde B, Vos R, Wauters J, Gouwy M, Proost P. Proteolytic inactivation of CXCL12 in the lungs and circulation of COVID-19 patients. Cell Mol Life Sci 2023; 80:234. [PMID: 37505242 PMCID: PMC11073220 DOI: 10.1007/s00018-023-04870-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/29/2023]
Abstract
The human chemokine stromal cell-derived factor-1 (SDF-1) or CXCL12 is involved in several homeostatic processes and pathologies through interaction with its cognate G protein-coupled receptor CXCR4. Recent research has shown that CXCL12 is present in the lungs and circulation of patients with coronavirus disease 2019 (COVID-19). However, the question whether the detected CXCL12 is bioactive was not addressed. Indeed, the activity of CXCL12 is regulated by NH2- and COOH-terminal post-translational proteolysis, which significantly impairs its biological activity. The aim of the present study was to characterize proteolytic processing of CXCL12 in broncho-alveolar lavage (BAL) fluid and blood plasma samples from critically ill COVID-19 patients. Therefore, we optimized immunosorbent tandem mass spectrometry proteoform analysis (ISTAMPA) for detection of CXCL12 proteoforms. In patient samples, this approach uncovered that CXCL12 is rapidly processed by site-specific NH2- and COOH-terminal proteolysis and ultimately degraded. This proteolytic inactivation occurred more rapidly in COVID-19 plasma than in COVID-19 BAL fluids, whereas BAL fluid samples from stable lung transplantation patients and the non-affected lung of lung cancer patients (control groups) hardly induced any processing of CXCL12. In COVID-19 BAL fluids with high proteolytic activity, processing occurred exclusively NH2-terminally and was predominantly mediated by neutrophil elastase. In low proteolytic activity BAL fluid and plasma samples, NH2- and COOH-terminal proteolysis by CD26 and carboxypeptidases were observed. Finally, protease inhibitors already approved for clinical use such as sitagliptin and sivelestat prevented CXCL12 processing and may therefore be of pharmacological interest to prolong CXCL12 half-life and biological activity in vivo.
Collapse
Affiliation(s)
- Seppe Cambier
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Rega - Herestraat 49, Box 1042, 3000, Leuven, Belgium
| | - Fabio Beretta
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Rega - Herestraat 49, Box 1042, 3000, Leuven, Belgium
| | - Noëmie Pörtner
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Rega - Herestraat 49, Box 1042, 3000, Leuven, Belgium
| | - Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Rega - Herestraat 49, Box 1042, 3000, Leuven, Belgium
| | - Ana Carolina de Carvalho
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Rega - Herestraat 49, Box 1042, 3000, Leuven, Belgium
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Department of Genetics, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Erik Martens
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Celine Aelbrecht
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Cato Jacobs
- Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Pierre Van Mol
- Laboratory of Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Els Wauters
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Philippe Meersseman
- Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Greet Hermans
- Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Bart Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Joost Wauters
- Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Rega - Herestraat 49, Box 1042, 3000, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Rega - Herestraat 49, Box 1042, 3000, Leuven, Belgium.
| |
Collapse
|
55
|
Park SY, Ter-Saakyan S, Faraci G, Lee HY. Immune cell identifier and classifier (ImmunIC) for single cell transcriptomic readouts. Sci Rep 2023; 13:12093. [PMID: 37495649 PMCID: PMC10372073 DOI: 10.1038/s41598-023-39282-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 07/22/2023] [Indexed: 07/28/2023] Open
Abstract
Single cell RNA sequencing has a central role in immune profiling, identifying specific immune cells as disease markers and suggesting therapeutic target genes of immune cells. Immune cell-type annotation from single cell transcriptomics is in high demand for dissecting complex immune signatures from multicellular blood and organ samples. However, accurate cell type assignment from single-cell RNA sequencing data alone is complicated by a high level of gene expression heterogeneity. Many computational methods have been developed to respond to this challenge, but immune cell annotation accuracy is not highly desirable. We present ImmunIC, a simple and robust tool for immune cell identification and classification by combining marker genes with a machine learning method. With over two million immune cells and half-million non-immune cells from 66 single cell RNA sequencing studies, ImmunIC shows 98% accuracy in the identification of immune cells. ImmunIC outperforms existing immune cell classifiers, categorizing into ten immune cell types with 92% accuracy. We determine peripheral blood mononuclear cell compositions of severe COVID-19 cases and healthy controls using previously published single cell transcriptomic data, permitting the identification of immune cell-type specific differential pathways. Our publicly available tool can maximize the utility of single cell RNA profiling by functioning as a stand-alone bioinformatic cell sorter, advancing cell-type specific immune profiling for the discovery of disease-specific immune signatures and therapeutic targets.
Collapse
Affiliation(s)
- Sung Yong Park
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Sonia Ter-Saakyan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Gina Faraci
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Ha Youn Lee
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
56
|
Zhou T, Gilliam NJ, Li S, Spandau S, Osborn RM, Connor S, Anderson CS, Mariani TJ, Thakar J, Dewhurst S, Mathews DH, Huang L, Sun Y. Generation and Functional Analysis of Defective Viral Genomes during SARS-CoV-2 Infection. mBio 2023; 14:e0025023. [PMID: 37074178 PMCID: PMC10294654 DOI: 10.1128/mbio.00250-23] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/28/2023] [Indexed: 04/20/2023] Open
Abstract
Defective viral genomes (DVGs) have been identified in many RNA viruses as a major factor influencing antiviral immune response and viral pathogenesis. However, the generation and function of DVGs in SARS-CoV-2 infection are less known. In this study, we elucidated DVG generation in SARS-CoV-2 and its relationship with host antiviral immune response. We observed DVGs ubiquitously from transcriptome sequencing (RNA-seq) data sets of in vitro infections and autopsy lung tissues of COVID-19 patients. Four genomic hot spots were identified for DVG recombination, and RNA secondary structures were suggested to mediate DVG formation. Functionally, bulk and single-cell RNA-seq analysis indicated the interferon (IFN) stimulation of SARS-CoV-2 DVGs. We further applied our criteria to the next-generation sequencing (NGS) data set from a published cohort study and observed a significantly higher amount and frequency of DVG in symptomatic patients than those in asymptomatic patients. Finally, we observed exceptionally diverse DVG populations in one immunosuppressive patient up to 140 days after the first positive test of COVID-19, suggesting for the first time an association between DVGs and persistent viral infections in SARS-CoV-2. Together, our findings strongly suggest a critical role of DVGs in modulating host IFN responses and symptom development, calling for further inquiry into the mechanisms of DVG generation and into how DVGs modulate host responses and infection outcome during SARS-CoV-2 infection. IMPORTANCE Defective viral genomes (DVGs) are generated ubiquitously in many RNA viruses, including SARS-CoV-2. Their interference activity to full-length viruses and IFN stimulation provide the potential for them to be used in novel antiviral therapies and vaccine development. SARS-CoV-2 DVGs are generated through the recombination of two discontinuous genomic fragments by viral polymerase complex, and this recombination is also one of the major mechanisms for the emergence of new coronaviruses. Focusing on the generation and function of SARS-CoV-2 DVGs, these studies identify new hot spots for nonhomologous recombination and strongly suggest that the secondary structures within viral genomes mediate the recombination. Furthermore, these studies provide the first evidence for IFN stimulation activity of de novo DVGs during natural SARS-CoV-2 infection. These findings set up the foundation for further mechanism studies of SARS-CoV-2 recombination and provide evidence to harness the immunostimulatory potential of DVGs in the development of a vaccine and antivirals for SARS-CoV-2.
Collapse
Affiliation(s)
- Terry Zhou
- Department of Immunology and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Nora J. Gilliam
- Department of Immunology and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Translational Biomedical Sciences PhD Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Sizhen Li
- School of Electrical Engineering & Computer Science, Oregon State University, Corvallis, Oregon, USA
| | - Simone Spandau
- Department of Immunology and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Raven M. Osborn
- Department of Immunology and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
- Translational Biomedical Sciences PhD Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Sarah Connor
- Department of Pediatrics and Center for Children’s Health Research, University of Rochester, Rochester, New York, USA
| | - Christopher S. Anderson
- Department of Pediatrics and Center for Children’s Health Research, University of Rochester, Rochester, New York, USA
| | - Thomas J. Mariani
- Department of Pediatrics and Center for Children’s Health Research, University of Rochester, Rochester, New York, USA
| | - Juilee Thakar
- Department of Immunology and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Stephen Dewhurst
- Department of Immunology and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
| | - David H. Mathews
- Department of Biochemistry & Biophysics and Center for RNA Biology, University of Rochester Medical Center, Rochester, New York, USA
| | - Liang Huang
- School of Electrical Engineering & Computer Science, Oregon State University, Corvallis, Oregon, USA
| | - Yan Sun
- Department of Immunology and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
57
|
Olivier T, Blomet J, Desmecht D. Central role of lung macrophages in SARS-CoV-2 physiopathology: a cross-model single-cell RNA-seq perspective. Front Immunol 2023; 14:1197588. [PMID: 37350967 PMCID: PMC10282834 DOI: 10.3389/fimmu.2023.1197588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Cytokine storms are considered a driving factor in coronavirus disease 2019 (COVID-19) severity. However, the triggering and resolution of this cytokine production, as well as the link between this phenomenon and infected cells, are still poorly understood. In this study, a cross-species scRNA-seq analysis showed that cytokine-producing macrophages together with pneumocytes were found to be the main contributors of viral transcripts in both Syrian hamsters and African green monkeys. Whatever the cell type, viral read-bearing cells show an apoptotic phenotype. A comparison of SARS-CoV-2 entry receptor candidates showed that Fc receptors are better correlated with infected cells than ACE2, NRP1, or AXL. Although both species show similar interferon responses, differences in adaptive immunity were highlighted. Lastly, Fc receptor and cytokine upregulation in M1 macrophages was found to correlate with a comprehensive interferon response. Based on these results, we propose a model in which lung macrophages play a central role in COVID-19 severity through antibody-dependent enhancement.
Collapse
Affiliation(s)
- Thibaut Olivier
- GAS Department, Prevor Research Laboratories, Valmondois, France
- Department of Pathology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liege, Liège, Belgium
| | - Joël Blomet
- GAS Department, Prevor Research Laboratories, Valmondois, France
| | - Daniel Desmecht
- Department of Pathology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liege, Liège, Belgium
| |
Collapse
|
58
|
Kokinos EK, Tsymbal SA, Galochkina AV, Bezlepkina SA, Nikolaeva JV, Vershinina SO, Shtro AA, Tatarskiy VV, Shtil AA, Broude EV, Roninson IB, Dukhinova M. Inhibition of Cyclin-Dependent Kinases 8/19 Restricts Bacterial and Virus-Induced Inflammatory Responses in Monocytes. Viruses 2023; 15:1292. [PMID: 37376593 PMCID: PMC10305654 DOI: 10.3390/v15061292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Hyperactivation of the immune system remains a dramatic, life-threatening complication of viral and bacterial infections, particularly during pneumonia. Therapeutic approaches to counteract local and systemic outbreaks of cytokine storm and to prevent tissue damage remain limited. Cyclin-dependent kinases 8 and 19 (CDK8/19) potentiate transcriptional responses to the altered microenvironment, but CDK8/19 potential in immunoregulation is not fully understood. In the present study, we investigated how a selective CDK8/19 inhibitor, Senexin B, impacts the immunogenic profiles of monocytic cells stimulated using influenza virus H1N1 or bacterial lipopolysaccharides. Senexin B was able to prevent the induction of gene expression of proinflammatory cytokines in THP1 and U937 cell lines and in human peripheral blood-derived mononuclear cells. Moreover, Senexin B substantially reduced functional manifestations of inflammation, including clustering and chemokine-dependent migration of THP1 monocytes and human pulmonary fibroblasts (HPF).
Collapse
Affiliation(s)
- Elena K Kokinos
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| | - Sergey A Tsymbal
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| | - Anastasia V Galochkina
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popov Street, 197376 Saint-Petersburg, Russia
| | - Svetlana A Bezlepkina
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| | - Julia V Nikolaeva
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popov Street, 197376 Saint-Petersburg, Russia
| | - Sofia O Vershinina
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| | - Anna A Shtro
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popov Street, 197376 Saint-Petersburg, Russia
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
| | - Alexander A Shtil
- Blokhin National Medical Research Center of Oncology, Kashirskoe Highway 24, 115478 Moscow, Russia
| | - Eugenia V Broude
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Sumter Street 715, Columbia, SC 29208, USA
| | - Igor B Roninson
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Sumter Street 715, Columbia, SC 29208, USA
| | - Marina Dukhinova
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| |
Collapse
|
59
|
Mohandas S, Jagannathan P, Henrich TJ, Sherif ZA, Bime C, Quinlan E, Portman MA, Gennaro M, Rehman J. Immune mechanisms underlying COVID-19 pathology and post-acute sequelae of SARS-CoV-2 infection (PASC). eLife 2023; 12:e86014. [PMID: 37233729 PMCID: PMC10219649 DOI: 10.7554/elife.86014] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
With a global tally of more than 500 million cases of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections to date, there are growing concerns about the post-acute sequelae of SARS-CoV-2 infection (PASC), also known as long COVID. Recent studies suggest that exaggerated immune responses are key determinants of the severity and outcomes of the initial SARS-CoV-2 infection as well as subsequent PASC. The complexity of the innate and adaptive immune responses in the acute and post-acute period requires in-depth mechanistic analyses to identify specific molecular signals as well as specific immune cell populations which promote PASC pathogenesis. In this review, we examine the current literature on mechanisms of immune dysregulation in severe COVID-19 and the limited emerging data on the immunopathology of PASC. While the acute and post-acute phases may share some parallel mechanisms of immunopathology, it is likely that PASC immunopathology is quite distinct and heterogeneous, thus requiring large-scale longitudinal analyses in patients with and without PASC after an acute SARS-CoV-2 infection. By outlining the knowledge gaps in the immunopathology of PASC, we hope to provide avenues for novel research directions that will ultimately lead to precision therapies which restore healthy immune function in PASC patients.
Collapse
Affiliation(s)
- Sindhu Mohandas
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Prasanna Jagannathan
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford UniversityStanfordUnited States
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Zaki A Sherif
- Department of Biochemistry & Molecular Biology, Howard University College of MedicineWashingtonUnited States
| | - Christian Bime
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, University of Arizona College of MedicineTucsonUnited States
| | - Erin Quinlan
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
| | - Michael A Portman
- Seattle Children’s Hospital, Division of Pediatric Cardiology, Department of Pediatrics, University of WashingtonSeattleUnited States
| | - Marila Gennaro
- Public Health Research Institute and Department of Medicine, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of MedicineChicagoUnited States
| |
Collapse
|
60
|
Drake KA, Talantov D, Tong GJ, Lin JT, Verheijden S, Katz S, Leung JM, Yuen B, Krishna V, Wu MJ, Sutherland A, Short SA, Kheradpour P, Mumbach M, Franz K, Trifonov V, Lucas MV, Merson J, Kim CC. Multi-omic Profiling Reveals Early Immunological Indicators for Identifying COVID-19 Progressors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.25.542297. [PMID: 37292797 PMCID: PMC10246026 DOI: 10.1101/2023.05.25.542297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to a rapid response by the scientific community to further understand and combat its associated pathologic etiology. A focal point has been on the immune responses mounted during the acute and post-acute phases of infection, but the immediate post-diagnosis phase remains relatively understudied. We sought to better understand the immediate post-diagnosis phase by collecting blood from study participants soon after a positive test and identifying molecular associations with longitudinal disease outcomes. Multi-omic analyses identified differences in immune cell composition, cytokine levels, and cell subset-specific transcriptomic and epigenomic signatures between individuals on a more serious disease trajectory (Progressors) as compared to those on a milder course (Non-progressors). Higher levels of multiple cytokines were observed in Progressors, with IL-6 showing the largest difference. Blood monocyte cell subsets were also skewed, showing a comparative decrease in non-classical CD14-CD16+ and intermediate CD14+CD16+ monocytes. Additionally, in the lymphocyte compartment, CD8+ T effector memory cells displayed a gene expression signature consistent with stronger T cell activation in Progressors. Importantly, the identification of these cellular and molecular immune changes occurred at the early stages of COVID-19 disease. These observations could serve as the basis for the development of prognostic biomarkers of disease risk and interventional strategies to improve the management of severe COVID-19.
Collapse
Affiliation(s)
| | | | - Gary J Tong
- Verily Life Sciences, South San Francisco, CA
| | - Jack T Lin
- Verily Life Sciences, South San Francisco, CA
| | | | - Samuel Katz
- Verily Life Sciences, South San Francisco, CA
| | | | | | | | | | | | | | | | | | - Kate Franz
- Verily Life Sciences, South San Francisco, CA
| | | | | | - James Merson
- Janssen Research & Development, LLC, San Diego, CA
| | | |
Collapse
|
61
|
Lee CM, Kim M, Kang CK, Choe PG, Kim NJ, Bang H, Cho T, Shin HM, Kim HR, Park WB, Oh MD. Different degree of cytokinemia and T-cell activation according to serum IL-6 levels in critical COVID-19. Front Immunol 2023; 14:1110874. [PMID: 37081872 PMCID: PMC10110916 DOI: 10.3389/fimmu.2023.1110874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
Introduction Tocilizumab, a humanized anti-interleukin-6 receptor (IL-6R) antibody, is recommended for the treatment of severe to critical coronavirus diseases 2019 (COVID-19). However, there were conflicting results on the efficacy of tocilizumab. Therefore, we hypothesized that the differences in tocilizumab efficacy may stem from the different immune responses of critical COVID-19 patients. In this study, we described two groups of immunologically distinct COVID-19 patients, based on their IL-6 response. Methods We prospectively enrolled critical COVID-19 patients, requiring oxygen support with a high flow nasal cannula or a mechanical ventilator, and analyzed their serial samples. An enzyme-linked immunosorbent assay and flow cytometry were used to evaluate the cytokine kinetics and cellular immune responses, respectively. Results A total of nine patients with critical COVID-19 were included. The high (n = 5) and low IL-6 (n = 4) groups were distinguished by their peak serum IL-6 levels, using 400 pg/mL as the cut-off value. Although the difference of flow cytometric data did not reach the level of statistical significance, the levels of pro-inflammatory cytokines and the frequencies of intermediate monocytes (CD14+CD16+), IFN-γ+ CD4+ or CD8+ T cells, and HLA-DR+PD-1+ CD4+ T cells were higher in the high IL-6 group than in the low IL-6 group. Conclusion There were distinctive two groups of critical COVID-19 according to serum IL-6 levels having different degrees of cytokinemia and T-cell responses. Our results indicate that the use of immune modulators should be more tailored in patients with critical COVID-19.
Collapse
Affiliation(s)
- Chan Mi Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Minji Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Anatomy & Cell Biology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chang Kyung Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Pyoeng Gyun Choe
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Nam Joong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyeeun Bang
- Research and development team 2, Molecular Diagnostics Division, Quantamatrix Inc., Seoul, Republic of Korea
| | - Taeeun Cho
- Research and development team 2, Molecular Diagnostics Division, Quantamatrix Inc., Seoul, Republic of Korea
| | - Hyun Mu Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea
| | - Hang-Rae Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Anatomy & Cell Biology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea
- Medical Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Myoung-don Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
62
|
Xia L, Yuan LZ, Hu YH, Liu JY, Hu GS, Qi RY, Zhang TY, Xiong HL, Zheng ZZ, Lin HW, Zhang JM, Yu C, Zhou M, Ma J, Cheng T, Chen RR, Guan Y, Xia NS, Liu W. A SARS-CoV-2-specific CAR-T-cell model identifies felodipine, fasudil, imatinib, and caspofungin as potential treatments for lethal COVID-19. Cell Mol Immunol 2023; 20:351-364. [PMID: 36864189 PMCID: PMC9979130 DOI: 10.1038/s41423-023-00985-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 01/10/2023] [Accepted: 02/06/2023] [Indexed: 03/04/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced cytokine storm is closely associated with coronavirus disease 2019 (COVID-19) severity and lethality. However, drugs that are effective against inflammation to treat lethal COVID-19 are still urgently needed. Here, we constructed a SARS-CoV-2 spike protein-specific CAR, and human T cells infected with this CAR (SARS-CoV-2-S CAR-T) and stimulated with spike protein mimicked the T-cell responses seen in COVID-19 patients, causing cytokine storm and displaying a distinct memory, exhausted, and regulatory T-cell phenotype. THP1 remarkably augmented cytokine release in SARS-CoV-2-S CAR-T cells when they were in coculture. Based on this "two-cell" (CAR-T and THP1 cells) model, we screened an FDA-approved drug library and found that felodipine, fasudil, imatinib, and caspofungin were effective in suppressing the release of cytokines, which was likely due to their ability to suppress the NF-κB pathway in vitro. Felodipine, fasudil, imatinib, and caspofungin were further demonstrated, although to different extents, to attenuate lethal inflammation, ameliorate severe pneumonia, and prevent mortality in a SARS-CoV-2-infected Syrian hamster model, which were also linked to their suppressive role in inflammation. In summary, we established a SARS-CoV-2-specific CAR-T-cell model that can be utilized as a tool for anti-inflammatory drug screening in a fast and high-throughput manner. The drugs identified herein have great potential for early treatment to prevent COVID-19 patients from cytokine storm-induced lethality in the clinic because they are safe, inexpensive, and easily accessible for immediate use in most countries.
Collapse
Affiliation(s)
- Lin Xia
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Lun-Zhi Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Ya-Hong Hu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Jun-Yi Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Guo-Sheng Hu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Ruo-Yao Qi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Tian-Ying Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Hua-Long Xiong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Zao-Zao Zheng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Hong-Wei Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Jia-Mo Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Chao Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Ming Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Jian Ma
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Ri-Rong Chen
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Guan
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ning-Shao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China.
| | - Wen Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China.
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China.
| |
Collapse
|
63
|
Schmitt CA, Tchkonia T, Niedernhofer LJ, Robbins PD, Kirkland JL, Lee S. COVID-19 and cellular senescence. Nat Rev Immunol 2023; 23:251-263. [PMID: 36198912 PMCID: PMC9533263 DOI: 10.1038/s41577-022-00785-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2022] [Indexed: 11/15/2022]
Abstract
The clinical severity of coronavirus disease 2019 (COVID-19) is largely determined by host factors. Recent advances point to cellular senescence, an ageing-related switch in cellular state, as a critical regulator of SARS-CoV-2-evoked hyperinflammation. SARS-CoV-2, like other viruses, can induce senescence and exacerbates the senescence-associated secretory phenotype (SASP), which is comprised largely of pro-inflammatory, extracellular matrix-degrading, complement-activating and pro-coagulatory factors secreted by senescent cells. These effects are enhanced in elderly individuals who have an increased proportion of pre-existing senescent cells in their tissues. SASP factors can contribute to a 'cytokine storm', tissue-destructive immune cell infiltration, endothelialitis (endotheliitis), fibrosis and microthrombosis. SASP-driven spreading of cellular senescence uncouples tissue injury from direct SARS-CoV-2-inflicted cellular damage in a paracrine fashion and can further amplify the SASP by increasing the burden of senescent cells. Preclinical and early clinical studies indicate that targeted elimination of senescent cells may offer a novel therapeutic opportunity to attenuate clinical deterioration in COVID-19 and improve resilience following infection with SARS-CoV-2 or other pathogens.
Collapse
Affiliation(s)
- Clemens A Schmitt
- Charité-Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumour Immunology, and Molekulares Krebsforschungszentrum-MKFZ, Campus Virchow Klinikum, Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Faculty of Medicine, Johannes Kepler University, Linz, Austria.
- Kepler University Hospital, Department of Hematology and Oncology, Linz, Austria.
- Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium), Partner site Berlin, Berlin, Germany.
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology, and Biochemistry, University of Minnesota, Minneapolis, MN, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology, and Biochemistry, University of Minnesota, Minneapolis, MN, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Soyoung Lee
- Charité-Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumour Immunology, and Molekulares Krebsforschungszentrum-MKFZ, Campus Virchow Klinikum, Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Faculty of Medicine, Johannes Kepler University, Linz, Austria.
| |
Collapse
|
64
|
Arduini A, Laprise F, Liang C. SARS-CoV-2 ORF8: A Rapidly Evolving Immune and Viral Modulator in COVID-19. Viruses 2023; 15:871. [PMID: 37112851 PMCID: PMC10141009 DOI: 10.3390/v15040871] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
The COVID-19 pandemic has resulted in upwards of 6.8 million deaths over the past three years, and the frequent emergence of variants continues to strain global health. Although vaccines have greatly helped mitigate disease severity, SARS-CoV-2 is likely to remain endemic, making it critical to understand its viral mechanisms contributing to pathogenesis and discover new antiviral therapeutics. To efficiently infect, this virus uses a diverse set of strategies to evade host immunity, accounting for its high pathogenicity and rapid spread throughout the COVID-19 pandemic. Behind some of these critical host evasion strategies is the accessory protein Open Reading Frame 8 (ORF8), which has gained recognition in SARS-CoV-2 pathogenesis due to its hypervariability, secretory property, and unique structure. This review discusses the current knowledge on SARS-CoV-2 ORF8 and proposes actualized functional models describing its pivotal roles in both viral replication and immune evasion. A better understanding of ORF8's interactions with host and viral factors is expected to reveal essential pathogenic strategies utilized by SARS-CoV-2 and inspire the development of novel therapeutics to improve COVID-19 disease outcomes.
Collapse
Affiliation(s)
- Ariana Arduini
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (A.A.); (F.L.)
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| | - Frederique Laprise
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (A.A.); (F.L.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (A.A.); (F.L.)
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
65
|
Urbiola-Salvador V, Lima de Souza S, Grešner P, Qureshi T, Chen Z. Plasma Proteomics Unveil Novel Immune Signatures and Biomarkers upon SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:ijms24076276. [PMID: 37047248 PMCID: PMC10093853 DOI: 10.3390/ijms24076276] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Several elements have an impact on COVID-19, including comorbidities, age and sex. To determine the protein profile changes in peripheral blood caused by a SARS-CoV-2 infection, a proximity extension assay was used to quantify 1387 proteins in plasma samples among 28 Finnish patients with COVID-19 with and without comorbidities and their controls. Key immune signatures, including CD4 and CD28, were changed in patients with comorbidities. Importantly, several unreported elevated proteins in patients with COVID-19, such as RBP2 and BST2, which show anti-microbial activity, along with proteins involved in extracellular matrix remodeling, including MATN2 and COL6A3, were identified. RNF41 was downregulated in patients compared to healthy controls. Our study demonstrates that SARS-CoV-2 infection causes distinct plasma protein changes in the presence of comorbidities despite the interpatient heterogeneity, and several novel potential biomarkers associated with a SARS-CoV-2 infection alone and in the presence of comorbidities were identified. Protein changes linked to the generation of SARS-CoV-2-specific antibodies, long-term effects and potential association with post-COVID-19 condition were revealed. Further study to characterize the identified plasma protein changes from larger cohorts with more diverse ethnicities of patients with COVID-19 combined with functional studies will facilitate the identification of novel diagnostic, prognostic biomarkers and potential therapeutic targets for patients with COVID-19.
Collapse
Affiliation(s)
- Víctor Urbiola-Salvador
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, 80-307 Gdańsk, Pomerania, Poland
| | - Suiane Lima de Souza
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, North Ostrobothnia, Finland
| | - Peter Grešner
- Department of Translational Oncology, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, Medical University of Gdańsk, 80-211 Gdańsk, Pomerania, Poland
| | - Talha Qureshi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, North Ostrobothnia, Finland
| | - Zhi Chen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, North Ostrobothnia, Finland
- Correspondence:
| |
Collapse
|
66
|
Aghamiri SS, Puniya BL, Amin R, Helikar T. A multiscale mechanistic model of human dendritic cells for in-silico investigation of immune responses and novel therapeutics discovery. Front Immunol 2023; 14:1112985. [PMID: 36993954 PMCID: PMC10040975 DOI: 10.3389/fimmu.2023.1112985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/22/2023] [Indexed: 03/14/2023] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells (APCs) with the unique ability to mediate inflammatory responses of the immune system. Given the critical role of DCs in shaping immunity, they present an attractive avenue as a therapeutic target to program the immune system and reverse immune disease disorders. To ensure appropriate immune response, DCs utilize intricate and complex molecular and cellular interactions that converge into a seamless phenotype. Computational models open novel frontiers in research by integrating large-scale interaction to interrogate the influence of complex biological behavior across scales. The ability to model large biological networks will likely pave the way to understanding any complex system in more approachable ways. We developed a logical and predictive model of DC function that integrates the heterogeneity of DCs population, APC function, and cell-cell interaction, spanning molecular to population levels. Our logical model consists of 281 components that connect environmental stimuli with various layers of the cell compartments, including the plasma membrane, cytoplasm, and nucleus to represent the dynamic processes within and outside the DC, such as signaling pathways and cell-cell interactions. We also provided three sample use cases to apply the model in the context of studying cell dynamics and disease environments. First, we characterized the DC response to Sars-CoV-2 and influenza co-infection by in-silico experiments and analyzed the activity level of 107 molecules that play a role in this co-infection. The second example presents simulations to predict the crosstalk between DCs and T cells in a cancer microenvironment. Finally, for the third example, we used the Kyoto Encyclopedia of Genes and Genomes enrichment analysis against the model's components to identify 45 diseases and 24 molecular pathways that the DC model can address. This study presents a resource to decode the complex dynamics underlying DC-derived APC communication and provides a platform for researchers to perform in-silico experiments on human DC for vaccine design, drug discovery, and immunotherapies.
Collapse
Affiliation(s)
| | | | - Rada Amin
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
67
|
Shao L, Wu B, Liu C, Chong W. VALPROIC ACID INHIBITS CLASSICAL MONOCYTE-DERIVED TISSUE FACTOR AND ALLEVIATES HEMORRHAGIC SHOCK-INDUCED ACUTE LUNG INJURY IN RATS. Shock 2023; 59:449-459. [PMID: 36443067 PMCID: PMC9997640 DOI: 10.1097/shk.0000000000002064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/27/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
ABSTRACT Background: Monocytes and monocyte-derived tissue factor (TF) promote the development of sepsis-induced acute lung injury (ALI). Classical monocytes (C-Mcs) can be induced to express TF. Valproic acid (VPA) alleviates hemorrhagic shock (HS)-induced ALI (HS/ALI) and inhibits TF expression in monocytes. We hypothesized that C-Mcs and C-Mc-derived TF promoted HS/ALI and that VPA could inhibit C-Mc-derived TF expression and attenuate HS/ALI. Methods: Wistar rats and THP-1 cells were used to evaluate our hypothesis. Monocyte subtypes were analyzed by flow cytometry; mRNA expression was measured by fluorescence quantitative polymerase chain reaction; protein expression was measured by Western blotting, immunofluorescence, or immunohistology; inflammatory cytokines levels were measured by enzyme-linked immunosorbent assay; and ALI scores were used to determine the degree of ALI. Results: The blood %C-Mcs and C-Mcs/non-C-Mcs ratios, monocyte TF levels, serum and/or lung inflammatory cytokine levels, and ALI scores of HS rats were significantly increased ( P < 0.05). After monocyte depletion and thrombin inhibition, the inflammatory cytokine levels and ALI scores were significantly decreased ( P < 0.05). VPA reduced the %C-Mcs and C-Mc/non-C-Mc ratios, TF expression, inflammatory cytokine levels, and ALI scores during HS ( P < 0.05) and inhibited HS-induced monocyte Egr-1 and p-ERK1/2 expression ( P < 0.05). VPA inhibited hypoxia-induced TF expression in THP-1 cells by regulating the p-ERK1/2-Egr-1 axis. Conclusion: C-Mcs and C-Mc-derived TF accelerate the development of HS/ALI by increasing thrombin production. VPA inhibits HS-induced C-Mc production of TF by regulating the p-ERK1/2-Egr-1 axis and alleviates HS/ALI.
Collapse
Affiliation(s)
- Lina Shao
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
- Intensive Care Unit, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
- Intensive Care Unit, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province, China
- Intensive Care Unit, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Bing Wu
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Chang Liu
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Chong
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
68
|
Walter LO, Cardoso CC, Santos-Pirath ÍM, Costa HZ, Gartner R, Werle I, Mohr ETB, da Rosa JS, Lubschinski TL, Felisberto M, Kretzer IF, Masukawa II, Vanny PDA, Luiz MC, de Moraes ACR, Dalmarco EM, Santos-Silva MC. T cell maturation is significantly affected by SARS-CoV-2 infection. Immunology 2023. [PMID: 36855300 DOI: 10.1111/imm.13635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory tract infection caused by the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). An adequate T cell response is essential not only for fighting disease but also for the creation of immune memory. Thus, the present study aims to evaluate the T cells of patients with moderate, severe and critical COVID-19 not only at the time of illness but also 2 months after diagnosis to observe whether changes in this compartment persist. In this study, 166 COVID-19 patients were stratified into moderate/severe and critical disease categories. The maturation and activation of T cells were evaluated through flow cytometry. In addition, Treg cells were analysed. Until 15 days after diagnosis, patients presented a reduction in absolute and relative T lymphocyte counts. After 2 months, in moderate/severe patients, the counts returned to a similar level as that of the control group. In convalescent patients who had a critical illness, absolute T lymphocyte values increased considerably. Patients with active disease did not show differentiation of T cells. Nonetheless, after 2 months, patients with critical COVID-19 showed a significant increase in CD4+ EMRA (CD45RA+ effector memory) T lymphocytes. Furthermore, COVID-19 patients showed delayed T cell activation and reduced CD8+ suppressor T cells even 2 months after diagnosis. A reduction in CD4+ Treg cells was also observed, and their numbers returned to a similar level as that of healthy controls in convalescent patients. The results demonstrate that COVID-19 patients have a delayed activation and differentiation of T cells. In addition, these patients have a great reduction of T cells with a suppressor phenotype.
Collapse
Affiliation(s)
- Laura Otto Walter
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Chandra Chiappin Cardoso
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Íris Mattos Santos-Pirath
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Heloisa Zorzi Costa
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Rafaela Gartner
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Isabel Werle
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Julia Salvan da Rosa
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Mariano Felisberto
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Iara Fabricia Kretzer
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Ivete Ioshiko Masukawa
- Infectious Disease Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil.,Infectious Disease Service, Nereu Ramos Hospital, State Health Department, Florianópolis, Brazil
| | - Patrícia de Almeida Vanny
- Infectious Disease Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Magali Chaves Luiz
- Infectious Disease Service, Nereu Ramos Hospital, State Health Department, Florianópolis, Brazil
| | - Ana Carolina Rabello de Moraes
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eduardo Monguilhott Dalmarco
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Maria Cláudia Santos-Silva
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil.,Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
69
|
Chen Y, Chen L, Xu H, Cao R, Morisseau C, Zhang M, Shi Y, Hammock BD, Wang J, Zhuang J, Liu Z, Chen G. Structure-Directed Discovery of Potent Soluble Epoxide Hydrolase Inhibitors for the Treatment of Inflammatory Diseases. J Med Chem 2023; 66:2979-3009. [PMID: 36689364 PMCID: PMC9974930 DOI: 10.1021/acs.jmedchem.2c01996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Soluble epoxide hydrolase (sEH) has been identified as an attractive target for anti-inflammatory drug design in recent years. Picomolar level compound G1 against sEH was obtained by introducing the hydrophilic group homopiperazine and hydrophobic fragment propionyl onto the structure of lead compound A. G1 showed good microsomal stability, a moderate plasma protein binding rate, and good oral bioavailability and was well tolerated in rats. G1 has significant analgesic effects on CFA-induced AIA mice, ameliorated the pancreatic injury in acute pancreatitis induced by l-arginine, reversed pancreatic injury, edema, and neutrophil infiltration, and increased the survival time of C57BL/6 mice in a lipopolysaccharide (LPS)-induced sepsis model. Moreover the expression levels of sEH, COX-2, NOS-2, vascular cell adhesion molecule (VCAM), IL-6, MCP-5, and tumor necrosis factor α (TNF-α) were measured by Western blot or enzyme-linked immunosorbent assay (ELISA), with varying degrees of decrease. These results suggested that G1 is a drug candidate worthy of further evaluation for the treatment of inflammation-induced diseases such as arthritis, acute pancreatitis, and sepsis.
Collapse
Affiliation(s)
- Yuanguang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huashen Xu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ruolin Cao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Christophe Morisseau
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA
| | - Maoying Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yajie Shi
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA
| | - Jieru Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Junning Zhuang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhongbo Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
70
|
Nevejan L, Strypens T, Van Nieuwenhove M, Boel A, Cattoir L, Van Vaerenbergh K, Meeus P, Bossuyt X, De Neve N, Van Hoovels L. Serial measurement of circulating calprotectin as a prognostic biomarker in COVID-19 patients in intensive care setting. Clin Chem Lab Med 2023; 61:494-502. [PMID: 36473060 DOI: 10.1515/cclm-2022-1165] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Circulating calprotectin (cCLP) has been shown to be a promising prognostic marker for COVID-19 severity. We aimed to investigate the prognostic value of serial measurements of cCLP in COVID-19 patients admitted to an intensive care unit (ICU). METHODS From November 2020 to May 2021, patients with COVID-19, admitted at the ICU of the OLV Hospital, Aalst, Belgium, were prospectively included. For sixty-six (66) patients, blood samples were collected at admission and subsequently every 48 h during ICU stay. On every sample (total n=301), a cCLP (EliA™ Calprotectin 2, Phadia 200, Thermo Fisher Scientific; serum/plasma protocol (for Research Use Only, -RUO-) and C-reactive protein (CRP; cobas c501/c503, Roche Diagnostics) analysis were performed. Linear mixed models were used to associate biomarkers levels with mortality, need for mechanical ventilation, length of stay at ICU (LOS-ICU) and medication use (antibiotics, corticosteroids, antiviral and immune suppressant/modulatory drugs). RESULTS Longitudinally higher levels of all biomarkers were associated with LOS-ICU and with the need for mechanical ventilation. Medication use and LOS-ICU were not associated with variations in cCLP and CRP levels. cCLP levels increased significantly during ICU hospitalization in the deceased group (n=21/66) but decreased in the non-deceased group (n=45/66). In contrast, CRP levels decreased non-significantly in both patient groups, although significantly longitudinally higher CRP levels were obtained in the deceased subgroup. CONCLUSIONS Serial measurements of cCLP provides prognostic information which can be useful to guide clinical management of COVID-19 patients in ICU setting.
Collapse
Affiliation(s)
- Louis Nevejan
- Department of Laboratory Medicine, OLV Hospital, Aalst, Belgium.,Department of Laboratory Medicine, University Hospital Leuven, Leuven, Belgium
| | - Thomas Strypens
- Department of Laboratory Medicine, OLV Hospital, Aalst, Belgium.,Department of Laboratory Medicine, University Hospital Leuven, Leuven, Belgium
| | - Mathias Van Nieuwenhove
- Department of Intensive Care Medicine, OLV Hospital, Aalst, Belgium.,Department of Anesthesiology, OLV Hospital, Aalst, Belgium
| | - An Boel
- Department of Laboratory Medicine, OLV Hospital, Aalst, Belgium
| | - Lien Cattoir
- Department of Laboratory Medicine, OLV Hospital, Aalst, Belgium
| | | | - Peter Meeus
- Department of Laboratory Medicine, OLV Hospital, Aalst, Belgium
| | - Xavier Bossuyt
- Department of Laboratory Medicine, University Hospital Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Nikolaas De Neve
- Department of Intensive Care Medicine, OLV Hospital, Aalst, Belgium.,Department of Anesthesiology, OLV Hospital, Aalst, Belgium
| | - Lieve Van Hoovels
- Department of Laboratory Medicine, OLV Hospital, Aalst, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| |
Collapse
|
71
|
He B, Xiao Y, Liang H, Huang Q, Du Y, Li Y, Garmire D, Sun D, Garmire LX. ASGARD is A Single-cell Guided Pipeline to Aid Repurposing of Drugs. Nat Commun 2023; 14:993. [PMID: 36813801 PMCID: PMC9945835 DOI: 10.1038/s41467-023-36637-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Single-cell RNA sequencing technology has enabled in-depth analysis of intercellular heterogeneity in various diseases. However, its full potential for precision medicine has yet to be reached. Towards this, we propose A Single-cell Guided Pipeline to Aid Repurposing of Drugs (ASGARD) that defines a drug score to recommend drugs by considering all cell clusters to address the intercellular heterogeneity within each patient. ASGARD shows significantly better average accuracy on single-drug therapy compared to two bulk-cell-based drug repurposing methods. We also demonstrated that it performs considerably better than other cell cluster-level predicting methods. In addition, we validate ASGARD using the drug response prediction method TRANSACT with Triple-Negative-Breast-Cancer patient samples. We find that many top-ranked drugs are either approved by the Food and Drug Administration or in clinical trials treating corresponding diseases. In conclusion, ASGARD is a promising drug repurposing recommendation tool guided by single-cell RNA-seq for personalized medicine. ASGARD is free for educational use at https://github.com/lanagarmire/ASGARD .
Collapse
Affiliation(s)
- Bing He
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yao Xiao
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Haodong Liang
- Department of Statistics, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Qianhui Huang
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yuheng Du
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yijun Li
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - David Garmire
- Department of Electrical Engineering and Computer Science, College of Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Lana X Garmire
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
72
|
Sharma SB, Melvin WJ, Audu CO, Bame M, Rhoads N, Wu W, Kanthi Y, Knight JS, Adili R, Holinstat MA, Wakefield TW, Henke PK, Moore BB, Gallagher KA, Obi AT. The histone methyltransferase MLL1/KMT2A in monocytes drives coronavirus-associated coagulopathy and inflammation. Blood 2023; 141:725-742. [PMID: 36493338 PMCID: PMC9743412 DOI: 10.1182/blood.2022015917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
Coronavirus-associated coagulopathy (CAC) is a morbid and lethal sequela of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. CAC results from a perturbed balance between coagulation and fibrinolysis and occurs in conjunction with exaggerated activation of monocytes/macrophages (MO/Mφs), and the mechanisms that collectively govern this phenotype seen in CAC remain unclear. Here, using experimental models that use the murine betacoronavirus MHVA59, a well-established model of SARS-CoV-2 infection, we identify that the histone methyltransferase mixed lineage leukemia 1 (MLL1/KMT2A) is an important regulator of MO/Mφ expression of procoagulant and profibrinolytic factors such as tissue factor (F3; TF), urokinase (PLAU), and urokinase receptor (PLAUR) (herein, "coagulopathy-related factors") in noninfected and infected cells. We show that MLL1 concurrently promotes the expression of the proinflammatory cytokines while suppressing the expression of interferon alfa (IFN-α), a well-known inducer of TF and PLAUR. Using in vitro models, we identify MLL1-dependent NF-κB/RelA-mediated transcription of these coagulation-related factors and identify a context-dependent, MLL1-independent role for RelA in the expression of these factors in vivo. As functional correlates for these findings, we demonstrate that the inflammatory, procoagulant, and profibrinolytic phenotypes seen in vivo after coronavirus infection were MLL1-dependent despite blunted Ifna induction in MO/Mφs. Finally, in an analysis of SARS-CoV-2 positive human samples, we identify differential upregulation of MLL1 and coagulopathy-related factor expression and activity in CD14+ MO/Mφs relative to noninfected and healthy controls. We also observed elevated plasma PLAU and TF activity in COVID-positive samples. Collectively, these findings highlight an important role for MO/Mφ MLL1 in promoting CAC and inflammation.
Collapse
Affiliation(s)
- Sriganesh B. Sharma
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - William J. Melvin
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Christopher O. Audu
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Monica Bame
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Nicole Rhoads
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Weisheng Wu
- Bioinformatics Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis & Inflammation, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Michael A. Holinstat
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Thomas W. Wakefield
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Peter K. Henke
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Katherine A. Gallagher
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Andrea T. Obi
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| |
Collapse
|
73
|
Brown B, Ojha V, Fricke I, Al-Sheboul SA, Imarogbe C, Gravier T, Green M, Peterson L, Koutsaroff IP, Demir A, Andrieu J, Leow CY, Leow CH. Innate and Adaptive Immunity during SARS-CoV-2 Infection: Biomolecular Cellular Markers and Mechanisms. Vaccines (Basel) 2023; 11:408. [PMID: 36851285 PMCID: PMC9962967 DOI: 10.3390/vaccines11020408] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus 2019 (COVID-19) pandemic was caused by a positive sense single-stranded RNA (ssRNA) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, other human coronaviruses (hCoVs) exist. Historical pandemics include smallpox and influenza, with efficacious therapeutics utilized to reduce overall disease burden through effectively targeting a competent host immune system response. The immune system is composed of primary/secondary lymphoid structures with initially eight types of immune cell types, and many other subtypes, traversing cell membranes utilizing cell signaling cascades that contribute towards clearance of pathogenic proteins. Other proteins discussed include cluster of differentiation (CD) markers, major histocompatibility complexes (MHC), pleiotropic interleukins (IL), and chemokines (CXC). The historical concepts of host immunity are the innate and adaptive immune systems. The adaptive immune system is represented by T cells, B cells, and antibodies. The innate immune system is represented by macrophages, neutrophils, dendritic cells, and the complement system. Other viruses can affect and regulate cell cycle progression for example, in cancers that include human papillomavirus (HPV: cervical carcinoma), Epstein-Barr virus (EBV: lymphoma), Hepatitis B and C (HB/HC: hepatocellular carcinoma) and human T cell Leukemia Virus-1 (T cell leukemia). Bacterial infections also increase the risk of developing cancer (e.g., Helicobacter pylori). Viral and bacterial factors can cause both morbidity and mortality alongside being transmitted within clinical and community settings through affecting a host immune response. Therefore, it is appropriate to contextualize advances in single cell sequencing in conjunction with other laboratory techniques allowing insights into immune cell characterization. These developments offer improved clarity and understanding that overlap with autoimmune conditions that could be affected by innate B cells (B1+ or marginal zone cells) or adaptive T cell responses to SARS-CoV-2 infection and other pathologies. Thus, this review starts with an introduction into host respiratory infection before examining invaluable cellular messenger proteins and then individual immune cell markers.
Collapse
Affiliation(s)
| | | | - Ingo Fricke
- Independent Immunologist and Researcher, 311995 Lamspringe, Germany
| | - Suhaila A Al-Sheboul
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Department of Medical Microbiology, International School of Medicine, Medipol University-Istanbul, Istanbul 34810, Turkey
| | | | - Tanya Gravier
- Independent Researcher, MPH, San Francisco, CA 94131, USA
| | | | | | | | - Ayça Demir
- Faculty of Medicine, Afyonkarahisar University, Istanbul 03030, Turkey
| | - Jonatane Andrieu
- Faculté de Médecine, Aix–Marseille University, 13005 Marseille, France
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, (INFORMM), Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| |
Collapse
|
74
|
Severe COVID-19 patients have impaired plasmacytoid dendritic cell-mediated control of SARS-CoV-2. Nat Commun 2023; 14:694. [PMID: 36755036 PMCID: PMC9907212 DOI: 10.1038/s41467-023-36140-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
Type I and III interferons (IFN-I/λ) are important antiviral mediators against SARS-CoV-2 infection. Here, we demonstrate that plasmacytoid dendritic cells (pDC) are the predominant IFN-I/λ source following their sensing of SARS-CoV-2-infected cells. Mechanistically, this short-range sensing by pDCs requires sustained integrin-mediated cell adhesion with infected cells. In turn, pDCs restrict viral spread by an IFN-I/λ response directed toward SARS-CoV-2-infected cells. This specialized function enables pDCs to efficiently turn-off viral replication, likely via a local response at the contact site with infected cells. By exploring the pDC response in SARS-CoV-2 patients, we further demonstrate that pDC responsiveness inversely correlates with the severity of the disease. The pDC response is particularly impaired in severe COVID-19 patients. Overall, we propose that pDC activation is essential to control SARS-CoV-2-infection. Failure to develop this response could be important to understand severe cases of COVID-19.
Collapse
|
75
|
Wei B, Liu Y, Li H, Peng Y, Luo Z. Impact of alcohol consumption on coronavirus disease 2019 severity: A systematic review and meta-analysis. J Med Virol 2023; 95:e28547. [PMID: 36734064 DOI: 10.1002/jmv.28547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Fear and misinformation lead to widespread myths in the coronavirus disease 2019 (COVID-19) pandemic, such as "consuming high-strength alcohol kills the virus in the inhaled air." However, whether alcohol consumption can affect COVID-19 has not been clarified yet. This study aims to investigate the impact of alcohol consumption on COVID-19 severity. PubMed, Embase, Cochrane Library, Central, CINAHL, ClinicalTrials.gov, and WHO-International Clinical Trials Registry Platform were searched until November 25, 2022. Forty studies (1,697,683 COVID-19 individuals) were analyzed. Brown (patients numbers: 1317, risk ratios [RR] = 1.58, 95% [confidence interval] CI = 1.31 to 1.90, I2 = 0.0%, p < 0.001), American (patients numbers: 3721, RR = 1.51, 95% CI = 1.30 to 1.75, I2 = 0.0%, p < 0.001), and European (patients numbers: 261,437, RR = 2.04, 95% CI = 1.96 to 2.13, I2 = 0.0%, p < 0.001) drinkers were at high risk of severe COVID-19, intensive care unit (ICU) admission, and invasive mechanical ventilation (IMV), respectively. Consistently, individuals with a drinking history were at high risk of severe COVID-19 (patients numbers: 5399, RR = 1.23, 95% CI = 1.02 to 1.48, I2 = 38.4%, p = 0.03) and ICU admission (patients numbers: 6995, RR = 1.32, 95% CI = 1.08 to 1.60, I2 = 46.6%, p = 0.01). In addition, current drinkers had an increased risk of symptomatic COVID-19. However, excessive drinkers were at high risk of COVID-19 hospitalization. Alcohol consumption intensifies COVID-19 severity and deteriorates its clinical outcomes. Here, we strongly propose that people do not drink alcohol during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Baozhu Wei
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
| | - Yang Liu
- Department of Endocrinology, China Resources and WISCO General Hospital, Wuhan, China
| | - Hang Li
- Department of Geratology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yuanyuan Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zhi Luo
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
76
|
Falck‐Jones S, Österberg B, Smed‐Sörensen A. Respiratory and systemic monocytes, dendritic cells, and myeloid-derived suppressor cells in COVID-19: Implications for disease severity. J Intern Med 2023; 293:130-143. [PMID: 35996885 PMCID: PMC9538918 DOI: 10.1111/joim.13559] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Since the beginning of the SARS-CoV-2 pandemic in 2020, researchers worldwide have made efforts to understand the mechanisms behind the varying range of COVID-19 disease severity. Since the respiratory tract is the site of infection, and immune cells differ depending on their anatomical location, studying blood is not sufficient to understand the full immunopathogenesis in patients with COVID-19. It is becoming increasingly clear that monocytes, dendritic cells (DCs), and monocytic myeloid-derived suppressor cells (M-MDSCs) are involved in the immunopathology of COVID-19 and may play important roles in determining disease severity. Patients with mild COVID-19 display an early antiviral (interferon) response in the nasopharynx, expansion of activated intermediate monocytes, and low levels of M-MDSCs in blood. In contrast, patients with severe COVID-19 seem to lack an early efficient induction of interferons, and skew towards a more suppressive response in blood. This is characterized by downregulation of activation markers and decreased functional capacity of blood monocytes and DCs, reduced circulating DCs, and increased levels of HLA-DRlo CD14+ M-MDSCs. These suppressive characteristics could potentially contribute to delayed T-cell responses in severe COVID-19 cases. In contrast, airways of patients with severe COVID-19 display hyperinflammation with elevated levels of inflammatory monocytes and monocyte-derived macrophages, and reduced levels of tissue-resident alveolar macrophages. These monocyte-derived cells contribute to excess inflammation by producing cytokines and chemokines. Here, we review the current knowledge on the role of monocytes, DCs, and M-MDSCs in COVID-19 and how alterations and the anatomical distribution of these cell populations may relate to disease severity.
Collapse
Affiliation(s)
- Sara Falck‐Jones
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Björn Österberg
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Anna Smed‐Sörensen
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska InstitutetKarolinska University HospitalStockholmSweden
| |
Collapse
|
77
|
Islam MM, Islam S, Ahmed R, Majumder M, Sarkar B, Himu MER, Kawser M, Hossain A, Mia MJ, Parag RR, Bulbul MRH, Ahmed S, Sattar MA, Biswas R, Das M, Rahman MM, Shil RK, Parial R, Chowdhury S, Das M, Noman ASM, Hossain MM. Reduced IFN-γ levels along with changes in hematologic and immunologic parameters are key to COVID-19 severity in Bangladeshi patients. Exp Hematol 2023; 118:53-64.e1. [PMID: 36574579 PMCID: PMC9701580 DOI: 10.1016/j.exphem.2022.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/08/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022]
Abstract
The manifestation of coronavirus disease 2019 (COVID-19) severity and mortality has been associated with dysregulation of the immune response, often influenced by racial disparities and conferred by changes in hematologic and immunologic parameters. These biological and hematologic parameters as well as cytokine profiles were investigated in a cohort of 61 COVID-19-positive patients (categorized into mild, moderate, and severe groups) from Bangladesh using standard analytical methods. The data reported that the interleukin (IL)-4 and IL-6 levels were significantly increased, whereas the levels of interferon (IFN)-γ were significantly reduced in patients with severe COVID-19 (p < 0.05) compared with those in patients with mild and/or moderate COVID-19. The extent of erythrocyte sedimentation rate (ESR); neutrophil count; and levels of ferritin, C-reactive protein (CRP), and D-dimer (p < 0.05) were found to be significantly increased, whereas the white blood cell (WBC), lymphocyte, eosinophil, and platelet counts (p < 0.05) were observed to be significantly reduced in patients with severe COVID-19 compared with those in the patients in other 2 groups. Our study exhibited a significantly higher IL-6-to-lymphocyte ratio in patients with severe COVID-19 than in those with mild and moderate COVID-19. The calculated neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), and ferritin-to-ESR ratio were significantly increased in patients with severe COVID-19. The increase in the IL-4 and IL-6 levels along with CRP and D-dimer levels may envisage a hyperinflammatory environment and immune dysregulation, which contribute to prolonged viral persistence, leading to severe disease. However, the reduced level of IFN-γ can be attributed to a less fatality toll in Bangladesh compared with that in the rest of the world.
Collapse
Affiliation(s)
- Mohammed Moinul Islam
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Shafiqul Islam
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh; Stem Cell Genetics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Ridwan Ahmed
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Mohit Majumder
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Bishu Sarkar
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Md Ejajur Rahman Himu
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Md Kawser
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Alamgir Hossain
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Mohammad Jewel Mia
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Rashed Rezwan Parag
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | | | - Shakeel Ahmed
- Bangladesh Institute of Tropical and Infectious Diseases, Chattogram, Bangladesh
| | - M A Sattar
- Department of Medicine, Chittagong Medical College and Hospital, Chattogram, Bangladesh
| | - Rajdeep Biswas
- Anaesthesia & ICU department, General Hospital, Chattogram, Bangladesh
| | - Moumita Das
- Anaesthesia & ICU department, General Hospital, Chattogram, Bangladesh
| | - Md Mizanur Rahman
- EuGEF Research Foundation, Chattogram, Bangladesh; Department of Biochemistry, Rangamati Medical College, Rangamati, Bangladesh
| | - Rajib Kumar Shil
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Ramendu Parial
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Srikanta Chowdhury
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Manisha Das
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Abu Shadat Mohammod Noman
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh
| | - Muhammad Mosaraf Hossain
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, Bangladesh; EuGEF Research Foundation, Chattogram, Bangladesh.
| |
Collapse
|
78
|
Boswell MT, Maimela T, Hameiri-Bowen D, Riley G, Malan A, Steyn N, Nolutshungu N, de Villiers TR, de Beer Z, Mathabathe J, Tshabalala K, Abdullah F, Ramlall R, Heystek M, Basu D, Rheeder P, Ueckermann V, van Hougenhouck-Tulleken W. COVID-19 severity and in-hospital mortality in an area with high HIV prevalence. South Afr J HIV Med 2023; 24:1412. [PMID: 36751479 PMCID: PMC9900246 DOI: 10.4102/sajhivmed.v24i1.1412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/10/2022] [Indexed: 02/04/2023] Open
Abstract
Background HIV infection causes immune dysregulation affecting T-cell and monocyte function, which may alter coronavirus disease 2019 (COVID-19) pathophysiology. Objectives We investigated the associations among clinical phenotypes, laboratory biomarkers, and hospitalisation outcomes in a cohort of people hospitalised with COVID-19 in a high HIV prevalence area. Method We conducted a prospective observational cohort study in Tshwane, South Africa. Respiratory disease severity was quantified using the respiratory oxygenation score. Analysed biomarkers included inflammatory and coagulation biomarkers, CD4 T-cell counts, and HIV-1 viral loads (HIVVL). Results The analysis included 558 patients, of whom 21.7% died during admission. The mean age was 54 years. A total of 82 participants were HIV-positive. People living with HIV (PLWH) were younger (mean age 46 years) than HIV-negative people; most were on antiretroviral treatment with a suppressed HIVVL (72%) and the median CD4 count was 159 (interquartile range: 66-397) cells/µL. After adjusting for age, HIV was not associated with increased risk of mortality during hospitalisation (age-adjusted hazard ratio = 1.1, 95% confidence interval: 0.6-2.0). Inflammatory biomarker levels were similar in PLWH and HIV-negative patients. Detectable HIVVL was associated with less severe respiratory disease. In PLWH, mortality was associated with higher levels of inflammatory biomarkers. Opportunistic infections, and other risk factors for severe COVID-19, were common in PLWH who died. Conclusion PLWH were not at increased risk of mortality and those with detectable HIVVL had less severe respiratory disease than those with suppressed HIVVL. What this study adds This study advances our understanding of severe COVID-19 in PLWH.
Collapse
Affiliation(s)
- Michael T. Boswell
- Department of Internal Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Tshegofatso Maimela
- Clinical Public Health Unit, Department of Public Health Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Dan Hameiri-Bowen
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | - George Riley
- Department of Internal Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | | | - Nickietta Steyn
- Department of Internal Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Nomonde Nolutshungu
- Department of Medical Immunology, University of Pretoria, Pretoria, South Africa
| | | | | | - John Mathabathe
- Clinical Public Health Unit, Department of Public Health Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Khanyisile Tshabalala
- Clinical Public Health Unit, Department of Public Health Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Fareed Abdullah
- South African Medical Research Council, Pretoria, South Africa
| | | | | | - Debashis Basu
- Clinical Public Health Unit, Department of Public Health Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Paul Rheeder
- Department of Internal Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Veronica Ueckermann
- Department of Internal Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | | |
Collapse
|
79
|
Lai YJ, Liu SH, Manachevakul S, Lee TA, Kuo CT, Bello D. Biomarkers in long COVID-19: A systematic review. Front Med (Lausanne) 2023; 10:1085988. [PMID: 36744129 PMCID: PMC9895110 DOI: 10.3389/fmed.2023.1085988] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/02/2023] [Indexed: 01/21/2023] Open
Abstract
Purpose Long COVID, also known as post-acute sequelae of COVID-19, refers to the constellation of long-term symptoms experienced by people suffering persistent symptoms for one or more months after SARS-CoV-2 infection. Blood biomarkers can be altered in long COVID patients; however, biomarkers associated with long COVID symptoms and their roles in disease progression remain undetermined. This study aims to systematically evaluate blood biomarkers that may act as indicators or therapeutic targets for long COVID. Methods A systematic literature review in PubMed, Embase, and CINAHL was performed on 18 August 2022. The search keywords long COVID-19 symptoms and biomarkers were used to filter out the eligible studies, which were then carefully evaluated. Results Identified from 28 studies and representing six biological classifications, 113 biomarkers were significantly associated with long COVID: (1) Cytokine/Chemokine (38, 33.6%); (2) Biochemical markers (24, 21.2%); (3) Vascular markers (20, 17.7%); (4) Neurological markers (6, 5.3%); (5) Acute phase protein (5, 4.4%); and (6) Others (20, 17.7%). Compared with healthy control or recovered patients without long COVID symptoms, 79 biomarkers were increased, 29 were decreased, and 5 required further determination in the long COVID patients. Of these, up-regulated Interleukin 6, C-reactive protein, and tumor necrosis factor alpha might serve as the potential diagnostic biomarkers for long COVID. Moreover, long COVID patients with neurological symptoms exhibited higher levels of neurofilament light chain and glial fibrillary acidic protein whereas those with pulmonary symptoms exhibited a higher level of transforming growth factor beta. Conclusion Long COVID patients present elevated inflammatory biomarkers after initial infection. Our study found significant associations between specific biomarkers and long COVID symptoms. Further investigations are warranted to identify a core set of blood biomarkers that can be used to diagnose and manage long COVID patients in clinical practice.
Collapse
Affiliation(s)
- Yun-Ju Lai
- School of Nursing, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, United States
| | - Shou-Hou Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sumatchara Manachevakul
- School of Nursing, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, United States
| | - Te-An Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Tse Kuo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Dhimiter Bello
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, United States
| |
Collapse
|
80
|
Carai P, González LF, Van Bruggen S, Spalart V, De Giorgio D, Geuens N, Martinod K, Jones EAV, Heymans S. Neutrophil inhibition improves acute inflammation in a murine model of viral myocarditis. Cardiovasc Res 2023; 118:3331-3345. [PMID: 35426438 PMCID: PMC9847559 DOI: 10.1093/cvr/cvac052] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/07/2022] [Accepted: 03/24/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Viral myocarditis (VM) is an inflammatory pathology of the myocardium triggered by a viral infection that may cause sudden death or heart failure (HF), especially in the younger population. Current treatments only stabilize and improve cardiac function without resolving the underlying inflammatory cause. The factors that induce VM to progress to HF are still uncertain, but neutrophils have been increasingly associated with the negative evolution of cardiac pathologies. The present study investigates the contribution of neutrophils to VM disease progression in different ways. METHODS AND RESULTS In a coxsackievirus B3- (CVB3) induced mouse model of VM, neutrophils and neutrophil extracellular traps (NETs) were prominent in the acute phase of VM as revealed by enzyme-linked immunosorbent assay analysis and immunostaining. Anti-Ly6G-mediated neutrophil blockade starting at model induction decreased cardiac necrosis and leucocyte infiltration, preventing monocyte and Ly6CHigh pro-inflammatory macrophage recruitment. Furthermore, genetic peptidylarginine deiminase 4-dependent NET blockade reduced cardiac damage and leucocyte recruitment, significantly decreasing cardiac monocyte and macrophage presence. Depleting neutrophils with anti-Ly6G antibodies at 7 days post-infection, after the acute phase, did not decrease cardiac inflammation. CONCLUSION Collectively, these results indicate that the repression of neutrophils and the related NET response in the acute phase of VM improves the pathological phenotype by reducing cardiac inflammation.
Collapse
Affiliation(s)
- Paolo Carai
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- CARIM, Maastricht University, Maastricht, The Netherlands
| | - Laura Florit González
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Cardiology, Experimental Cardiology Laboratory, Utrecht University, Utrecht, The Netherlands
| | - Stijn Van Bruggen
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Valerie Spalart
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Daria De Giorgio
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milan, Italy
| | - Nadéche Geuens
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Elizabeth Anne Vincent Jones
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- CARIM, Maastricht University, Maastricht, The Netherlands
| | - Stephane Heymans
- Centre for Vascular and Molecular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- CARIM, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
81
|
Wigerblad G, Warner SA, Ramos-Benitez MJ, Kardava L, Tian X, Miao R, Reger R, Chakraborty M, Wong S, Kanthi Y, Suffredini AF, Dell’Orso S, Brooks S, King C, Shlobin O, Nathan SD, Cohen J, Moir S, Childs RW, Kaplan MJ, Chertow DS, Strich JR. Spleen tyrosine kinase inhibition restores myeloid homeostasis in COVID-19. SCIENCE ADVANCES 2023; 9:eade8272. [PMID: 36598976 PMCID: PMC9812373 DOI: 10.1126/sciadv.ade8272] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Spleen tyrosine kinase (SYK) is a previously unidentified therapeutic target that inhibits neutrophil and macrophage activation in coronavirus disease 2019 (COVID-19). Fostamatinib, a SYK inhibitor, was studied in a phase 2 placebo-controlled randomized clinical trial and was associated with improvements in many secondary end points related to efficacy. Here, we used a multiomic approach to evaluate cellular and soluble immune mediator responses of patients enrolled in this trial. We demonstrated that SYK inhibition was associated with reduced neutrophil activation, increased circulation of mature neutrophils (CD10+CD33-), and decreased circulation of low-density granulocytes and polymorphonuclear myeloid-derived suppressor cells (HLA-DR-CD33+CD11b-). SYK inhibition was also associated with normalization of transcriptional activity in circulating monocytes relative to healthy controls, an increase in frequency of circulating nonclassical and HLA-DRhi classical monocyte populations, and restoration of interferon responses. Together, these data suggest that SYK inhibition may mitigate proinflammatory myeloid cellular and soluble mediator responses thought to contribute to immunopathogenesis of severe COVID-19.
Collapse
Affiliation(s)
- Gustaf Wigerblad
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD, USA
| | - Seth A. Warner
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Marcos J. Ramos-Benitez
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA
- Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, Bethesda, MD, USA
- Ponce Health Science University and Ponce Research Institute, Department of Basic Sciences, School of Medicine, Ponce, Puerto Rico, USA
| | - Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Rui Miao
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Robert Reger
- Laboratory of Transplantation Immunotherapy, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Mala Chakraborty
- Laboratory of Transplantation Immunotherapy, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Susan Wong
- Laboratory of Transplantation Immunotherapy, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Anthony F. Suffredini
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Stefania Dell’Orso
- Genomic Technology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephen Brooks
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher King
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Oksana Shlobin
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Steven D. Nathan
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Jonathan Cohen
- Adventist Healthcare Shady Grove Medical Center, Rockville, MD, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Richard W. Childs
- Laboratory of Transplantation Immunotherapy, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
- United States Public Health Service Commissioned Corps, Rockville, MD, USA
| | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD, USA
| | - Daniel S. Chertow
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
- United States Public Health Service Commissioned Corps, Rockville, MD, USA
| | - Jeffrey R. Strich
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA
- United States Public Health Service Commissioned Corps, Rockville, MD, USA
| |
Collapse
|
82
|
Hopkins FR, Govender M, Svanberg C, Nordgren J, Waller H, Nilsdotter-Augustinsson Å, Henningsson AJ, Hagbom M, Sjöwall J, Nyström S, Larsson M. Major alterations to monocyte and dendritic cell subsets lasting more than 6 months after hospitalization for COVID-19. Front Immunol 2023; 13:1082912. [PMID: 36685582 PMCID: PMC9846644 DOI: 10.3389/fimmu.2022.1082912] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction After more than two years the Coronavirus disease-19 (COVID-19) pandemic continues to burden healthcare systems and economies worldwide, and it is evident that the effects on the immune system can persist for months post-infection. The activity of myeloid cells such as monocytes and dendritic cells (DC) is essential for correct mobilization of the innate and adaptive responses to a pathogen. Impaired levels and responses of monocytes and DC to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is likely to be a driving force behind the immune dysregulation that characterizes severe COVID-19. Methods Here, we followed a cohort of COVID-19 patients hospitalized during the early waves of the pandemic for 6-7 months. The levels and phenotypes of circulating monocyte and DC subsets were assessed to determine both the early and long-term effects of the SARS-CoV-2 infection. Results We found increased monocyte levels that persisted for 6-7 months, mostly attributed to elevated levels of classical monocytes. Myeloid derived suppressor cells were also elevated over this period. While most DC subsets recovered from an initial decrease, we found elevated levels of cDC2/cDC3 at the 6-7 month timepoint. Analysis of functional markers on monocytes and DC revealed sustained reduction in program death ligand 1 (PD-L1) expression but increased CD86 expression across almost all cell types examined. Finally, C-reactive protein (CRP) correlated positively to the levels of intermediate monocytes and negatively to the recovery of DC subsets. Conclusion By exploring the myeloid compartments, we show here that alterations in the immune landscape remain more than 6 months after severe COVID-19, which could be indicative of ongoing healing and/or persistence of viral antigens.
Collapse
Affiliation(s)
- Francis R. Hopkins
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Melissa Govender
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Cecilia Svanberg
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan Nordgren
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hjalmar Waller
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Åsa Nilsdotter-Augustinsson
- Division of Infection and Inflammation, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Infectious Diseases, Linköping University, Linköping, Sweden
| | - Anna J. Henningsson
- Division of Infection and Inflammation, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Division of Clinical Microbiology, Department of Laboratory Medicine in Jönköping, Ryhov County Hospital, Jönköping, Sweden
| | - Marie Hagbom
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanna Sjöwall
- Division of Infection and Inflammation, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Infectious Diseases, Linköping University, Linköping, Sweden
| | - Sofia Nyström
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Linköping University, Linköping, Sweden
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
83
|
López-Martínez C, Martín-Vicente P, Gómez de Oña J, López-Alonso I, Gil-Peña H, Cuesta-Llavona E, Fernández-Rodríguez M, Crespo I, Salgado Del Riego E, Rodríguez-García R, Parra D, Fernández J, Rodríguez-Carrio J, Jimeno-Demuth FJ, Dávalos A, Chapado LA, Coto E, Albaiceta GM, Amado-Rodríguez L. Transcriptomic clustering of critically ill COVID-19 patients. Eur Respir J 2023; 61:13993003.00592-2022. [PMID: 36104291 PMCID: PMC9478362 DOI: 10.1183/13993003.00592-2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 08/19/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Infections caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may cause a severe disease, termed coronavirus disease 2019 (COVID-19), with significant mortality. Host responses to this infection, mainly in terms of systemic inflammation, have emerged as key pathogenetic mechanisms and their modulation has shown a mortality benefit. METHODS In a cohort of 56 critically ill COVID-19 patients, peripheral blood transcriptomes were obtained at admission to an intensive care unit (ICU) and clustered using an unsupervised algorithm. Differences in gene expression, circulating microRNAs (c-miRNAs) and clinical data between clusters were assessed, and circulating cell populations estimated from sequencing data. A transcriptomic signature was defined and applied to an external cohort to validate the findings. RESULTS We identified two transcriptomic clusters characterised by expression of either interferon-related or immune checkpoint genes, respectively. Steroids have cluster-specific effects, decreasing lymphocyte activation in the former but promoting B-cell activation in the latter. These profiles have different ICU outcomes, despite no major clinical differences at ICU admission. A transcriptomic signature was used to identify these clusters in two external validation cohorts (with 50 and 60 patients), yielding similar results. CONCLUSIONS These results reveal different underlying pathogenetic mechanisms and illustrate the potential of transcriptomics to identify patient endotypes in severe COVID-19 with the aim to ultimately personalise their therapies.
Collapse
Affiliation(s)
- Cecilia López-Martínez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Madrid, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Paula Martín-Vicente
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Madrid, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Juan Gómez de Oña
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Servicio de Genética Molecular, Hospital Universitario Central de Asturias, Oviedo, Spain
- Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Inés López-Alonso
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Madrid, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain
| | - Helena Gil-Peña
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Servicio de Pediatría, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Elías Cuesta-Llavona
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Servicio de Genética Molecular, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Margarita Fernández-Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Irene Crespo
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Madrid, Spain
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | - Estefanía Salgado Del Riego
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Unidad de Cuidados Intensivos Polivalente, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Raquel Rodríguez-García
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Madrid, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Diego Parra
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Javier Fernández
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Servicio de Microbiología, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Javier Rodríguez-Carrio
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | | | - Alberto Dávalos
- Instituto Madrileño de Estudios Avanzados (IMDEA) Alimentación, CEI UAM+CSIC, Madrid, Spain
| | - Luis A Chapado
- Instituto Madrileño de Estudios Avanzados (IMDEA) Alimentación, CEI UAM+CSIC, Madrid, Spain
| | - Eliecer Coto
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Servicio de Genética Molecular, Hospital Universitario Central de Asturias, Oviedo, Spain
- Red de Investigación Renal (REDINREN), Madrid, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Guillermo M Albaiceta
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Madrid, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
- G.M. Albaiceta and L. Amado-Rodríguez share last authorship
| | - Laura Amado-Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Madrid, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
- G.M. Albaiceta and L. Amado-Rodríguez share last authorship
| |
Collapse
|
84
|
Van Bruggen S, Martinod K. The coming of age of neutrophil extracellular traps in thrombosis: Where are we now and where are we headed? Immunol Rev 2022; 314:376-398. [PMID: 36560865 DOI: 10.1111/imr.13179] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thrombosis remains a major problem in our society, manifesting across multiple demographic groups and with high associated morbidity and mortality. Thrombus development is the result of a complex mechanism in which multiple cell types and soluble factors play a crucial role. One cell that has gained the most attention in recent years is the neutrophil. This key member of the innate immune system can form neutrophil extracellular traps (NETs) in response to activating stimuli in circulation. NETs form a scaffold for thrombus formation, both initiating the process and stabilizing the final product. As the first responders of the host immune system, neutrophils have the flexibility to recognize a variety of molecules and can quickly interact with a range of different cell types. This trait makes them sensitive to exogenous stimuli. NET formation in response to pathogens is well established, leading to immune-mediated thrombus formation or immunothrombosis. NETs can also be formed during sterile inflammation through the activation of neutrophils by fellow immune cells including platelets, or activated endothelium. In chronic inflammatory settings, NETs can ultimately promote the development of tissue fibrosis, with organ failure as an end-stage outcome. In this review, we discuss the different pathways through which neutrophils can be activated toward NET formation and how these processes can result in a shared outcome: thrombus formation. Finally, we evaluate these different interactions and mechanisms for their potential as therapeutic targets, with neutrophil-targeted therapies providing a future approach to treating thrombosis. In contrast to current practices, such treatment could result in reduced pathogenic blood clot formation without increasing the risk of bleeding.
Collapse
Affiliation(s)
- Stijn Van Bruggen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
85
|
Bosteels C, Van Damme KFA, De Leeuw E, Declercq J, Maes B, Bosteels V, Hoste L, Naesens L, Debeuf N, Deckers J, Cole B, Pardons M, Weiskopf D, Sette A, Weygaerde YV, Malfait T, Vandecasteele SJ, Demedts IK, Slabbynck H, Allard S, Depuydt P, Van Braeckel E, De Clercq J, Martens L, Dupont S, Seurinck R, Vandamme N, Haerynck F, Roychowdhury DF, Vandekerckhove L, Guilliams M, Tavernier SJ, Lambrecht BN. Loss of GM-CSF-dependent instruction of alveolar macrophages in COVID-19 provides a rationale for inhaled GM-CSF treatment. Cell Rep Med 2022; 3:100833. [PMID: 36459994 PMCID: PMC9663750 DOI: 10.1016/j.xcrm.2022.100833] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/12/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022]
Abstract
GM-CSF promotes myelopoiesis and inflammation, and GM-CSF blockade is being evaluated as a treatment for COVID-19-associated hyperinflammation. Alveolar GM-CSF is, however, required for monocytes to differentiate into alveolar macrophages (AMs) that control alveolar homeostasis. By mapping cross-species AM development to clinical lung samples, we discovered that COVID-19 is marked by defective GM-CSF-dependent AM instruction and accumulation of pro-inflammatory macrophages. In a multi-center, open-label RCT in 81 non-ventilated COVID-19 patients with respiratory failure, we found that inhalation of rhu-GM-CSF did not improve mean oxygenation parameters compared with standard treatment. However, more patients on GM-CSF had a clinical response, and GM-CSF inhalation induced higher numbers of virus-specific CD8 effector lymphocytes and class-switched B cells, without exacerbating systemic hyperinflammation. This translational proof-of-concept study provides a rationale for further testing of inhaled GM-CSF as a non-invasive treatment to improve alveolar gas exchange and simultaneously boost antiviral immunity in COVID-19. This study is registered at ClinicalTrials.gov (NCT04326920) and EudraCT (2020-001254-22).
Collapse
Affiliation(s)
- Cedric Bosteels
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Karel F A Van Damme
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Elisabeth De Leeuw
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Jozefien Declercq
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Bastiaan Maes
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Victor Bosteels
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Laboratory of ER Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium
| | - Levi Hoste
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Primary Immunodeficiency Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Leslie Naesens
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Primary Immunodeficiency Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Nincy Debeuf
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Julie Deckers
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Basiel Cole
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Marion Pardons
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Daniela Weiskopf
- Center for Autoimmunity and Inflammation and Center for Infectious Diseases and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Autoimmunity and Inflammation and Center for Infectious Diseases and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | | | - Thomas Malfait
- Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | | | - Ingel K Demedts
- Department of Pulmonary Medicine, AZ Delta General Hospital, 8800 Roeselare, Belgium
| | - Hans Slabbynck
- Department of Pulmonary Medicine, ZNA General Hospital, 2000 Antwerp, Belgium
| | - Sabine Allard
- Department of Internal Medicine, Universitair Ziekenhuis Brussel, 1000 Brussels, Belgium
| | - Pieter Depuydt
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Intensive Care Unit, Ghent University Hospital, 9000 Ghent, Belgium
| | - Eva Van Braeckel
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Jozefien De Clercq
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Infectious Diseases, Ghent University Hospital, 9000 Ghent, Belgium
| | - Liesbet Martens
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, 9000 Ghent, Belgium
| | - Sam Dupont
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Ruth Seurinck
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, 9000 Ghent, Belgium; Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000 Ghent, Belgium
| | - Niels Vandamme
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, 9000 Ghent, Belgium; VIB Single Cell Core, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Primary Immunodeficiency Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | | | - Linos Vandekerckhove
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Infectious Diseases, Ghent University Hospital, 9000 Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, 9000 Ghent, Belgium
| | - Simon J Tavernier
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Primary Immunodeficiency Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium; Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, 9000 Ghent, Belgium; Laboratory of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| |
Collapse
|
86
|
Gindlhuber J, Tomin T, Wiesenhofer F, Zacharias M, Liesinger L, Demichev V, Kratochwill K, Gorkiewicz G, Schittmayer M, Birner-Gruenberger R. Proteomic profiling of end-stage COVID-19 lung biopsies. Clin Proteomics 2022; 19:46. [PMID: 36526981 PMCID: PMC9758034 DOI: 10.1186/s12014-022-09386-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
The outbreak of a novel coronavirus (SARS-CoV-2) in 2019 led to a worldwide pandemic, which remains an integral part of our lives to this day. Coronavirus disease (COVID-19) is a flu like condition, often accompanied by high fever and respiratory distress. In some cases, conjointly with other co-morbidities, COVID-19 can become severe, leading to lung arrest and even death. Although well-known from a clinical standpoint, the mechanistic understanding of lethal COVID-19 is still rudimentary. Studying the pathology and changes on a molecular level associated with the resulting COVID-19 disease is impeded by the highly infectious nature of the virus and the concomitant sampling challenges. We were able to procure COVID-19 post-mortem lung tissue specimens by our collaboration with the BSL-3 laboratory of the Biobanking and BioMolecular resources Research Infrastructure Austria which we subjected to state-of-the-art quantitative proteomic analysis to better understand the pulmonary manifestations of lethal COVID-19. Lung tissue samples from age-matched non-COVID-19 patients who died within the same period were used as controls. Samples were subjected to parallel accumulation-serial fragmentation combined with data-independent acquisition (diaPASEF) on a timsTOF Pro and obtained raw data was processed using DIA-NN software. Here we report that terminal COVID-19 patients display an increase in inflammation, acute immune response and blood clot formation (with concomitant triggering of fibrinolysis). Furthermore, we describe that COVID-19 diseased lungs undergo severe extracellular matrix restructuring, which was corroborated on the histopathological level. However, although undergoing an injury, diseased lungs seem to have impaired proliferative and tissue repair signalling, with several key kinase-mediated signalling pathways being less active. This might provide a mechanistic link to post-acute sequelae of COVID-19 (PASC; "Long COVID"). Overall, we emphasize the importance of histopathological patient stratification when interpreting molecular COVID-19 data.
Collapse
Affiliation(s)
- Juergen Gindlhuber
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Tamara Tomin
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
| | - Florian Wiesenhofer
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Martin Zacharias
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Laura Liesinger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Vadim Demichev
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Gregor Gorkiewicz
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Matthias Schittmayer
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria.
| | - Ruth Birner-Gruenberger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria.
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria.
| |
Collapse
|
87
|
Ghosh N, Saha I, Plewczynski D. Unveiling the Biomarkers of Cancer and COVID-19 and Their Regulations in Different Organs by Integrating RNA-Seq Expression and Protein-Protein Interactions. ACS OMEGA 2022; 7:43589-43602. [PMID: 36506181 PMCID: PMC9730762 DOI: 10.1021/acsomega.2c04389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 06/17/2023]
Abstract
Cancer and COVID-19 have killed millions of people worldwide. COVID-19 is even more dangerous to people with comorbidities such as cancer. Thus, it is imperative to identify the key human genes or biomarkers that can be targeted to develop novel prognosis and therapeutic strategies. The transcriptomic data provided by the next-generation sequencing technique makes this identification very convenient. Hence, mRNA (messenger ribonucleic acid) expression data of 2265 cancer and 282 normal patients were considered, while for COVID-19 assessment, 784 and 425 COVID-19 and normal patients were taken, respectively. Initially, volcano plots were used to identify the up- and down-regulated genes for both cancer and COVID-19. Thereafter, protein-protein interaction (PPI) networks were prepared by combining all the up- and down-regulated genes for each of cancer and COVID-19. Subsequently, such networks were analyzed to identify the top 10 genes with the highest degree of connection to provide the biomarkers. Interestingly, these genes were all up-regulated for cancer, while they were down-regulated for COVID-19. This study had also identified common genes between cancer and COVID-19, all of which were up-regulated in both the diseases. This analysis revealed that FN1 was highly up-regulated in different organs for cancer, while EEF2 was dysregulated in most organs affected by COVID-19. Then, functional enrichment analysis was performed to identify significant biological processes. Finally, the drugs for cancer and COVID-19 biomarkers and the common genes between them were identified using the Enrichr online web tool. These drugs include lucanthone, etoposide, and methotrexate, targeting the biomarkers for cancer, while paclitaxel is an important drug for COVID-19.
Collapse
Affiliation(s)
- Nimisha Ghosh
- Faculty
of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw 02-097, Poland
- Department
of Computer Science and Information Technology, Institute of Technical
Education and Research, Siksha ‘O’
Anusandhan (Deemed to Be University), Bhubaneswar 751030 Odisha, India
| | - Indrajit Saha
- Department
of Computer Science and Engineering, National
Institute of Technical Teachers’ Training and Research, Kolkata 700106 West Bengal, India
| | - Dariusz Plewczynski
- Laboratory
of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw 02-097, Poland
- Laboratory
of Bioinformatics and Computational Genomics, Faculty of Mathematics
and Information Science, Warsaw University
of Technology, Warsaw 00-662, Poland
| |
Collapse
|
88
|
Chattopadhyay P, Khare K, Kumar M, Mishra P, Anand A, Maurya R, Gupta R, Sahni S, Gupta A, Wadhwa S, Yadav A, Devi P, Tardalkar K, Joshi M, Sethi T, Pandey R. Single-cell multiomics revealed the dynamics of antigen presentation, immune response and T cell activation in the COVID-19 positive and recovered individuals. Front Immunol 2022; 13:1034159. [PMID: 36532041 PMCID: PMC9755500 DOI: 10.3389/fimmu.2022.1034159] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Despite numerous efforts to describe COVID-19's immunological landscape, there is still a gap in our understanding of the virus's infections after-effects, especially in the recovered patients. This would be important to understand as we now have huge number of global populations infected by the SARS-CoV-2 as well as variables inclusive of VOCs, reinfections, and vaccination breakthroughs. Furthermore, single-cell transcriptome alone is often insufficient to understand the complex human host immune landscape underlying differential disease severity and clinical outcome. Methods By combining single-cell multi-omics (Whole Transcriptome Analysis plus Antibody-seq) and machine learning-based analysis, we aim to better understand the functional aspects of cellular and immunological heterogeneity in the COVID-19 positive, recovered and the healthy individuals. Results Based on single-cell transcriptome and surface marker study of 163,197 cells (124,726 cells after data QC) from the 33 individuals (healthy=4, COVID-19 positive=16, and COVID-19 recovered=13), we observed a reduced MHC Class-I-mediated antigen presentation and dysregulated MHC Class-II-mediated antigen presentation in the COVID-19 patients, with restoration of the process in the recovered individuals. B-cell maturation process was also impaired in the positive and the recovered individuals. Importantly, we discovered that a subset of the naive T-cells from the healthy individuals were absent from the recovered individuals, suggesting a post-infection inflammatory stage. Both COVID-19 positive patients and the recovered individuals exhibited a CD40-CD40LG-mediated inflammatory response in the monocytes and T-cell subsets. T-cells, NK-cells, and monocyte-mediated elevation of immunological, stress and antiviral responses were also seen in the COVID-19 positive and the recovered individuals, along with an abnormal T-cell activation, inflammatory response, and faster cellular transition of T cell subtypes in the COVID-19 patients. Importantly, above immune findings were used for a Bayesian network model, which significantly revealed FOS, CXCL8, IL1β, CST3, PSAP, CD45 and CD74 as COVID-19 severity predictors. Discussion In conclusion, COVID-19 recovered individuals exhibited a hyper-activated inflammatory response with the loss of B cell maturation, suggesting an impeded post-infection stage, necessitating further research to delineate the dynamic immune response associated with the COVID-19. To our knowledge this is first multi-omic study trying to understand the differential and dynamic immune response underlying the sample subtypes.
Collapse
Affiliation(s)
- Partha Chattopadhyay
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kriti Khare
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manish Kumar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Pallavi Mishra
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Alok Anand
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Ranjeet Maurya
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rohit Gupta
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Shweta Sahni
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Ayushi Gupta
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Saruchi Wadhwa
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Aanchal Yadav
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Priti Devi
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kishore Tardalkar
- Department of Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Meghnad Joshi
- Department of Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Tavpritesh Sethi
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Rajesh Pandey
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
89
|
Mohan AA, Olson LB, Naqvi IA, Morrison SA, Kraft BD, Chen L, Que LG, Ma Q, Barkauskas CE, Kirk A, Nair SK, Sullenger BA, Kasotakis G. Age and Comorbidities Predict COVID-19 Outcome, Regardless of Innate Immune Response Severity: A Single Institutional Cohort Study. Crit Care Explor 2022; 4:e0799. [PMID: 36506827 PMCID: PMC9726311 DOI: 10.1097/cce.0000000000000799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic has claimed over eight hundred thousand lives in the United States alone, with older individuals and those with comorbidities being at higher risk of severe disease and death. Although severe acute respiratory syndrome coronavirus 2-induced hyperinflammation is one of the mechanisms underlying the high mortality, the association between age and innate immune responses in COVID-19 mortality remains unclear. DESIGN Flow cytometry of fresh blood and multiplexed inflammatory chemokine measurements of sera were performed on samples collected longitudinally from our cohort. Aggregate impact of comorbid conditions was calculated with the Charlson Comorbidity Index, and association between patient factors and outcomes was calculated via Cox proportional hazard analysis and repeated measures analysis of variance. SETTING A cohort of severely ill COVID-19 patients requiring ICU admission was followed prospectively. PATIENTS In total, 67 patients (46 male, age 59 ± 14 yr) were included in the study. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Mortality in our cohort was 41.8%. We identified older age (hazard ratio [HR] 1.09 [95% CI 1.07-1.11]; p = 0.001), higher comorbidity index (HR 1.24 [95% CI 1.14-1.35]; p = 0.039), and hyponatremia (HR 0.90 [95% CI 0.82-0.99]; p = 0.026) to each independently increase risk for death in COVID-19. We also found that neutrophilia (R = 0.2; p = 0.017), chemokine C-C motif ligand (CCL) 2 (R = 0.3; p = 0.043), and C-X-C motif chemokine ligand 9 (CXCL9) (R = 0.3; p = 0.050) were weakly but significantly correlated with mortality. Older age was associated with lower monocyte (R = -0.2; p = 0.006) and cluster of differentiation (CD) 16+ cell counts (R = -0.2; p = 0.002) and increased CCL11 concentration (R = 0.3; p = 0.050). Similarly, younger patients (< 65 yr) demonstrated a rise in CD4 (b-coefficient = 0.02; p = 0.036) and CD8 (0.01; p = 0.001) counts, as well as CCL20 (b-coefficient = 6.8; p = 0.036) during their ICU stay. This CD8 count rise was also associated with survival (b-coefficient = 0.01; p = 0.023). CONCLUSIONS Age, comorbidities, and hyponatremia independently predict mortality in severe COVID-19. Neutrophilia and higher CCL2 and CXCL9 levels are also associated with higher mortality, while independent of age.
Collapse
Affiliation(s)
| | - Lyra B Olson
- Department of Surgery, Duke University, Durham, NC
| | | | | | | | - Lingye Chen
- Department of Surgery, Duke University, Durham, NC
| | | | - Qing Ma
- Department of Surgery, Duke University, Durham, NC
| | | | - Allan Kirk
- Department of Surgery, Duke University, Durham, NC
| | - Smita K Nair
- Department of Surgery, Duke University, Durham, NC
- Departments of Neurosurgery and Pathology, Duke University, Durham, NC
| | - Bruce A Sullenger
- Department of Surgery, Duke University, Durham, NC
- Departments of Neurosurgery and Pathology, Duke University, Durham, NC
- Departments of Neurosurgery, Pharmacology, and Cancer Biology, Duke University, Durham, NC
| | | |
Collapse
|
90
|
Ruenjaiman V, Sodsai P, Kueanjinda P, Bunrasmee W, Klinchanhom S, Reantragoon R, Tunvirachaisakul C, Manothummetha K, Mejun N, Liengswangwong K, Torvorapanit P, Paitoonpong L, Putcharoen O, Palaga T, Hirankarn N, study team 2TungwongkitsiriAbhichayaMittrakulkijChanyaChiewbangyangFarsaiKaewsrihawongJanistaSanpakitJirayuKulkiatprasertKanokphetMunkongKhemmachatKeawthawonNanthidaWattanakulNatchanonLimchanachonNatdanaiRoopsuwankunNatthapatChaosuwannakijNatthasiniLarpanekananPasinPitakkitnukunPawitHomswadPongponRatanapraisornSamapitchAtchariyapakornSarunyapongVongphanichSasathamonJessadapornchaiSirapatAvihingsanonTetonPiyasathapornpongThanatorn, Mittrakulkij C, Chiewbangyang F, Kaewsrihawong J, Sanpakit J, Kulkiatprasert K, Munkong K, Keawthawon N, Wattanakul N, Limchanachon N, Roopsuwankun N, Chaosuwannakij N, Larpanekanan P, Pitakkitnukun P, Homswad P, Ratanapraisorn S, Atchariyapakorn S, Vongphanich S, Jessadapornchai S, Avihingsanon T, Piyasathapornpong T. Impact of SARS-CoV-2 infection on the profiles and responses of innate immune cells after recovery. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:993-1004. [PMID: 36220753 PMCID: PMC9519362 DOI: 10.1016/j.jmii.2022.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/30/2022] [Accepted: 09/24/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUNDS SARS-CoV-2 infection results in a broad spectrum of clinical outcomes, ranging from asymptomatic to severe symptoms and death. Most COVID-19 pathogenesis is associated with hyperinflammatory conditions driven primarily by myeloid cell lineages. The long-term effects of SARS-CoV-2 infection post recovery include various symptoms. METHODS We performed a longitudinal study of the innate immune profiles 1 and 3 months after recovery in the Thai cohort by comparing patients with mild, moderate, and severe clinical symptoms using peripheral blood mononuclear cells (n = 62). RESULTS Significant increases in the frequencies of monocytes compared to controls and NK cells compared to mild and moderate patients were observed in severe patients 1-3 months post recovery. Increased polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were observed in all recovered patients, even after 3 months. Increased IL-6 and TNFα levels in monocytes were observed 1 month after recovery in response to lipopolysaccharide (LPS) stimulation, while decreased CD86 and HLA-DR levels were observed regardless of stimulation. A multiplex analysis of serum cytokines performed at 1 month revealed that most innate cytokines, except for TNFα, IL4/IL-13 (Th2) and IFNγ (Th1), were elevated in recovered patients in a severity-dependent manner. Finally, the myelopoiesis cytokines G-CSF and GM-CSF were higher in all patient groups. Increased monocytes and IL-6- and TNFα-producing cells were significantly associated with long COVID-19 symptoms. CONCLUSIONS These results reveal that COVID-19 infection influences the frequencies and functions of innate immune cells for up to 3 months after recovery, which may potentially lead to some of the long COVID symptoms.
Collapse
Affiliation(s)
- Vichaya Ruenjaiman
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pimpayao Sodsai
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Patipark Kueanjinda
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Worawan Bunrasmee
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Siriwan Klinchanhom
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rangsima Reantragoon
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | | | - Kasama Manothummetha
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nuthchaya Mejun
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kaewkwan Liengswangwong
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pattama Torvorapanit
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Leilani Paitoonpong
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Opass Putcharoen
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tanapat Palaga
- Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand,Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand,Corresponding author. Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Nierenberg D, Flores O, Fox D, Sip YYL, Finn CM, Ghozlan H, Cox A, Coathup M, McKinstry KK, Zhai L, Khaled AR. Macromolecules Absorbed from Influenza Infection-Based Sera Modulate the Cellular Uptake of Polymeric Nanoparticles. Biomimetics (Basel) 2022; 7:biomimetics7040219. [PMID: 36546919 PMCID: PMC9775140 DOI: 10.3390/biomimetics7040219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
Optimizing the biological identity of nanoparticles (NPs) for efficient tumor uptake remains challenging. The controlled formation of a protein corona on NPs through protein absorption from biofluids could favor a biological identity that enables tumor accumulation. To increase the diversity of proteins absorbed by NPs, sera derived from Influenza A virus (IAV)-infected mice were used to pre-coat NPs formed using a hyperbranched polyester polymer (HBPE-NPs). HBPE-NPs, encapsulating a tracking dye or cancer drug, were treated with sera from days 3-6 of IAV infection (VS3-6), and uptake of HBPE-NPs by breast cancer cells was examined. Cancer cells demonstrated better uptake of HBPE-NPs pre-treated with VS3-6 over polyethylene glycol (PEG)-HBPE-NPs, a standard NP surface modification. The uptake of VS5 pre-treated HBPE-NPs by monocytic cells (THP-1) was decreased over PEG-HBPE-NPs. VS5-treated HBPE-NPs delivered a cancer drug more efficiently and displayed better in vivo distribution over controls, remaining stable even after interacting with endothelial cells. Using a proteomics approach, proteins absorbed from sera-treated HBPE-NPs were identified, such as thrombospondin-1 (TSP-1), that could bind multiple cancer cell receptors. Our findings indicate that serum collected during an immune response to infection is a rich source of macromolecules that are absorbed by NPs and modulate their biological identity, achieving rationally designed uptake by targeted cell types.
Collapse
Affiliation(s)
- Daniel Nierenberg
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Orielyz Flores
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - David Fox
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
- Department of Chemistry, College of Science, University of Central Florida, Orlando, FL 32816, USA
| | - Yuen Yee Li Sip
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
- Department of Materials Science and Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, FL 32816, USA
| | - Caroline M. Finn
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Heba Ghozlan
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Amanda Cox
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Melanie Coathup
- Biionix Cluster and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Karl Kai McKinstry
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
| | - Lei Zhai
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
- Department of Chemistry, College of Science, University of Central Florida, Orlando, FL 32816, USA
- Department of Materials Science and Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, FL 32816, USA
| | - Annette R. Khaled
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
- Correspondence: ; Tel.: +1-407-266-7035
| |
Collapse
|
92
|
Mortezaee K, Majidpoor J. Cellular immune states in SARS-CoV-2-induced disease. Front Immunol 2022; 13:1016304. [PMID: 36505442 PMCID: PMC9726761 DOI: 10.3389/fimmu.2022.1016304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
The general immune state plays important roles against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Cells of the immune system are encountering rapid changes during the acute phase of SARS-CoV-2-induced disease. Reduced fraction of functional CD8+ T cells, disrupted cross-talking between CD8+ T cells with dendritic cells (DCs), and impaired immunological T-cell memory, along with the higher presence of hyperactive neutrophils, high expansion of myeloid-derived suppressor cells (MDSCs) and non-classical monocytes, and attenuated cytotoxic capacity of natural killer (NK) cells, are all indicative of low efficient immunity against viral surge within the body. Immune state and responses from pro- or anti-inflammatory cells of the immune system to SARS-CoV-2 are discussed in this review. We also suggest some strategies to enhance the power of immune system against SARS-CoV-2-induced disease.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran,*Correspondence: Keywan Mortezaee, ;
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
93
|
Jiang ST, Liu YG, Zhang L, Sang XT, Xu YY, Lu X. Systems biology approach reveals a common molecular basis for COVID-19 and non-alcoholic fatty liver disease (NAFLD). Eur J Med Res 2022; 27:251. [PMCID: PMC9664052 DOI: 10.1186/s40001-022-00865-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background
Patients with non-alcoholic fatty liver disease (NAFLD) may be more susceptible to coronavirus disease 2019 (COVID-19) and even more likely to suffer from severe COVID-19. Whether there is a common molecular pathological basis for COVID-19 and NAFLD remains to be identified. The present study aimed to elucidate the transcriptional alterations shared by COVID-19 and NAFLD and to identify potential compounds targeting both diseases.
Methods
Differentially expressed genes (DEGs) for COVID-19 and NAFLD were extracted from the GSE147507 and GSE89632 datasets, and common DEGs were identified using the Venn diagram. Subsequently, we constructed a protein–protein interaction (PPI) network based on the common DEGs and extracted hub genes. Then, we performed gene ontology (GO) and pathway analysis of common DEGs. In addition, transcription factors (TFs) and miRNAs regulatory networks were constructed, and drug candidates were identified.
Results
We identified a total of 62 common DEGs for COVID-19 and NAFLD. The 10 hub genes extracted based on the PPI network were IL6, IL1B, PTGS2, JUN, FOS, ATF3, SOCS3, CSF3, NFKB2, and HBEGF. In addition, we also constructed TFs–DEGs, miRNAs–DEGs, and protein–drug interaction networks, demonstrating the complex regulatory relationships of common DEGs.
Conclusion
We successfully extracted 10 hub genes that could be used as novel therapeutic targets for COVID-19 and NAFLD. In addition, based on common DEGs, we propose some potential drugs that may benefit patients with COVID-19 and NAFLD.
Collapse
|
94
|
Hanata N, Ota M, Tsuchida Y, Nagafuchi Y, Okamura T, Shoda H, Fujio K. Serum extracellular traps associate with the activation of myeloid cells in SLE patients with the low level of anti-DNA antibodies. Sci Rep 2022; 12:18397. [PMID: 36319843 PMCID: PMC9626644 DOI: 10.1038/s41598-022-23076-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are involved in systemic lupus erythematosus (SLE). We sought to cluster SLE patients based on serum NET levels. Serum NET levels were higher in SLE patients than healthy controls. Frequencies of pleuritis and myositis were increased in patients with high serum NET levels. Serum NET levels negatively correlated with anti-double stranded DNA (anti-dsDNA) antibody titers and C1q-binding immune complexes, but positively correlated with C-reactive protein (CRP) and monocyte counts. Neutrophil transcriptome analysis demonstrated no difference in NET-associated signatures, irrespective of serum NET levels, suggesting anti-dsDNA antibody-mediated clearance of NETs. In serum, NET levels were significantly correlated with myeloid cell-derived inflammatory molecules. Serum NET-based cluster analysis revealed 3 groups of patients based on serum NET and CRP levels, anti-dsDNA antibody titers, and monocyte count. Monocytes were consistently activated following NET-containing immune complex (NET-IC) stimulation. In conclusion, SLE patients with high serum NET levels had lower anti-dsDNA antibody titers and higher inflammatory responses. NET-IC-stimulated monocytes might associate with an inflammatory response characterized by elevated CRP levels. These findings can apply to precision medicine, as inflammatory processes, rather than antibody-dependent processes, can be targeted in specific subpopulations of SLE patients.
Collapse
Affiliation(s)
- Norio Hanata
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mineto Ota
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumi Tsuchida
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Okamura
- grid.26999.3d0000 0001 2151 536XDepartment of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Shoda
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
95
|
Rossi E, Mutti L, Morrione A, Giordano A. Neuro-Immune Interactions in Severe COVID-19 Infection. Pathogens 2022; 11:1256. [PMID: 36365007 PMCID: PMC9699641 DOI: 10.3390/pathogens11111256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a new coronavirus that has affected the world since 2019. Interstitial pneumonia is the most common clinical presentation, but additional symptoms have been reported, including neurological manifestations. Severe forms of infection, especially in elderly patients, present as an excessive inflammatory response called "cytokine storm", which can lead to acute respiratory distress syndrome (ARDS), multiorgan failure and death. Little is known about the relationship between symptoms and clinical outcomes or the characteristics of virus-host interactions. The aim of this narrative review is to highlight possible links between neurological involvement and respiratory damage mediated by pathological inflammatory pathways in SARS-CoV-2 infection. We will focus on neuro-immune interactions and age-related immunity decline and discuss some pathological mechanisms that contribute to negative outcomes in COVID-19 patients. Furthermore, we will describe available therapeutic strategies and their effects on COVID-19 neurological symptoms.
Collapse
Affiliation(s)
- Elena Rossi
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Italian Group for Research and Therapy for Mesothelioma (GIMe), 27058 Voghera, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
96
|
Lebourgeois S, David A, Chenane HR, Granger V, Menidjel R, Fidouh N, Noël B, Delelis O, Richetta C, Charpentier C, Chollet-Martin S, Descamps D, Visseaux B, de Chaisemartin L. Differential activation of human neutrophils by SARS-CoV-2 variants of concern. Front Immunol 2022; 13:1010140. [PMID: 36389717 PMCID: PMC9646985 DOI: 10.3389/fimmu.2022.1010140] [Citation(s) in RCA: 12] [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: 08/02/2022] [Accepted: 10/14/2022] [Indexed: 07/22/2023] Open
Abstract
The emerging SARS-CoV-2 virus has affected the entire world with over 600 million confirmed cases and 6.5 million deaths as of September 2022. Since the beginning of the pandemic, several variants of SARS-CoV-2 have emerged, with different infectivity and virulence. Several studies suggest an important role of neutrophils in SARS-Cov-2 infection severity, but data about direct activation of neutrophils by the virus is scarce. Here, we studied the in vitro activation of human neutrophils by SARS-CoV-2 variants of concern (VOCs). In our work, we show that upon stimulation with SARS-Cov-2 infectious particles, human healthy resting neutrophils upregulate activation markers, degranulate IL-8, produce Reactive Oxygen Species and release Neutrophil Extracellular Traps. Neutrophil activation was dependent on TLR7/8 and IRF3/STING. We then compared the activation potential of neutrophils by SARS-CoV-2 variants and showed a significantly increased activation by the Delta variant and a decreased activation by the Omicron variant as compared to the initial strain. In this study, we demonstrate that the SARS-Cov-2 virus can directly activate neutrophils in COVID-19 and that the different VOCs had differences in neutrophil activation intensity that mirror the differences of clinical severity. These data highlight the need to address neutrophil-virus interactions as a potential target for therapeutic intervention in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Samuel Lebourgeois
- Université Paris Cité, Infection Antimicrobials Modelling Evolution (IAME), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Ambroise David
- Assistance Publique - Hôpitaux de Paris (AP-HP), University Hospital Bichat-Claude Bernard, Laboratoire d’Immunologie, Paris, France
| | - Houssem Redha Chenane
- Université Paris Cité, Infection Antimicrobials Modelling Evolution (IAME), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Vanessa Granger
- Assistance Publique - Hôpitaux de Paris (AP-HP), University Hospital Bichat-Claude Bernard, Laboratoire d’Immunologie, Paris, France
- Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale (INSERM), Châtenay-Malabry, France
| | - Reyene Menidjel
- Université Paris Cité, Infection Antimicrobials Modelling Evolution (IAME), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Nadhira Fidouh
- Assistance Publique - Hôpitaux de Paris (AP-HP), University Hospital Bichat-Claude Bernard, Laboratoire de Virologie, Paris, France
| | - Benoît Noël
- Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale (INSERM), Châtenay-Malabry, France
| | - Olivier Delelis
- LBPA-Laboratoire Biologie Pharmacologie Appliquée, Ecole Normal Supérieur (ENS) Paris-Saclay, Centre National de la Recherche Scientifique (CNRS) Unité Mix de Recherche (UMR), Université Paris-Saclay, Gif-sur-yvette, France
| | - Clémence Richetta
- LBPA-Laboratoire Biologie Pharmacologie Appliquée, Ecole Normal Supérieur (ENS) Paris-Saclay, Centre National de la Recherche Scientifique (CNRS) Unité Mix de Recherche (UMR), Université Paris-Saclay, Gif-sur-yvette, France
| | - Charlotte Charpentier
- Université Paris Cité, Infection Antimicrobials Modelling Evolution (IAME), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), University Hospital Bichat-Claude Bernard, Laboratoire de Virologie, Paris, France
| | - Sylvie Chollet-Martin
- Assistance Publique - Hôpitaux de Paris (AP-HP), University Hospital Bichat-Claude Bernard, Laboratoire d’Immunologie, Paris, France
- Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale (INSERM), Châtenay-Malabry, France
| | - Diane Descamps
- Université Paris Cité, Infection Antimicrobials Modelling Evolution (IAME), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), University Hospital Bichat-Claude Bernard, Laboratoire de Virologie, Paris, France
| | - Benoit Visseaux
- Université Paris Cité, Infection Antimicrobials Modelling Evolution (IAME), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), University Hospital Bichat-Claude Bernard, Laboratoire de Virologie, Paris, France
| | - Luc de Chaisemartin
- Assistance Publique - Hôpitaux de Paris (AP-HP), University Hospital Bichat-Claude Bernard, Laboratoire d’Immunologie, Paris, France
- Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale (INSERM), Châtenay-Malabry, France
| |
Collapse
|
97
|
Potere N, Del Buono MG, Caricchio R, Cremer PC, Vecchié A, Porreca E, Dalla Gasperina D, Dentali F, Abbate A, Bonaventura A. Interleukin-1 and the NLRP3 inflammasome in COVID-19: Pathogenetic and therapeutic implications. EBioMedicine 2022; 85:104299. [PMID: 36209522 PMCID: PMC9536001 DOI: 10.1016/j.ebiom.2022.104299] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/01/2022] [Accepted: 09/16/2022] [Indexed: 11/11/2022] Open
Abstract
A hyperinflammatory response during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection crucially worsens clinical evolution of coronavirus disease 2019 (COVID-19). The interaction between SARS-CoV-2 and angiotensin-converting enzyme 2 (ACE2) triggers the activation of the NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome. Enhanced inflammasome activity has been associated with increased disease severity and poor prognosis. Evidence suggests that inflammasome activation and interleukin-1β (IL-1β) release aggravate pulmonary injury and induce hypercoagulability, favoring progression to respiratory failure and widespread thrombosis eventually leading to multiorgan failure and death. Observational studies with the IL-1 blockers anakinra and canakinumab provided promising results. In the SAVE-MORE trial, early treatment with anakinra significantly shortened hospital stay and improved survival in patients with moderate-to-severe COVID-19. In this review, we summarize current evidence supporting the pathogenetic role of the NLRP3 inflammasome and IL-1β in COVID-19, and discuss clinical trials testing IL-1 inhibition in COVID-19.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medicine and Ageing Sciences and Department of Innovative Technologies in Medicine and Dentistry, G. D'Annunzio University, Chieti, Italy
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Paul C. Cremer
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alessandra Vecchié
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | - Ettore Porreca
- Department of Medicine and Ageing Sciences and Department of Innovative Technologies in Medicine and Dentistry, G. D'Annunzio University, Chieti, Italy
| | | | - Francesco Dentali
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi, Varese, Italy,Corresponding author.
| |
Collapse
|
98
|
Malengier-Devlies B, Filtjens J, Ahmadzadeh K, Boeckx B, Vandenhaute J, De Visscher A, Bernaerts E, Mitera T, Jacobs C, Vanderbeke L, Van Mol P, Van Herck Y, Hermans G, Meersseman P, Wilmer A, Gouwy M, Garg AD, Humblet-Baron S, De Smet F, Martinod K, Wauters E, Proost P, Wouters C, Leclercq G, Lambrechts D, Wauters J, Matthys P. Severe COVID-19 patients display hyper-activated NK cells and NK cell-platelet aggregates. Front Immunol 2022; 13:861251. [PMID: 36275702 PMCID: PMC9581751 DOI: 10.3389/fimmu.2022.861251] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/15/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is characterised by a broad spectrum of clinical and pathological features. Natural killer (NK) cells play an important role in innate immune responses to viral infections. Here, we analysed the phenotype and activity of NK cells in the blood of COVID-19 patients using flow cytometry, single-cell RNA-sequencing (scRNA-seq), and a cytotoxic killing assay. In the plasma of patients, we quantified the main cytokines and chemokines. Our cohort comprises COVID-19 patients hospitalised in a low-care ward unit (WARD), patients with severe COVID-19 disease symptoms hospitalised in intensive care units (ICU), and post-COVID-19 patients, who were discharged from hospital six weeks earlier. NK cells from hospitalised COVID-19 patients displayed an activated phenotype with substantial differences between WARD and ICU patients and the timing when samples were taken post-onset of symptoms. While NK cells from COVID-19 patients at an early stage of infection showed increased expression of the cytotoxic molecules perforin and granzyme A and B, NK cells from patients at later stages of COVID-19 presented enhanced levels of IFN-γ and TNF-α which were measured ex vivo in the absence of usual in vitro stimulation. These activated NK cells were phenotyped as CD49a+CD69a+CD107a+ cells, and their emergence in patients correlated to the number of neutrophils, and plasma IL-15, a key cytokine in NK cell activation. Despite lower amounts of cytotoxic molecules in NK cells of patients with severe symptoms, majority of COVID-19 patients displayed a normal cytotoxic killing of Raji tumour target cells. In vitro stimulation of patients blood cells by IL-12+IL-18 revealed a defective IFN-γ production in NK cells of ICU patients only, indicative of an exhausted phenotype. ScRNA-seq revealed, predominantly in patients with severe COVID-19 disease symptoms, the emergence of an NK cell subset with a platelet gene signature that we identified by flow and imaging cytometry as aggregates of NK cells with CD42a+CD62P+ activated platelets. Post-COVID-19 patients show slow recovery of NK cell frequencies and phenotype. Our study points to substantial changes in NK cell phenotype during COVID-19 disease and forms a basis to explore the contribution of platelet-NK cell aggregates to antiviral immunity against SARS-CoV-2 and disease pathology.
Collapse
Affiliation(s)
- Bert Malengier-Devlies
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Jessica Filtjens
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Kourosh Ahmadzadeh
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Bram Boeckx
- Laboratory of Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Jessica Vandenhaute
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Amber De Visscher
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Eline Bernaerts
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Tania Mitera
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Cato Jacobs
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Lore Vanderbeke
- Laboratory of Clinical Bacteriology and Mycology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Pierre Van Mol
- Laboratory of Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Yannick Van Herck
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Greet Hermans
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Philippe Meersseman
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Alexander Wilmer
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Abhishek D. Garg
- Laboratory for Cell Stress & Immunity (CSI), Department of Cellular and Molecular Medicine (CMM), KU Leuven, Leuven, Belgium
| | - Stephanie Humblet-Baron
- Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Els Wauters
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Carine Wouters
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Georges Leclercq
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Joost Wauters
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
99
|
Vanderheiden A, Klein RS. Neuroinflammation and COVID-19. Curr Opin Neurobiol 2022; 76:102608. [PMID: 35863101 PMCID: PMC9239981 DOI: 10.1016/j.conb.2022.102608] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 01/11/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has caused a historic pandemic of respiratory disease. COVID-19 also causes acute and post-acute neurological symptoms, which range from mild, such as headaches, to severe, including hemorrhages. Current evidence suggests that there is no widespread infection of the central nervous system (CNS) by SARS-CoV-2, thus what is causing COVID-19 neurological disease? Here, we review potential immunological mechanisms driving neurological disease in COVID-19 patients. We begin by discussing the implications of imbalanced peripheral immunity on CNS function. Next, we examine the evidence for dysregulation of the blood-brain barrier during SARS-CoV-2 infection. Last, we discuss the role myeloid cells may play in promoting COVID-19 neurological disease. Combined, we highlight the role of innate immunity in COVID-19 neuroinflammation and suggest areas for future research.
Collapse
Affiliation(s)
- Abigail Vanderheiden
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA; Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Robyn S Klein
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA; Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Departments of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Departments of Neurosciences, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
100
|
Abstract
SARS-CoV-2, the virus that causes coronavirus disease (COVID)-19, has become a persistent global health threat. Individuals who are symptomatic for COVID-19 frequently exhibit respiratory illness, which is often accompanied by neurological symptoms of anosmia and fatigue. Mounting clinical data also indicate that many COVID-19 patients display long-term neurological disorders postinfection such as cognitive decline, which emphasizes the need to further elucidate the effects of COVID-19 on the central nervous system. In this review article, we summarize an emerging body of literature describing the impact of SARS-CoV-2 infection on central nervous system (CNS) health and highlight important areas of future investigation.
Collapse
Affiliation(s)
- Nick R. Natale
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
- Global Biothreats Graduate Training Program, University of Virginia, Charlottesville, VA, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
- Global Biothreats Graduate Training Program, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, USA
| | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
- Global Biothreats Graduate Training Program, University of Virginia, Charlottesville, VA, USA
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Pathology, University of Virginia Health System, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, USA
| |
Collapse
|