101
|
Wang W, Chai Z, Cooper ME, Zimmet PZ, Guo H, Ding J, Yang F, Chen X, Lin X, Zhang K, Zhong Q, Li Z, Zhang P, Wu Z, Guan X, Zhang L, He K. High Fasting Blood Glucose Level With Unknown Prior History of Diabetes Is Associated With High Risk of Severe Adverse COVID-19 Outcome. Front Endocrinol (Lausanne) 2021; 12:791476. [PMID: 34956098 PMCID: PMC8692378 DOI: 10.3389/fendo.2021.791476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/15/2021] [Indexed: 01/08/2023] Open
Abstract
Background We aimed to understand how glycaemic levels among COVID-19 patients impact their disease progression and clinical complications. Methods We enrolled 2,366 COVID-19 patients from Huoshenshan hospital in Wuhan. We stratified the COVID-19 patients into four subgroups by current fasting blood glucose (FBG) levels and their awareness of prior diabetic status, including patients with FBG<6.1mmol/L with no history of diabetes (group 1), patients with FBG<6.1mmol/L with a history of diabetes diagnosed (group 2), patients with FBG≥6.1mmol/L with no history of diabetes (group 3) and patients with FBG≥6.1mmol/L with a history of diabetes diagnosed (group 4). A multivariate cause-specific Cox proportional hazard model was used to assess the associations between FBG levels or prior diabetic status and clinical adversities in COVID-19 patients. Results COVID-19 patients with higher FBG and unknown diabetes in the past (group 3) are more likely to progress to the severe or critical stage than patients in other groups (severe: 38.46% vs 23.46%-30.70%; critical 7.69% vs 0.61%-3.96%). These patients also have the highest abnormal level of inflammatory parameters, complications, and clinical adversities among all four groups (all p<0.05). On day 21 of hospitalisation, group 3 had a significantly higher risk of ICU admission [14.1% (9.6%-18.6%)] than group 4 [7.0% (3.7%-10.3%)], group 2 [4.0% (0.2%-7.8%)] and group 1 [2.1% (1.4%-2.8%)], (P<0.001). Compared with group 1 who had low FBG, group 3 demonstrated 5 times higher risk of ICU admission events during hospitalisation (HR=5.38, 3.46-8.35, P<0.001), while group 4, where the patients had high FBG and prior diabetes diagnosed, also showed a significantly higher risk (HR=1.99, 1.12-3.52, P=0.019), but to a much lesser extent than in group 3. Conclusion Our study shows that COVID-19 patients with current high FBG levels but unaware of pre-existing diabetes, or possibly new onset diabetes as a result of COVID-19 infection, have a higher risk of more severe adverse outcomes than those aware of prior diagnosis of diabetes and those with low current FBG levels.
Collapse
Affiliation(s)
- Wenjun Wang
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Translational Medical Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Artificial Intelligence Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Big Data Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhonglin Chai
- Department of Diabetes, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Mark E. Cooper
- Department of Diabetes, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Paul Z. Zimmet
- Department of Diabetes, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Hua Guo
- Department of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Junyu Ding
- Department of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Feifei Yang
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Translational Medical Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Artificial Intelligence Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Big Data Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xu Chen
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Translational Medical Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Artificial Intelligence Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Big Data Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xixiang Lin
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Translational Medical Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Artificial Intelligence Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Big Data Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Kai Zhang
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Qin Zhong
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Translational Medical Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Artificial Intelligence Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Big Data Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zongren Li
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Translational Medical Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Artificial Intelligence Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Big Data Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Peifang Zhang
- BioMind Technology, Zhongguancun Medical Engineering Center, Beijing, China
| | - Zhenzhou Wu
- BioMind Technology, Zhongguancun Medical Engineering Center, Beijing, China
| | - Xizhou Guan
- Department of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Lei Zhang
- Department of Diabetes, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC, Australia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Kunlun He
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Translational Medical Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Artificial Intelligence Research Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical Big Data Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
102
|
Raines NH, Cheung MD, Wilson LS, Edberg JC, Erdmann NB, Schmaier AA, Berryhill TF, Manickas-Hill Z, Li JZ, Yu XG, Agarwal A, Barnes S, Parikh SM. Nicotinamide Adenine Dinucleotide Biosynthetic Impairment and Urinary Metabolomic Alterations Observed in Hospitalized Adults With COVID-19-Related Acute Kidney Injury. Kidney Int Rep 2021; 6:3002-3013. [PMID: 34541422 PMCID: PMC8439094 DOI: 10.1016/j.ekir.2021.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/03/2021] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) is common in COVID-19 and associated with increased morbidity and mortality. We investigated alterations in the urine metabolome to test the hypothesis that impaired nicotinamide adenine dinucleotide (NAD+) biosynthesis and other deficiencies in energy metabolism in the kidney, previously characterized in ischemic, toxic, and inflammatory etiologies of AKI, will be present in COVID-19-associated AKI. METHODS This is a case-control study among the following 2 independent populations of adults hospitalized with COVID-19: a critically ill population in Boston, Massachusetts, and a general population in Birmingham, Alabama. The cases had AKI stages 2 or 3 by Kidney Disease Improving Global Outcomes (KDIGO) criteria; the controls had no AKI. Metabolites were measured by liquid chromatography-mass spectrometry. RESULTS A total of 14 cases and 14 controls were included from Boston and 8 cases and 10 controls from Birmingham. Increased urinary quinolinate-to-tryptophan ratio (Q/T), found with impaired NAD+ biosynthesis, was present in the cases at each location and pooled across locations (median [interquartile range]: 1.34 [0.59-2.96] in cases, 0.31 [0.13-1.63] in controls, P = 0.0013). Altered energy metabolism and purine metabolism contributed to a distinct urinary metabolomic signature that differentiated patients with and without AKI (supervised random forest class error: 2 of 28 in Boston, 0 of 18 in Birmingham). CONCLUSION Urinary metabolites spanning multiple biochemical pathways differentiate AKI versus non-AKI in patients hospitalized with COVID-19 and suggest a conserved impairment in NAD+ biosynthesis, which may present a novel therapeutic target to mitigate COVID-19-associated AKI.
Collapse
Affiliation(s)
- Nathan H. Raines
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew D. Cheung
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Landon S. Wilson
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeffrey C. Edberg
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nathaniel B. Erdmann
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alec A. Schmaier
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Taylor F. Berryhill
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zachary Manickas-Hill
- Ragon Institute of the Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard University, Massachusetts General Hospital, Cambridge, Massachusetts, USA
| | - Jonathan Z. Li
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Xu G. Yu
- Ragon Institute of the Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard University, Massachusetts General Hospital, Cambridge, Massachusetts, USA
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Stephen Barnes
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samir M. Parikh
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
103
|
Matsuyama T, Yoshinaga SK, Shibue K, Mak TW. Comorbidity-associated glutamine deficiency is a predisposition to severe COVID-19. Cell Death Differ 2021; 28:3199-3213. [PMID: 34663907 PMCID: PMC8522258 DOI: 10.1038/s41418-021-00892-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 vaccinations have greatly reduced COVID-19 cases, but we must continue to develop our understanding of the nature of the disease and its effects on human immunity. Previously, we suggested that a dysregulated STAT3 pathway following SARS-Co-2 infection ultimately leads to PAI-1 activation and cascades of pathologies. The major COVID-19-associated metabolic risks (old age, hypertension, cardiovascular diseases, diabetes, and obesity) share high PAI-1 levels and could predispose certain groups to severe COVID-19 complications. In this review article, we describe the common metabolic profile that is shared between all of these high-risk groups and COVID-19. This profile not only involves high levels of PAI-1 and STAT3 as previously described, but also includes low levels of glutamine and NAD+, coupled with overproduction of hyaluronan (HA). SARS-CoV-2 infection exacerbates this metabolic imbalance and predisposes these patients to the severe pathophysiologies of COVID-19, including the involvement of NETs (neutrophil extracellular traps) and HA overproduction in the lung. While hyperinflammation due to proinflammatory cytokine overproduction has been frequently documented, it is recently recognized that the immune response is markedly suppressed in some cases by the expansion and activity of MDSCs (myeloid-derived suppressor cells) and FoxP3+ Tregs (regulatory T cells). The metabolomics profiles of severe COVID-19 patients and patients with advanced cancer are similar, and in high-risk patients, SARS-CoV-2 infection leads to aberrant STAT3 activation, which promotes a cancer-like metabolism. We propose that glutamine deficiency and overproduced HA is the central metabolic characteristic of COVID-19 and its high-risk groups. We suggest the usage of glutamine supplementation and the repurposing of cancer drugs to prevent the development of severe COVID-19 pneumonia.
Collapse
Affiliation(s)
- Toshifumi Matsuyama
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| | | | - Kimitaka Shibue
- Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Tak W Mak
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2M9, Canada
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, M5G 1L7, Canada
- Department of Immunology, University of Toronto, 101 College Street, Toronto, ON, M5G 1L7, Canada
- Department of Pathology, University of Hong Kong, Hong Kong, Pok Fu Lam, 999077, Hong Kong
| |
Collapse
|
104
|
Wang Q, Zhou J, Liu T, Yang N, Mi X, Han D, Han Y, Chen L, Liu K, Zheng H, Zhang J, Lin X, Li Y, Hong J, Li Z, Guo X. Predictive Value of Preoperative Profiling of Serum Metabolites for Emergence Agitation After General Anesthesia in Adult Patients. Front Mol Biosci 2021; 8:739227. [PMID: 34746231 PMCID: PMC8566542 DOI: 10.3389/fmolb.2021.739227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/20/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Emergence agitation (EA) in adult patients under general anesthesia leads to increased postoperative complications and heavy medical burden. Unfortunately, its pathogenesis has not been clarified until now. The purpose of the present study was to explore the relationship between preoperative serum metabolites and EA. Methods: We used an untargeted metabolic analysis method to investigate the different metabolomes in the serum of EA patients and non-EA patients undergoing elective surgical procedures after the induction of general anesthesia. A Richmond Agitation-Sedation Scale score ≥ +2 was diagnosed as EA during postoperative emergence. Non-EA patients were matched with EA patients according to general characteristics. Preoperative serum samples of the two groups were collected to investigate the association between serum metabolites and EA development. Results: The serum samples of 16 EA patients with 34 matched non-EA patients were obtained for metabolic analysis. After screening and alignment with databases, 31 altered metabolites were detected between the two groups. These metabolites were mainly involved in the metabolism of lipids, purines, and amino acids. Analyses of receiver-operating characteristic curves showed that the preoperative alterations of choline, cytidine, glycerophosphocholine, L-phenylalanine, oleamide, and inosine may be associated with adult EA. Conclusion: Multiple metabolic abnormalities (including those for lipids, purines, and amino acids) and other pathological processes (e.g., neurotransmitter imbalance and oxidative stress) may contribute to EA. Several altered metabolites in serum before surgery may have predictive value for EA diagnosis. This study might afford new metabolic clues for the understanding of EA pathogenesis.
Collapse
Affiliation(s)
- Qian Wang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jiansuo Zhou
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yongzheng Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Lei Chen
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Kaixi Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Hongcai Zheng
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jing Zhang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiaona Lin
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yitong Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jingshu Hong
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
105
|
Tayara H, Abdelbaky I, To Chong K. Recent omics-based computational methods for COVID-19 drug discovery and repurposing. Brief Bioinform 2021; 22:6355836. [PMID: 34423353 DOI: 10.1093/bib/bbab339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/09/2021] [Indexed: 12/22/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the main reason for the increasing number of deaths worldwide. Although strict quarantine measures were followed in many countries, the disease situation is still intractable. Thus, it is needed to utilize all possible means to confront this pandemic. Therefore, researchers are in a race against the time to produce potential treatments to cure or reduce the increasing infections of COVID-19. Computational methods are widely proving rapid successes in biological related problems, including diagnosis and treatment of diseases. Many efforts in recent months utilized Artificial Intelligence (AI) techniques in the context of fighting the spread of COVID-19. Providing periodic reviews and discussions of recent efforts saves the time of researchers and helps to link their endeavors for a faster and efficient confrontation of the pandemic. In this review, we discuss the recent promising studies that used Omics-based data and utilized AI algorithms and other computational tools to achieve this goal. We review the established datasets and the developed methods that were basically directed to new or repurposed drugs, vaccinations and diagnosis. The tools and methods varied depending on the level of details in the available information such as structures, sequences or metabolic data.
Collapse
Affiliation(s)
- Hilal Tayara
- School of international Engineering and Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Ibrahim Abdelbaky
- Artificial Intelligence Department, Faculty of Computers and Artificial Intelligence, Benha University, Banha 13518, Egypt
| | - Kil To Chong
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, Jeollabukdo 54896, Republic of Korea.,Advances Electronics and Information Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
106
|
Shen T, Wang T. Metabolic Reprogramming in COVID-19. Int J Mol Sci 2021; 22:ijms222111475. [PMID: 34768906 PMCID: PMC8584248 DOI: 10.3390/ijms222111475] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022] Open
Abstract
Plenty of research has revealed virus induced alternations in metabolic pathways, which is known as metabolic reprogramming. Studies focusing on COVID-19 have uncovered significant changes in metabolism, resulting in the perspective that COVID-19 is a metabolic disease. Reprogramming of amino acid, glucose, cholesterol and fatty acid is distinctive characteristic of COVID-19 infection. These metabolic changes in COVID-19 have a critical role not only in producing energy and virus constituent elements, but also in regulating immune response, offering new insights into COVID-19 pathophysiology. Remarkably, metabolic reprogramming provides great opportunities for developing novel biomarkers and therapeutic agents for COVID-19 infection. Such novel agents are expected to be effective adjuvant therapies. In this review, we integrate present studies about major metabolic reprogramming in COVID-19, as well as the possibility of targeting reprogrammed metabolism to combat virus infection.
Collapse
Affiliation(s)
- Tao Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China;
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China;
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
- Correspondence:
| |
Collapse
|
107
|
Single-Cell Multiomics Analysis for Drug Discovery. Metabolites 2021; 11:metabo11110729. [PMID: 34822387 PMCID: PMC8623556 DOI: 10.3390/metabo11110729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 02/02/2023] Open
Abstract
Given the heterogeneity seen in cell populations within biological systems, analysis of single cells is necessary for studying mechanisms that cannot be identified on a bulk population level. There are significant variations in the biological and physiological function of cell populations due to the functional differences within, as well as between, single species as a result of the specific proteome, transcriptome, and metabolome that are unique to each individual cell. Single-cell analysis proves crucial in providing a comprehensive understanding of the biological and physiological properties underlying human health and disease. Omics technologies can help to examine proteins (proteomics), RNA molecules (transcriptomics), and the chemical processes involving metabolites (metabolomics) in cells, in addition to genomes. In this review, we discuss the value of multiomics in drug discovery and the importance of single-cell multiomics measurements. We will provide examples of the benefits of applying single-cell omics technologies in drug discovery and development. Moreover, we intend to show how multiomics offers the opportunity to understand the detailed events which produce or prevent disease, and ways in which the separate omics disciplines complement each other to build a broader, deeper knowledge base.
Collapse
|
108
|
Khodadoust MM. Inferring a causal relationship between ceramide levels and COVID-19 respiratory distress. Sci Rep 2021; 11:20866. [PMID: 34675292 PMCID: PMC8531370 DOI: 10.1038/s41598-021-00286-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
A causal relationship between plasma ceramide concentration and respiratory distress symptoms in COVID-19 patients is inferred. In this study, plasma samples of 52 individuals infected with COVID-19 were utilized in a lipidomic analysis. Lipids belonging to the ceramide class exhibited a 400-fold increase in total plasma concentration in infected patients. Further analysis led to the demonstration of concentration dependency for severe COVID-19 respiratory symptoms in a subclass of ceramides. The subclasses Cer(d18:0/24:1), Cer(d18:1/24:1), and Cer(d18:1/22:0) were shown to be increased by 48-, 40-, and 33-fold, respectively, in infected plasma samples and to 116-, 91- and 50-fold, respectively, in plasma samples with respiratory distress. Hence, monitoring plasma ceramide concentration, can be a valuable tool for measuring effects of therapies on COVID-19 respiratory distress patients.
Collapse
|
109
|
Suvarna K, Salkar A, Palanivel V, Bankar R, Banerjee N, Gayathri J Pai M, Srivastava A, Singh A, Khatri H, Agrawal S, Shrivastav O, Shastri J, Srivastava S. A Multi-omics Longitudinal Study Reveals Alteration of the Leukocyte Activation Pathway in COVID-19 Patients. J Proteome Res 2021; 20:4667-4680. [PMID: 34379420 PMCID: PMC8370121 DOI: 10.1021/acs.jproteome.1c00215] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Indexed: 12/24/2022]
Abstract
Severe coronavirus disease 2019 (COVID-19) infection may lead to lung injury, multi-organ failure, and eventually death. Cytokine storm due to excess cytokine production has been associated with fatality in severe infections. However, the specific molecular signatures associated with the elevated immune response are yet to be elucidated. We performed a mass-spectrometry-based proteomic and metabolomic analysis of COVID-19 plasma samples collected at two time points. Using Orbitrap Fusion LC-MS/MS-based label-free proteomic analysis, we identified around 10 significant proteins, 32 significant peptides, and 5 metabolites that were dysregulated at the severe time points. Few of these proteins identified by quantitative proteomics were validated using the multiple reaction monitoring (MRM) assay. Integrated pathway analysis using distinct proteomic and metabolomic signatures revealed alterations in complement and coagulation cascade, platelet aggregation, myeloid leukocyte activation pathway, and arginine metabolism. Further, we highlight the role of leukocyte activation and arginine metabolism in COVID-19 pathogenesis and targeting these pathways for COVID-19 therapeutics.
Collapse
Affiliation(s)
- Kruthi Suvarna
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| | - Akanksha Salkar
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| | - Viswanthram Palanivel
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| | - Renuka Bankar
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| | - Nirjhar Banerjee
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| | - Medha Gayathri J Pai
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| | - Alisha Srivastava
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
- University of Delhi, New
Delhi, Delhi 110021, India
| | - Avinash Singh
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| | - Harsh Khatri
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| | - Sachee Agrawal
- Kasturba Hospital for Infectious
Diseases, Chinchpokli, Mumbai, Maharashtra 400034,
India
| | - Om Shrivastav
- Kasturba Hospital for Infectious
Diseases, Chinchpokli, Mumbai, Maharashtra 400034,
India
| | - Jayanthi Shastri
- Kasturba Hospital for Infectious
Diseases, Chinchpokli, Mumbai, Maharashtra 400034,
India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Powai, Mumbai 400076,
India
| |
Collapse
|
110
|
Theken KN, Tang SY, Sengupta S, FitzGerald GA. The roles of lipids in SARS-CoV-2 viral replication and the host immune response. J Lipid Res 2021; 62:100129. [PMID: 34599996 PMCID: PMC8480132 DOI: 10.1016/j.jlr.2021.100129] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
The significant morbidity and mortality associated with severe acute respiratory syndrome coronavirus 2 infection has underscored the need for novel antiviral strategies. Lipids play essential roles in the viral life cycle. The lipid composition of cell membranes can influence viral entry by mediating fusion or affecting receptor conformation. Upon infection, viruses can reprogram cellular metabolism to remodel lipid membranes and fuel the production of new virions. Furthermore, several classes of lipid mediators, including eicosanoids and sphingolipids, can regulate the host immune response to viral infection. Here, we summarize the existing literature on the mechanisms through which these lipid mediators may regulate viral burden in COVID-19. Furthermore, we define the gaps in knowledge and identify the core areas in which lipids offer therapeutic promise for severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Katherine N Theken
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Oral Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Soon Yew Tang
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shaon Sengupta
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
111
|
Lavorgna G, Cavalli G, Dagna L, Gregori S, Larcher A, Landoni G, Ciceri F, Montorsi F, Salonia A. A virus-free cellular model recapitulates several features of severe COVID-19. Sci Rep 2021; 11:17473. [PMID: 34471195 PMCID: PMC8410838 DOI: 10.1038/s41598-021-96875-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
As for all newly-emergent pathogens, SARS-CoV-2 presents with a relative paucity of clinical information and experimental models, a situation hampering both the development of new effective treatments and the prediction of future outbreaks. Here, we find that a simple virus-free model, based on publicly available transcriptional data from human cell lines, is surprisingly able to recapitulate several features of the clinically relevant infections. By segregating cell lines (n = 1305) from the CCLE project on the base of their sole angiotensin-converting enzyme 2 (ACE2) mRNA content, we found that overexpressing cells present with molecular features resembling those of at-risk patients, including senescence, impairment of antibody production, epigenetic regulation, DNA repair and apoptosis, neutralization of the interferon response, proneness to an overemphasized innate immune activity, hyperinflammation by IL-1, diabetes, hypercoagulation and hypogonadism. Likewise, several pathways were found to display a differential expression between sexes, with males being in the least advantageous position, thus suggesting that the model could reproduce even the sex-related disparities observed in the clinical outcome of patients with COVID-19. Overall, besides validating a new disease model, our data suggest that, in patients with severe COVID-19, a baseline ground could be already present and, as a consequence, the viral infection might simply exacerbate a variety of latent (or inherent) pre-existing conditions, representing therefore a tipping point at which they become clinically significant.
Collapse
Affiliation(s)
- Giovanni Lavorgna
- grid.18887.3e0000000417581884Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giulio Cavalli
- grid.15496.3fUniversity Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- grid.15496.3fUniversity Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Gregori
- grid.18887.3e0000000417581884San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandro Larcher
- grid.18887.3e0000000417581884Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giovanni Landoni
- grid.15496.3fUniversity Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Anesthesia and Intensive Care Department, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Fabio Ciceri
- grid.15496.3fUniversity Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Hematology and Bone Marrow Transplant Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Montorsi
- grid.18887.3e0000000417581884Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy ,grid.15496.3fUniversity Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Salonia
- grid.18887.3e0000000417581884Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy ,grid.15496.3fUniversity Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
112
|
Tang Y, Hu L, Liu Y, Zhou B, Qin X, Ye J, Shen M, Wu Z, Zhang P. Possible mechanisms of cholesterol elevation aggravating COVID-19. Int J Med Sci 2021; 18:3533-3543. [PMID: 34522180 PMCID: PMC8436106 DOI: 10.7150/ijms.62021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/04/2021] [Indexed: 12/23/2022] Open
Abstract
Importance: Despite the availability of a vaccine against the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), humans will have to live with this virus and the after-effects of the coronavirus disease 2019 (COVID-19) infection for a long time. Cholesterol plays an important role in the infection and prognosis of SARS-CoV-2, and the study of its mechanism is of great significance not only for the treatment of COVID-19 but also for research on generic antiviral drugs. Observations: Cholesterol promotes the development of atherosclerosis by activating NLR family pyrin domain containing 3 (NLRP3), and the resulting inflammatory environment indirectly contributes to COVID-19 infection and subsequent deterioration. In in vitro studies, membrane cholesterol increased the number of viral entry sites on the host cell membrane and the number of angiotensin-converting enzyme 2 (ACE2) receptors in the membrane fusion site. Previous studies have shown that the fusion protein of the virus interacts with cholesterol, and the spike protein of SARS-CoV-2 also requires cholesterol to enter the host cells. Cholesterol in blood interacts with the spike protein to promote the entry of spike cells, wherein the scavenger receptor class B type 1 (SR-B1) plays an important role. Because of the cardiovascular protective effects of lipid-lowering therapy and the additional anti-inflammatory effects of lipid-lowering drugs, it is currently recommended to continue lipid-lowering therapy for patients with COVID-19, but the safety of extremely low LDL-C is questionable. Conclusions and Relevance: Cholesterol can indirectly increase the susceptibility of patients to SARS-CoV-2 and increase the risk of death from COVID-19, which are mediated by NLRP3 and atherosclerotic plaques, respectively. Cholesterol present in the host cell membrane, virus, and blood may also directly participate in the virus cell entry process, but the specific mechanism still needs further study. Patients with COVID-19 are recommended to continue lipid-lowering therapy.
Collapse
Affiliation(s)
- Yan Tang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 235 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
- Zhujiang Hospital, Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, No. 6, Chenggui Road, East District, Zhongshan, 528403, Guangdong, People's Republic of China
| | - Longtai Hu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 235 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
- School of Traditional Chinese Medicine, Southern Medical University, No. 6, Chenggui Road, East District, Zhongshan, 528403, Guangdong, People's Republic of China
| | - Yi Liu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 235 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
- Zhujiang Hospital, Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, No. 6, Chenggui Road, East District, Zhongshan, 528403, Guangdong, People's Republic of China
| | - Bangyi Zhou
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 235 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
- Zhujiang Hospital, Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, No. 6, Chenggui Road, East District, Zhongshan, 528403, Guangdong, People's Republic of China
| | - Xiaohuan Qin
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 235 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
- Zhujiang Hospital, Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, No. 6, Chenggui Road, East District, Zhongshan, 528403, Guangdong, People's Republic of China
| | - Jujian Ye
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 235 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
- Zhujiang Hospital, Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, No. 6, Chenggui Road, East District, Zhongshan, 528403, Guangdong, People's Republic of China
| | - Maoze Shen
- Department of Cardiology, Raoping County People's Hospital, 161 Caichang Street, Huanggang Town, Chaozhou, 515700, Guangdong, People's Republic of China
| | - Zhijian Wu
- Department of Cardiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, No. 6, Chenggui Road, East District, Zhongshan, 528403, Guangdong, People's Republic of China
| | - Peidong Zhang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 235 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
| |
Collapse
|
113
|
Sindelar M, Stancliffe E, Schwaiger-Haber M, Anbukumar DS, Adkins-Travis K, Goss CW, O’Halloran JA, Mudd PA, Liu WC, Albrecht RA, García-Sastre A, Shriver LP, Patti GJ. Longitudinal metabolomics of human plasma reveals prognostic markers of COVID-19 disease severity. Cell Rep Med 2021; 2:100369. [PMID: 34308390 PMCID: PMC8292035 DOI: 10.1016/j.xcrm.2021.100369] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/01/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
There is an urgent need to identify which COVID-19 patients will develop life-threatening illness so that medical resources can be optimally allocated and rapid treatment can be administered early in the disease course, when clinical management is most effective. To aid in the prognostic classification of disease severity, we perform untargeted metabolomics on plasma from 339 patients, with samples collected at six longitudinal time points. Using the temporal metabolic profiles and machine learning, we build a predictive model of disease severity. We discover that a panel of metabolites measured at the time of study entry successfully determines disease severity. Through analysis of longitudinal samples, we confirm that most of these markers are directly related to disease progression and that their levels return to baseline upon disease recovery. Finally, we validate that these metabolites are also altered in a hamster model of COVID-19.
Collapse
Affiliation(s)
- Miriam Sindelar
- Department of Chemistry, Washington University, St. Louis, MO, USA
- Department of Medicine, Washington University, St. Louis, MO, USA
| | - Ethan Stancliffe
- Department of Chemistry, Washington University, St. Louis, MO, USA
- Department of Medicine, Washington University, St. Louis, MO, USA
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Washington University, St. Louis, MO, USA
- Department of Medicine, Washington University, St. Louis, MO, USA
| | - Dhanalakshmi S. Anbukumar
- Department of Chemistry, Washington University, St. Louis, MO, USA
- Department of Medicine, Washington University, St. Louis, MO, USA
| | | | - Charles W. Goss
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Philip A. Mudd
- Department of Emergency Medicine, Washington University, St. Louis, MO, USA
| | - Wen-Chun Liu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Randy A. Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Leah P. Shriver
- Department of Chemistry, Washington University, St. Louis, MO, USA
- Department of Medicine, Washington University, St. Louis, MO, USA
| | - Gary J. Patti
- Department of Chemistry, Washington University, St. Louis, MO, USA
- Department of Medicine, Washington University, St. Louis, MO, USA
- Siteman Cancer Center, Washington University, St. Louis, MO, USA
| |
Collapse
|
114
|
Jiang X, Chen X, Chen Z, Yu J, Lou H, Wu J. High-Throughput Salivary Metabolite Profiling on an Ultralow Noise Tip-Enhanced Laser Desorption Ionization Mass Spectrometry Platform for Noninvasive Diagnosis of Early Lung Cancer. J Proteome Res 2021; 20:4346-4356. [PMID: 34342461 DOI: 10.1021/acs.jproteome.1c00310] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Lung cancer (LC) is a widespread cancer that is the cause of the highest mortality rate accounting for 25% of all cancer deaths. To date, most LC patients are diagnosed at the advanced stage owing to the lack of obvious symptoms in the early stage and the limitations of current clinical diagnostic techniques. Therefore, developing a high throughput technique for early screening is of great importance. In this work, we established an effective and rapid salivary metabolic analysis platform for early LC diagnosis and combined metabolomics and transcriptomics to reveal the metabolic fluctuations correlated to LC. Saliva samples were collected from a total of 150 volunteers including 89 patients with early LC, 11 patients with advanced LC, and 50 healthy controls. The metabolic profiling of noninvasive samples was investigated on an ultralow noise TELDI-MS platform. In addition, data normalization methods were screened and assessed to overcome the MS signal variation caused by individual difference for biomarker mining. For untargeted metabolic profiling of saliva samples, around 264 peaks could be reliably detected in each sample. After multivariate analysis, 23 metabolites were sorted out and verified to be related to the dysfunction of the amino acid and nucleotide metabolism in early LC. Notably, transcriptomic data from online TCGA repository were utilized to support findings from the salivary metabolomics experiment, including the disorder of amino acid biosynthesis and amino acid metabolism. Based on the verified differential metabolites, early LC patients could be clearly distinguished from healthy controls with a sensitivity of 97.2% and a specificity of 92%. The ultralow noise TELDI-MS platform displayed satisfactory ability to explore salivary metabolite information and discover potential biomarkers that may help develop a noninvasive screening tool for early LC.
Collapse
Affiliation(s)
- Xinrong Jiang
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Xiaoming Chen
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, China.,Well-Healthcare Technologies Co., Hangzhou 310051, China
| | - Zhao Chen
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jiekai Yu
- Institute of Cancer Research, The Second Affiliated Hospital of Zhejiang University, Hangzhou 310009, China
| | - Haizhou Lou
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jianmin Wu
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
115
|
Tsoukalas D, Sarandi E, Georgaki S. The snapshot of metabolic health in evaluating micronutrient status, the risk of infection and clinical outcome of COVID-19. Clin Nutr ESPEN 2021; 44:173-187. [PMID: 34330463 PMCID: PMC8234252 DOI: 10.1016/j.clnesp.2021.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022]
Abstract
COVID-19 has re-established the significance of analyzing the organism through a metabolic perspective to uncover the dynamic interconnections within the biological systems. The role of micronutrient status and metabolic health emerge as pivotal in COVID-19 pathogenesis and the immune system's response. Metabolic disruption, proceeding from modifiable factors, has been proposed as a significant risk factor accounting for infection susceptibility, disease severity and risk for post-COVID complications. Metabolomics, the comprehensive study and quantification of intermediates and products of metabolism, is a rapidly evolving field and a novel tool in biomarker discovery. In this article, we propose that leveraging insulin resistance biomarkers along with biomarkers of micronutrient deficiencies, will allow for a diagnostic window and provide functional therapeutic targets. Specifically, metabolomics can be applied as: a. At-home test to assess the risk of infection and propose nutritional support, b. A screening tool for high-risk COVID-19 patients to develop serious illness during hospital admission and prioritize medical support, c(i). A tool to match nutritional support with specific nutrient requirements for mildly ill patients to reduce the risk for hospitalization, and c(ii). for critically ill patients to reduce recovery time and risk of post-COVID complications, d. At-home test to monitor metabolic health and reduce post-COVID symptomatology. Metabolic rewiring offers potential virtues towards disease prevention, dissection of high-risk patients, taking actionable therapeutic measures, as well as shielding against post-COVID syndrome.
Collapse
Affiliation(s)
- Dimitris Tsoukalas
- European Institute of Nutritional Medicine, 00198 Rome, Italy; Metabolomic Medicine, Health Clinic for Autoimmune and Chronic Diseases, 10674 Athens, Greece.
| | - Evangelia Sarandi
- Metabolomic Medicine, Health Clinic for Autoimmune and Chronic Diseases, 10674 Athens, Greece; Laboratory of Toxicology and Forensic Sciences, Medical School, University of Crete, 71003 Heraklion, Greece.
| | - Spyridoula Georgaki
- Metabolomic Medicine, Health Clinic for Autoimmune and Chronic Diseases, 10674 Athens, Greece.
| |
Collapse
|
116
|
Molecular Phenomic Approaches to Deconvolving the Systemic Effects of SARS-CoV-2 Infection and Post-acute COVID-19 Syndrome. PHENOMICS 2021; 1:143-150. [PMID: 35233558 PMCID: PMC8295979 DOI: 10.1007/s43657-021-00020-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/25/2021] [Accepted: 06/17/2021] [Indexed: 11/09/2022]
Abstract
SARS COV-2 infection causes acute and frequently severe respiratory disease with associated multi-organ damage and systemic disturbances in many biochemical pathways. Metabolic phenotyping provides deep insights into the complex immunopathological problems that drive the resulting COVID-19 disease and is also a source of novel metrics for assessing patient recovery. A multiplatform metabolic phenotyping approach to studying the pathology and systemic metabolic sequelae of COVID-19 is considered here, together with a framework for assessing post-acute COVID-19 Syndrome (PACS) that is a major long-term health consequence for many patients. The sudden emergence of the disease presents a biological discovery challenge as we try to understand the pathological mechanisms of the disease and develop effective mitigation strategies. This requires technologies to measure objectively the extent and sub-phenotypes of the disease at the molecular level. Spectroscopic methods can reveal metabolic sub-phenotypes and new biomarkers that can be monitored during the acute disease phase and beyond. This approach is scalable and translatable to other pathologies and provides as an exemplar strategy for the investigation of other emergent zoonotic diseases with complex immunological drivers, multi-system involvements and diverse persistent symptoms.
Collapse
|
117
|
Mussap M, Fanos V. Could metabolomics drive the fate of COVID-19 pandemic? A narrative review on lights and shadows. Clin Chem Lab Med 2021; 59:1891-1905. [PMID: 34332518 DOI: 10.1515/cclm-2021-0414] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Human Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) infection activates a complex interaction host/virus, leading to the reprogramming of the host metabolism aimed at the energy supply for viral replication. Alterations of the host metabolic homeostasis strongly influence the immune response to SARS-CoV-2, forming the basis of a wide range of outcomes, from the asymptomatic infection to the onset of COVID-19 and up to life-threatening acute respiratory distress syndrome, vascular dysfunction, multiple organ failure, and death. Deciphering the molecular mechanisms associated with the individual susceptibility to SARS-CoV-2 infection calls for a system biology approach; this strategy can address multiple goals, including which patients will respond effectively to the therapeutic treatment. The power of metabolomics lies in the ability to recognize endogenous and exogenous metabolites within a biological sample, measuring their concentration, and identifying perturbations of biochemical pathways associated with qualitative and quantitative metabolic changes. Over the last year, a limited number of metabolomics- and lipidomics-based clinical studies in COVID-19 patients have been published and are discussed in this review. Remarkable alterations in the lipid and amino acid metabolism depict the molecular phenotype of subjects infected by SARS-CoV-2; notably, structural and functional data on the lipids-virus interaction may open new perspectives on targeted therapeutic interventions. Several limitations affect most metabolomics-based studies, slowing the routine application of metabolomics. However, moving metabolomics from bench to bedside cannot imply the mere determination of a given metabolite panel; rather, slotting metabolomics into clinical practice requires the conversion of metabolic patient-specific data into actionable clinical applications.
Collapse
Affiliation(s)
- Michele Mussap
- Laboratory Medicine, Department of Surgical Sciences, School of Medicine, University of Cagliari, Monserrato, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, School of Medicine, University of Cagliari, Monserrato, Italy
| |
Collapse
|
118
|
Gray N, Lawler NG, Zeng AX, Ryan M, Bong SH, Boughton BA, Bizkarguenaga M, Bruzzone C, Embade N, Wist J, Holmes E, Millet O, Nicholson JK, Whiley L. Diagnostic Potential of the Plasma Lipidome in Infectious Disease: Application to Acute SARS-CoV-2 Infection. Metabolites 2021; 11:467. [PMID: 34357361 PMCID: PMC8306636 DOI: 10.3390/metabo11070467] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
Improved methods are required for investigating the systemic metabolic effects of SARS-CoV-2 infection and patient stratification for precision treatment. We aimed to develop an effective method using lipid profiles for discriminating between SARS-CoV-2 infection, healthy controls, and non-SARS-CoV-2 respiratory infections. Targeted liquid chromatography-mass spectrometry lipid profiling was performed on discovery (20 SARS-CoV-2-positive; 37 healthy controls; 22 COVID-19 symptoms but SARS-CoV-2negative) and validation (312 SARS-CoV-2-positive; 100 healthy controls) cohorts. Orthogonal projection to latent structure-discriminant analysis (OPLS-DA) and Kruskal-Wallis tests were applied to establish discriminant lipids, significance, and effect size, followed by logistic regression to evaluate classification performance. OPLS-DA reported separation of SARS-CoV-2 infection from healthy controls in the discovery cohort, with an area under the curve (AUC) of 1.000. A refined panel of discriminant features consisted of six lipids from different subclasses (PE, PC, LPC, HCER, CER, and DCER). Logistic regression in the discovery cohort returned a training ROC AUC of 1.000 (sensitivity = 1.000, specificity = 1.000) and a test ROC AUC of 1.000. The validation cohort produced a training ROC AUC of 0.977 (sensitivity = 0.855, specificity = 0.948) and a test ROC AUC of 0.978 (sensitivity = 0.948, specificity = 0.922). The lipid panel was also able to differentiate SARS-CoV-2-positive individuals from SARS-CoV-2-negative individuals with COVID-19-like symptoms (specificity = 0.818). Lipid profiling and multivariate modelling revealed a signature offering mechanistic insights into SARS-CoV-2, with strong predictive power, and the potential to facilitate effective diagnosis and clinical management.
Collapse
Affiliation(s)
- Nicola Gray
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
- Centre for Computational and Systems Medicine, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Nathan G. Lawler
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
- Centre for Computational and Systems Medicine, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Annie Xu Zeng
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
| | - Monique Ryan
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
| | - Sze How Bong
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
| | - Berin A. Boughton
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
- Centre for Computational and Systems Medicine, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Maider Bizkarguenaga
- Centro de Investigación Cooperativa en Biociencias—CIC bioGUNE, Precision Medicine and Metabolism Laboratory, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Spain; (M.B.); (C.B.); (N.E.)
| | - Chiara Bruzzone
- Centro de Investigación Cooperativa en Biociencias—CIC bioGUNE, Precision Medicine and Metabolism Laboratory, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Spain; (M.B.); (C.B.); (N.E.)
| | - Nieves Embade
- Centro de Investigación Cooperativa en Biociencias—CIC bioGUNE, Precision Medicine and Metabolism Laboratory, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Spain; (M.B.); (C.B.); (N.E.)
| | - Julien Wist
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
- Centre for Computational and Systems Medicine, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK
| | - Oscar Millet
- Centro de Investigación Cooperativa en Biociencias—CIC bioGUNE, Precision Medicine and Metabolism Laboratory, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Spain; (M.B.); (C.B.); (N.E.)
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
- Centre for Computational and Systems Medicine, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Institute of Global Health Innovation, Faculty Building South Kensington Campus, Imperial College London, London SW7 2AZ, UK
| | - Luke Whiley
- Australian National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia; (N.G.); (N.G.L.); (A.X.Z.); (M.R.); (S.H.B.); (B.A.B.); (J.W.); (E.H.)
- Centre for Computational and Systems Medicine, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| |
Collapse
|
119
|
López-Hernández Y, Monárrez-Espino J, Oostdam ASHV, Delgado JEC, Zhang L, Zheng J, Valdez JJO, Mandal R, González FDLO, Moreno JCB, Trejo-Medinilla FM, López JA, Moreno JAE, Wishart DS. Targeted metabolomics identifies high performing diagnostic and prognostic biomarkers for COVID-19. Sci Rep 2021; 11:14732. [PMID: 34282210 PMCID: PMC8290000 DOI: 10.1038/s41598-021-94171-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/06/2021] [Indexed: 12/14/2022] Open
Abstract
Research exploring the development and outcome of COVID-19 infections has led to the need to find better diagnostic and prognostic biomarkers. This cross-sectional study used targeted metabolomics to identify potential COVID-19 biomarkers that predicted the course of the illness by assessing 110 endogenous plasma metabolites from individuals admitted to a local hospital for diagnosis/treatment. Patients were classified into four groups (≈ 40 each) according to standard polymerase chain reaction (PCR) COVID-19 testing and disease course: PCR-/controls (i.e., non-COVID controls), PCR+/not-hospitalized, PCR+/hospitalized, and PCR+/intubated. Blood samples were collected within 2 days of admission/PCR testing. Metabolite concentration data, demographic data and clinical data were used to propose biomarkers and develop optimal regression models for the diagnosis and prognosis of COVID-19. The area under the receiver operating characteristic curve (AUC; 95% CI) was used to assess each models' predictive value. A panel that included the kynurenine: tryptophan ratio, lysoPC a C26:0, and pyruvic acid discriminated non-COVID controls from PCR+/not-hospitalized (AUC = 0.947; 95% CI 0.931-0.962). A second panel consisting of C10:2, butyric acid, and pyruvic acid distinguished PCR+/not-hospitalized from PCR+/hospitalized and PCR+/intubated (AUC = 0.975; 95% CI 0.968-0.983). Only lysoPC a C28:0 differentiated PCR+/hospitalized from PCR+/intubated patients (AUC = 0.770; 95% CI 0.736-0.803). If additional studies with targeted metabolomics confirm the diagnostic value of these plasma biomarkers, such panels could eventually be of clinical use in medical practice.
Collapse
Affiliation(s)
- Yamilé López-Hernández
- Cátedras-CONACyT, Consejo Nacional de Ciencia y Tecnologia, 03940, México, México.
- Autonomous University of Zacatecas, 98000, Zacatecas, Mexico.
| | - Joel Monárrez-Espino
- Christus Muguerza Hospital Chihuahua-University of Monterrey, 31000, Chihuahua, Mexico.
| | | | - Julio Enrique Castañeda Delgado
- Cátedras-CONACyT, Consejo Nacional de Ciencia y Tecnologia, 03940, México, México
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, 98000, Zacatecas, México
| | - Lun Zhang
- The Metabolomics Innovation Center, University of Alberta, Edmonton, AB, T6G1C9, Canada
| | - Jiamin Zheng
- The Metabolomics Innovation Center, University of Alberta, Edmonton, AB, T6G1C9, Canada
| | - Juan José Oropeza Valdez
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, 98000, Zacatecas, México
| | - Rupasri Mandal
- The Metabolomics Innovation Center, University of Alberta, Edmonton, AB, T6G1C9, Canada
| | - Fátima de Lourdes Ochoa González
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, 98000, Zacatecas, México
- Doctorado en Ciencias Básicas, Universidad Autónoma de Zacatecas, Zacatecas, México
| | - Juan Carlos Borrego Moreno
- Departmento de Epidemiología, Hospital General de Zona #1 "Emilio Varela Luján", Instituto Mexicano del Seguro Social, 98000, Zacatecas, México
| | - Flor M Trejo-Medinilla
- Autonomous University of Zacatecas, 98000, Zacatecas, Mexico
- Doctorado en Ciencias Básicas, Universidad Autónoma de Zacatecas, Zacatecas, México
| | - Jesús Adrián López
- MicroRNAs Laboratory, Academic Unit for Biological Sciences, Autonomous University of Zacatecas, 98000, Zacatecas, Mexico
| | - José Antonio Enciso Moreno
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, 98000, Zacatecas, México
| | - David S Wishart
- The Metabolomics Innovation Center, University of Alberta, Edmonton, AB, T6G1C9, Canada
| |
Collapse
|
120
|
Masuda R, Lodge S, Nitschke P, Spraul M, Schaefer H, Bong SH, Kimhofer T, Hall D, Loo RL, Bizkarguenaga M, Bruzzone C, Gil-Redondo R, Embade N, Mato JM, Holmes E, Wist J, Millet O, Nicholson JK. Integrative Modeling of Plasma Metabolic and Lipoprotein Biomarkers of SARS-CoV-2 Infection in Spanish and Australian COVID-19 Patient Cohorts. J Proteome Res 2021; 20:4139-4152. [PMID: 34251833 DOI: 10.1021/acs.jproteome.1c00458] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Quantitative plasma lipoprotein and metabolite profiles were measured on an autonomous community of the Basque Country (Spain) cohort consisting of hospitalized COVID-19 patients (n = 72) and a matched control group (n = 75) and a Western Australian (WA) cohort consisting of (n = 17) SARS-CoV-2 positives and (n = 20) healthy controls using 600 MHz 1H nuclear magnetic resonance (NMR) spectroscopy. Spanish samples were measured in two laboratories using one-dimensional (1D) solvent-suppressed and T2-filtered methods with in vitro diagnostic quantification of lipoproteins and metabolites. SARS-CoV-2 positive patients and healthy controls from both populations were modeled and cross-projected to estimate the biological similarities and validate biomarkers. Using the top 15 most discriminatory variables enabled construction of a cross-predictive model with 100% sensitivity and specificity (within populations) and 100% sensitivity and 82% specificity (between populations). Minor differences were observed between the control metabolic variables in the two cohorts, but the lipoproteins were virtually indistinguishable. We observed highly significant infection-related reductions in high-density lipoprotein (HDL) subfraction 4 phospholipids, apolipoproteins A1 and A2,that have previously been associated with negative regulation of blood coagulation and fibrinolysis. The Spanish and Australian diagnostic SARS-CoV-2 biomarkers were mathematically and biologically equivalent, demonstrating that NMR-based technologies are suitable for the study of the comparative pathology of COVID-19 via plasma phenotyping.
Collapse
Affiliation(s)
- Reika Masuda
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Samantha Lodge
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Manfred Spraul
- Bruker Biospin GmbH, Silberstreifen, Ettlingen 76275, Germany
| | | | - Sze-How Bong
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Drew Hall
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Ruey Leng Loo
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Maider Bizkarguenaga
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Chiara Bruzzone
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Rubén Gil-Redondo
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Nieves Embade
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - José M Mato
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Section for Nutrition Research, Department of Metabolism, Nutrition and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, U.K
| | - Julien Wist
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Chemistry Department, Universidad del Valle, 76001 Cali, Colombia
| | - Oscar Millet
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Jeremy K Nicholson
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Institute of Global Health Innovation, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K
| |
Collapse
|
121
|
Boffito M, Back DJ, Flexner C, Sjö P, Blaschke TF, Horby PW, Cattaneo D, Acosta EP, Anderson P, Owen A. Toward Consensus on Correct Interpretation of Protein Binding in Plasma and Other Biological Matrices for COVID-19 Therapeutic Development. Clin Pharmacol Ther 2021; 110:64-68. [PMID: 33113246 PMCID: PMC8359231 DOI: 10.1002/cpt.2099] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/17/2020] [Indexed: 12/19/2022]
Abstract
The urgent global public health need presented by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has brought scientists from diverse backgrounds together in an unprecedented international effort to rapidly identify interventions. There is a pressing need to apply clinical pharmacology principles and this has already been recognized by several other groups. However, one area that warrants additional specific consideration relates to plasma and tissue protein binding that broadly influences pharmacokinetics and pharmacodynamics. The principles of free drug theory have been forged and applied across drug development but are not currently being routinely applied for SARS-CoV-2 antiviral drugs. Consideration of protein binding is of critical importance to candidate selection but requires correct interpretation, in a drug-specific manner, to avoid either underinterpretation or overinterpretation of its consequences. This paper represents a consensus from international researchers seeking to apply historical knowledge, which has underpinned highly successful antiviral drug development for other viruses, such as HIV and hepatitis C virus for decades.
Collapse
Affiliation(s)
- Marta Boffito
- Chelsea & Westminster HospitalLondonUK
- Department of Infectious DiseaseImperial College LondonLondonUK
| | - David J. Back
- Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Charles Flexner
- Bloomberg School of Public HealthJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Peter Sjö
- Drugs for Neglected Diseases Initiative (DNDi)GenevaSwitzerland
| | - Terrence F. Blaschke
- Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| | - Peter W. Horby
- Centre for Tropical Medicine and Global HealthNuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Dario Cattaneo
- Unit of Clinical PharmacologyASST FatebenefratelliSacco University HospitalMilanItaly
| | - Edward P. Acosta
- Department of Pharmacology and ToxicologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Peter Anderson
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of ColoradoAuroraColoradoUSA
| | - Andrew Owen
- Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
- Centre of Excellence in Long‐acting Therapeutics (CELT)University of LiverpoolUK
| |
Collapse
|
122
|
Tounta V, Liu Y, Cheyne A, Larrouy-Maumus G. Metabolomics in infectious diseases and drug discovery. Mol Omics 2021; 17:376-393. [PMID: 34125125 PMCID: PMC8202295 DOI: 10.1039/d1mo00017a] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022]
Abstract
Metabolomics has emerged as an invaluable tool that can be used along with genomics, transcriptomics and proteomics to understand host-pathogen interactions at small-molecule levels. Metabolomics has been used to study a variety of infectious diseases and applications. The most common application of metabolomics is for prognostic and diagnostic purposes, specifically the screening of disease-specific biomarkers by either NMR-based or mass spectrometry-based metabolomics. In addition, metabolomics is of great significance for the discovery of druggable metabolic enzymes and/or metabolic regulators through the use of state-of-the-art flux analysis, for example, via the elucidation of metabolic mechanisms. This review discusses the application of metabolomics technologies to biomarker screening, the discovery of drug targets in infectious diseases such as viral, bacterial and parasite infections and immunometabolomics, highlights the challenges associated with accessing metabolite compartmentalization and discusses the available tools for determining local metabolite concentrations.
Collapse
Affiliation(s)
- Vivian Tounta
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College LondonLondonUK
| | - Yi Liu
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College LondonLondonUK
| | - Ashleigh Cheyne
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College LondonLondonUK
| | - Gerald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College LondonLondonUK
| |
Collapse
|
123
|
Praissman JL, Wells L. Proteomics-Based Insights Into the SARS-CoV-2-Mediated COVID-19 Pandemic: A Review of the First Year of Research. Mol Cell Proteomics 2021; 20:100103. [PMID: 34089862 PMCID: PMC8176883 DOI: 10.1016/j.mcpro.2021.100103] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/24/2021] [Indexed: 02/08/2023] Open
Abstract
In late 2019, a virus subsequently named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in China and led to a worldwide pandemic of the disease termed coronavirus disease 2019. The global health threat posed by this pandemic led to an extremely rapid and robust mobilization of the scientific and medical communities as evidenced by the publication of more than 10,000 peer-reviewed articles and thousands of preprints in the first year of the pandemic alone. With the publication of the initial genome sequence of SARS-CoV-2, the proteomics community immediately joined this effort publishing, to date, more than 100 peer-reviewed proteomics studies and submitting many more preprints to preprint servers. In this review, we focus on peer-reviewed articles published on the proteome, glycoproteome, and glycome of SARS-CoV-2. At a basic level, proteomic studies provide valuable information on quantitative aspects of viral infection course; information on the identities, sites, and microheterogeneity of post-translational modifications; and, information on protein-protein interactions. At a biological systems level, these studies elucidate host cell and tissue responses, characterize antibodies and other immune system factors in infection, suggest biomarkers that may be useful for diagnosis and disease-course monitoring, and help in the development or repurposing of potential therapeutics. Here, we summarize results from selected early studies to provide a perspective on the current rapidly evolving literature.
Collapse
Affiliation(s)
- Jeremy L Praissman
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
124
|
Holmes E, Wist J, Masuda R, Lodge S, Nitschke P, Kimhofer T, Loo RL, Begum S, Boughton B, Yang R, Morillon AC, Chin ST, Hall D, Ryan M, Bong SH, Gay M, Edgar DW, Lindon JC, Richards T, Yeap BB, Pettersson S, Spraul M, Schaefer H, Lawler NG, Gray N, Whiley L, Nicholson JK. Incomplete Systemic Recovery and Metabolic Phenoreversion in Post-Acute-Phase Nonhospitalized COVID-19 Patients: Implications for Assessment of Post-Acute COVID-19 Syndrome. J Proteome Res 2021; 20:3315-3329. [PMID: 34009992 PMCID: PMC8147448 DOI: 10.1021/acs.jproteome.1c00224] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Indexed: 12/15/2022]
Abstract
We present a multivariate metabotyping approach to assess the functional recovery of nonhospitalized COVID-19 patients and the possible biochemical sequelae of "Post-Acute COVID-19 Syndrome", colloquially known as long-COVID. Blood samples were taken from patients ca. 3 months after acute COVID-19 infection with further assessment of symptoms at 6 months. Some 57% of the patients had one or more persistent symptoms including respiratory-related symptoms like cough, dyspnea, and rhinorrhea or other nonrespiratory symptoms including chronic fatigue, anosmia, myalgia, or joint pain. Plasma samples were quantitatively analyzed for lipoproteins, glycoproteins, amino acids, biogenic amines, and tryptophan pathway intermediates using Nuclear Magnetic Resonance (NMR) spectroscopy and mass spectrometry. Metabolic data for the follow-up patients (n = 27) were compared with controls (n = 41) and hospitalized severe acute respiratory syndrome SARS-CoV-2 positive patients (n = 18, with multiple time-points). Univariate and multivariate statistics revealed variable patterns of functional recovery with many patients exhibiting residual COVID-19 biomarker signatures. Several parameters were persistently perturbed, e.g., elevated taurine (p = 3.6 × 10-3 versus controls) and reduced glutamine/glutamate ratio (p = 6.95 × 10-8 versus controls), indicative of possible liver and muscle damage and a high energy demand linked to more generalized tissue repair or immune function. Some parameters showed near-complete normalization, e.g., the plasma apolipoprotein B100/A1 ratio was similar to that of healthy controls but significantly lower (p = 4.2 × 10-3) than post-acute COVID-19 patients, reflecting partial reversion of the metabolic phenotype (phenoreversion) toward the healthy metabolic state. Plasma neopterin was normalized in all follow-up patients, indicative of a reduction in the adaptive immune activity that has been previously detected in active SARS-CoV-2 infection. Other systemic inflammatory biomarkers such as GlycA and the kynurenine/tryptophan ratio remained elevated in some, but not all, patients. Correlation analysis, principal component analysis (PCA), and orthogonal-partial least-squares discriminant analysis (O-PLS-DA) showed that the follow-up patients were, as a group, metabolically distinct from controls and partially comapped with the acute-phase patients. Significant systematic metabolic differences between asymptomatic and symptomatic follow-up patients were also observed for multiple metabolites. The overall metabolic variance of the symptomatic patients was significantly greater than that of nonsymptomatic patients for multiple parameters (χ2p = 0.014). Thus, asymptomatic follow-up patients including those with post-acute COVID-19 Syndrome displayed a spectrum of multiple persistent biochemical pathophysiology, suggesting that the metabolic phenotyping approach may be deployed for multisystem functional assessment of individual post-acute COVID-19 patients.
Collapse
Affiliation(s)
- Elaine Holmes
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
- Department of Metabolism, Digestion, and Reproduction,
Faculty of Medicine, Imperial College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Julien Wist
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
- Chemistry Department, Universidad del
Valle, 76001 Cali, Colombia
| | - Reika Masuda
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Samantha Lodge
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Ruey Leng Loo
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Sofina Begum
- Department of Metabolism, Digestion, and Reproduction,
Faculty of Medicine, Imperial College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Berin Boughton
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Rongchang Yang
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Aude-Claire Morillon
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Sung-Tong Chin
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Drew Hall
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Monique Ryan
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Sze-How Bong
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Melvin Gay
- Bruker Pty. Ltd., Preston,
VIC 3072, Australia
| | - Dale W. Edgar
- State Adult Burn Unit, Fiona Stanley
Hospital, Murdoch, WA 6150, Australia
- Burn Injury Research Node, The University
of Notre Dame, Fremantle, WA 6160, Australia
| | - John C. Lindon
- Department of Surgery and Cancer, Faculty of
Medicine, Imperial College London, London SW7 2AZ,
U.K.
| | - Toby Richards
- Department of Surgery, Fiona Stanley Hospital, Medical
School, University of Western Australia,Harry Perkins Building,
Murdoch, Perth, WA 6150, Australia
| | - Bu B. Yeap
- Department of Endocrinology and Diabetes, Fiona
Stanley Hospital, Medical School, University of Western
Australia, Harry Perkins Building, Murdoch, Perth, WA 6150,
Australia
| | - Sven Pettersson
- Singapore National NeuroScience
Centre, Mandalay Road, Singapore 308232,
Singapore
- Lee Kong Chian School of Medicine.
Nanyang Technological University, Mandalay Road, Singapore
308232, Singapore
- Department of Life Science Centre,
Sunway University, Kuala Lumpur 47500,
Malaysia
| | | | | | - Nathan G. Lawler
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Nicola Gray
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Luke Whiley
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Perron Institute for Neurological and
Translational Science, Nedlands, WA 6009,
Australia
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
- Institute of Global Health Innovation,
Imperial College London, Level 1, Faculty Building, South
Kensington Campus, London SW7 2AZ, U.K.
| |
Collapse
|
125
|
Dagenais-Lussier X, Loucif H, Beji C, Telittchenko R, Routy JP, van Grevenynghe J. Latest developments in tryptophan metabolism: Understanding its role in B cell immunity. Cytokine Growth Factor Rev 2021; 59:111-117. [DOI: 10.1016/j.cytogfr.2021.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
|
126
|
Wendt R, Thijs L, Kalbitz S, Mischak H, Siwy J, Raad J, Metzger J, Neuhaus B, Leyen HVD, Dudoignon E, Mebazaa A, Spasovski G, Milenkova M, Canevska-Talevska A, Czerwieńska B, Wiecek A, Peters B, Nilsson Å, Schwab M, Rothfuss K, Lübbert C, Staessen JA, Beige J. A urinary peptidomic profile predicts outcome in SARS-CoV-2-infected patients. EClinicalMedicine 2021; 36:100883. [PMID: 33969282 PMCID: PMC8092440 DOI: 10.1016/j.eclinm.2021.100883] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/07/2021] [Accepted: 04/16/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COVID-19 prediction models based on clinical characteristics, routine biochemistry and imaging, have been developed, but little is known on proteomic markers reflecting the molecular pathophysiology of disease progression. METHODS The multicentre (six European study sites) Prospective Validation of a Proteomic Urine Test for Early and Accurate Prognosis of Critical Course Complications in Patients with SARS-CoV-2 Infection Study (Crit-COV-U) is recruiting consecutive patients (≥ 18 years) with PCR-confirmed SARS-CoV-2 infection. A urinary proteomic biomarker (COV50) developed by capillary-electrophoresis-mass spectrometry (CE-MS) technology, comprising 50 sequenced peptides and identifying the parental proteins, was evaluated in 228 patients (derivation cohort) with replication in 99 patients (validation cohort). Death and progression along the World Health Organization (WHO) Clinical Progression Scale were assessed up to 21 days after the initial PCR test. Statistical methods included logistic regression, receiver operating curve (ROC) analysis and comparison of the area under the curve (AUC). FINDINGS In the derivation cohort, 23 patients died, and 48 developed worse WHO scores. The odds ratios (OR) for death per 1 standard deviation (SD) increment in COV50 were 3·52 (95% CI, 2·02-6·13, p <0·0001) unadjusted and 2·73 (1·25-5·95, p = 0·012) adjusted for sex, age, baseline WHO score, body mass index (BMI) and comorbidities. For WHO scale progression, the corresponding OR were 2·63 (1·80-3·85, p<0·0001) and 3·38 (1·85-6·17, p<0·0001), respectively. The area under the curve (AUC) for COV50 as a continuously distributed variable was 0·80 (0·72-0·88) for mortality and 0·74 (0·66-0·81) for worsening WHO score. The optimised COV50 thresholds for mortality and worsening WHO score were 0·47 and 0·04 with sensitivity/specificity of 87·0 (74·6%) and 77·1 (63·9%), respectively. On top of covariates, COV50 improved the AUC, albeit borderline for death, from 0·78 to 0·82 (p = 0·11) and 0·84 (p = 0·052) for mortality and from 0·68 to 0·78 (p = 0·0097) and 0·75 (p = 0·021) for worsening WHO score. The validation cohort findings were confirmatory. INTERPRETATION This first CRIT-COV-U report proves the concept that urinary proteomic profiling generates biomarkers indicating adverse COVID-19 outcomes, even at an early disease stage, including WHO stages 1-3. These findings need to be consolidated in an upcoming final dataset. FUNDING The German Federal Ministry of Health funded the study.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany
| | - Lutgarde Thijs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Diseases, University of Leuven, Belgium
| | - Sven Kalbitz
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany
| | - Harald Mischak
- Mosaiques-Diagnostics GmbH, Hannover, Germany
- Institute of Cardiovascular and Medical Sciences, Glasgow, United Kingdom
| | | | - Julia Raad
- Mosaiques-Diagnostics GmbH, Hannover, Germany
| | | | - Barbara Neuhaus
- Hannover Clinical Trial Center, Medizinische Hochschule, Hannover, Germany
| | | | - Emmanuel Dudoignon
- Department of Anaesthesiology and Intensive Care, Hôpital Saint Louis-Lariboisière, U942 Inserm MASCOT, Université de Paris, Paris, France
| | - Alexandre Mebazaa
- Department of Anaesthesiology and Intensive Care, Hôpital Saint Louis-Lariboisière, U942 Inserm MASCOT, Université de Paris, Paris, France
| | - Goce Spasovski
- Department Nephrology, Cyril and Methodius University, Skopje, Republic of North Macedonia
| | - Mimoza Milenkova
- Department Nephrology, Cyril and Methodius University, Skopje, Republic of North Macedonia
| | | | - Beata Czerwieńska
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Björn Peters
- Department of Nephrology, Skaraborg Hospital, Skövde, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Åsa Nilsson
- Research and Development Centre (FoU), Skaraborg Hospital, Skövde, Sweden
| | - Matthias Schwab
- Margarete-Fischer-Bosch Institute for Clinical Pharmacology and Department for Gastroenterology, Hepatology and Endocrinology, Robert Bosch Hospital, Stuttgart, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University of Tuebingen, Germany
| | - Katja Rothfuss
- Margarete-Fischer-Bosch Institute for Clinical Pharmacology and Department for Gastroenterology, Hepatology and Endocrinology, Robert Bosch Hospital, Stuttgart, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany
- Division of Infectious Diseases and Tropical Medicine, Leipzig University Medical Center, Leipzig, Germany
| | - Jan A. Staessen
- Non-Profit Research Institute Alliance for the Promotion of Preventive Medicine, Mechelen, Belgium
- Belgium, Biomedical Sciences Group, Faculty of Medicine, University of Leuven, Leuven, Belgium
| | - Joachim Beige
- Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany
- Martin-Luther-University Halle-Wittenberg, Halle an der Saale, Germany
- Corresponding author at: Department of Infectious Diseases/Tropical Medicine, Nephrology/KfH Renal Unit and Rheumatology, St. Georg Hospital Leipzig, Delitzscher Strasse 141, Leipzig DE 04129, Germany.
| | | |
Collapse
|
127
|
Collier ME, Zhang S, Scrutton NS, Giorgini F. Inflammation control and improvement of cognitive function in COVID-19 infections: is there a role for kynurenine 3-monooxygenase inhibition? Drug Discov Today 2021; 26:1473-1481. [PMID: 33609782 PMCID: PMC7889466 DOI: 10.1016/j.drudis.2021.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/26/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
The novel respiratory virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), emerged during late 2019 and spread rapidly across the world. It is now recognised that the nervous system can be affected in COVID-19, with several studies reporting long-term cognitive problems in patients. The metabolic pathway of tryptophan degradation, known as the kynurenine pathway (KP), is significantly activated in patients with COVID-19. KP metabolites have roles in regulating both inflammatory/immune responses and neurological functions. In this review, we speculate on the effects of KP activation in patients with COVID-19, and how modulation of this pathway might impact inflammation and reduce neurological symptoms.
Collapse
Affiliation(s)
- Mary Ew Collier
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK.
| | - Shaowei Zhang
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Nigel S Scrutton
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
128
|
Lawler NG, Gray N, Kimhofer T, Boughton B, Gay M, Yang R, Morillon AC, Chin ST, Ryan M, Begum S, Bong SH, Coudert JD, Edgar D, Raby E, Pettersson S, Richards T, Holmes E, Whiley L, Nicholson JK. Systemic Perturbations in Amine and Kynurenine Metabolism Associated with Acute SARS-CoV-2 Infection and Inflammatory Cytokine Responses. J Proteome Res 2021; 20:2796-2811. [PMID: 33724837 PMCID: PMC7986977 DOI: 10.1021/acs.jproteome.1c00052] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Indexed: 01/06/2023]
Abstract
We performed quantitative metabolic phenotyping of blood plasma in parallel with cytokine/chemokine analysis from participants who were either SARS-CoV-2 (+) (n = 10) or SARS-CoV-2 (-) (n = 49). SARS-CoV-2 positivity was associated with a unique metabolic phenotype and demonstrated a complex systemic response to infection, including severe perturbations in amino acid and kynurenine metabolic pathways. Nine metabolites were elevated in plasma and strongly associated with infection (quinolinic acid, glutamic acid, nicotinic acid, aspartic acid, neopterin, kynurenine, phenylalanine, 3-hydroxykynurenine, and taurine; p < 0.05), while four metabolites were lower in infection (tryptophan, histidine, indole-3-acetic acid, and citrulline; p < 0.05). This signature supports a systemic metabolic phenoconversion following infection, indicating possible neurotoxicity and neurological disruption (elevations of 3-hydroxykynurenine and quinolinic acid) and liver dysfunction (reduction in Fischer's ratio and elevation of taurine). Finally, we report correlations between the key metabolite changes observed in the disease with concentrations of proinflammatory cytokines and chemokines showing strong immunometabolic disorder in response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nathan G. Lawler
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Nicola Gray
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Berin Boughton
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Melvin Gay
- Bruker Pty Ltd., Preston,
VIC 3072, Australia
| | - Rongchang Yang
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Aude-Claire Morillon
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Sung-Tong Chin
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Monique Ryan
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Sofina Begum
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
- Department of Metabolism Digestion and Reproduction,
Faculty of Medicine, Imperial College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Sze How Bong
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Jerome D. Coudert
- Centre for Molecular Medicine & Innovative
Therapeutics, Murdoch University, Perth, WA 6150,
Australia
| | - Dale Edgar
- State Adult Burn Unit, Fiona Stanley
Hospital, Murdoch, WA 6150, Australia
- Burn Injury Research Node, The University of
Notre Dame, Fremantle, WA 6160, Australia
- Fiona Wood Foundation,
Murdoch, WA 6150, Australia
| | - Edward Raby
- Department of Microbiology, PathWest
Laboratory Medicine, Perth, WA 6009, Australia
- Department of Infectious Diseases, Fiona
Stanley Hospital, Perth, WA 6150, Australia
| | - Sven Pettersson
- Singapore National Neuro Science
Centre, Singapore Mandalay Road, Singapore 308232,
Singapore
- Lee Kong Chian School of Medicine,
Nanyang Technological University, Mandalay Road, Singapore
308232, Singapore
- Department of Life Science Centre,
Sunway University, 55100 Kuala Lumpur,
Malaysia
| | - Toby Richards
- Medical School, Faculty of Health and Medical
Sciences, University of Western Australia, Nedlands, WA 6009,
Australia
| | - Elaine Holmes
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
- Department of Metabolism Digestion and Reproduction,
Faculty of Medicine, Imperial College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Luke Whiley
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
- Perron Institute for Neurological and
Translational Science, Nedlands, WA 6009,
Australia
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
- Medical School, Faculty of Health and Medical
Sciences, University of Western Australia, Nedlands, WA 6009,
Australia
- Institute of Global Health Innovation,
Imperial College London, Level 1, Faculty Building South
Kensington Campus, London SW7 2AZ, U.K.
| |
Collapse
|
129
|
Suvarna K, Biswas D, Pai MGJ, Acharjee A, Bankar R, Palanivel V, Salkar A, Verma A, Mukherjee A, Choudhury M, Ghantasala S, Ghosh S, Singh A, Banerjee A, Badaya A, Bihani S, Loya G, Mantri K, Burli A, Roy J, Srivastava A, Agrawal S, Shrivastav O, Shastri J, Srivastava S. Proteomics and Machine Learning Approaches Reveal a Set of Prognostic Markers for COVID-19 Severity With Drug Repurposing Potential. Front Physiol 2021; 12:652799. [PMID: 33995121 PMCID: PMC8120435 DOI: 10.3389/fphys.2021.652799] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
The pestilential pathogen SARS-CoV-2 has led to a seemingly ceaseless pandemic of COVID-19. The healthcare sector is under a tremendous burden, thus necessitating the prognosis of COVID-19 severity. This in-depth study of plasma proteome alteration provides insights into the host physiological response towards the infection and also reveals the potential prognostic markers of the disease. Using label-free quantitative proteomics, we performed deep plasma proteome analysis in a cohort of 71 patients (20 COVID-19 negative, 18 COVID-19 non-severe, and 33 severe) to understand the disease dynamics. Of the 1200 proteins detected in the patient plasma, 38 proteins were identified to be differentially expressed between non-severe and severe groups. The altered plasma proteome revealed significant dysregulation in the pathways related to peptidase activity, regulated exocytosis, blood coagulation, complement activation, leukocyte activation involved in immune response, and response to glucocorticoid biological processes in severe cases of SARS-CoV-2 infection. Furthermore, we employed supervised machine learning (ML) approaches using a linear support vector machine model to identify the classifiers of patients with non-severe and severe COVID-19. The model used a selected panel of 20 proteins and classified the samples based on the severity with a classification accuracy of 0.84. Putative biomarkers such as angiotensinogen and SERPING1 and ML-derived classifiers including the apolipoprotein B, SERPINA3, and fibrinogen gamma chain were validated by targeted mass spectrometry-based multiple reaction monitoring (MRM) assays. We also employed an in silico screening approach against the identified target proteins for the therapeutic management of COVID-19. We shortlisted two FDA-approved drugs, namely, selinexor and ponatinib, which showed the potential of being repurposed for COVID-19 therapeutics. Overall, this is the first most comprehensive plasma proteome investigation of COVID-19 patients from the Indian population, and provides a set of potential biomarkers for the disease severity progression and targets for therapeutic interventions.
Collapse
Affiliation(s)
- Kruthi Suvarna
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Deeptarup Biswas
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Medha Gayathri J. Pai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Arup Acharjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Renuka Bankar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Viswanthram Palanivel
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Akanksha Salkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ayushi Verma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Amrita Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Manisha Choudhury
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Saicharan Ghantasala
- Centre for Research in Nanotechnology and Sciences, Indian Institute of Technology Bombay, Mumbai, India
| | - Susmita Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Avinash Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Arghya Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Apoorva Badaya
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India
| | - Surbhi Bihani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Gaurish Loya
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Krishi Mantri
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ananya Burli
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Jyotirmoy Roy
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Alisha Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Department of Genetics, University of Delhi, New Delhi, India
| | - Sachee Agrawal
- Kasturba Hospital for Infectious Diseases, Mumbai, India
| | - Om Shrivastav
- Kasturba Hospital for Infectious Diseases, Mumbai, India
| | | | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
130
|
Wu J, Zhao M, Li C, Zhang Y, Wang DW. The SARS-CoV-2 induced targeted amino acid profiling in patients at hospitalized and convalescent stage. Biosci Rep 2021; 41:BSR20204201. [PMID: 33625490 PMCID: PMC7955102 DOI: 10.1042/bsr20204201] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/10/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has induced an ongoing global health crisis. Here we utilized a combination of targeted amino acids (AAs) and clinical biochemical profiling to analyze the plasma of coronavirus disease 2019 (COVID-19) subjects at the hospitalization stage and 1-month post-infection convalescent stage, respectively, to investigate the systematic injury during COVID-19 disease progress. We found the virus-induced inflammatory status and reduced liver synthesis capacity in hospitalized patients, which manifested with increased branched-chain AAs (BCAAs), aromatic AAs (AAAs), one-carbon related metabolites, and decreased methionine. Most of these disturbances during infection recover except for the increased levels of medium-chain acylcarnitines (ACs) in the convalescent subjects, implying the existence of incomplete fatty acids oxidation during recovery periods. Our results suggested that the imbalance of the AA profiling in COVID-19 patients. The majority of disturbed AAs recovered in 1 month. The incomplete fatty acid oxidation products suggested it might take longer time for convalescent patients to get complete recovery.
Collapse
Affiliation(s)
- Junfang Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center, Peking University, Beijing 100191, China
| | - Chenze Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yuxuan Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| |
Collapse
|
131
|
Feingold KR. The bidirectional link between HDL and COVID-19 infections. J Lipid Res 2021; 62:100067. [PMID: 33741421 PMCID: PMC7963524 DOI: 10.1016/j.jlr.2021.100067] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/21/2022] Open
Affiliation(s)
- Kenneth R Feingold
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
132
|
Lodge S, Nitschke P, Kimhofer T, Wist J, Bong SH, Loo RL, Masuda R, Begum S, Richards T, Lindon JC, Bermel W, Reinsperger T, Schaefer H, Spraul M, Holmes E, Nicholson JK. Diffusion and Relaxation Edited Proton NMR Spectroscopy of Plasma Reveals a High-Fidelity Supramolecular Biomarker Signature of SARS-CoV-2 Infection. Anal Chem 2021; 93:3976-3986. [PMID: 33577736 PMCID: PMC7908063 DOI: 10.1021/acs.analchem.0c04952] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
We have applied nuclear magnetic resonance spectroscopy based plasma phenotyping to reveal diagnostic molecular signatures of SARS-CoV-2 infection via combined diffusional and relaxation editing (DIRE). We compared plasma from healthy age-matched controls (n = 26) with SARS-CoV-2 negative non-hospitalized respiratory patients and hospitalized respiratory patients (n = 23 and 11 respectively) with SARS-CoV-2 rRT-PCR positive respiratory patients (n = 17, with longitudinal sampling time-points). DIRE data were modelled using principal component analysis and orthogonal projections to latent structures discriminant analysis (O-PLS-DA), with statistical cross-validation indices indicating excellent model generalization for the classification of SARS-CoV-2 positivity for all comparator groups (area under the receiver operator characteristic curve = 1). DIRE spectra show biomarker signal combinations conferred by differential concentrations of metabolites with selected molecular mobility properties. These comprise the following: (a) composite N-acetyl signals from α-1-acid glycoprotein and other glycoproteins (designated GlycA and GlycB) that were elevated in SARS-CoV-2 positive patients [p = 2.52 × 10-10 (GlycA) and 1.25 × 10-9 (GlycB) vs controls], (b) two diagnostic supramolecular phospholipid composite signals that were identified (SPC-A and SPC-B) from the -+N-(CH3)3 choline headgroups of lysophosphatidylcholines carried on plasma glycoproteins and from phospholipids in high-density lipoprotein subfractions (SPC-A) together with a phospholipid component of low-density lipoprotein (SPC-B). The integrals of the summed SPC signals (SPCtotal) were reduced in SARS-CoV-2 positive patients relative to both controls (p = 1.40 × 10-7) and SARS-CoV-2 negative patients (p = 4.52 × 10-8) but were not significantly different between controls and SARS-CoV-2 negative patients. The identity of the SPC signal components was determined using one and two dimensional diffusional, relaxation, and statistical spectroscopic experiments. The SPCtotal/GlycA ratios were also significantly different for control versus SARS-CoV-2 positive patients (p = 1.23 × 10-10) and for SARS-CoV-2 negatives versus positives (p = 1.60 × 10-9). Thus, plasma SPCtotal and SPCtotal/GlycA are proposed as sensitive molecular markers for SARS-CoV-2 positivity that could effectively augment current COVID-19 diagnostics and may have value in functional assessment of the disease recovery process in patients with long-term symptoms.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Philipp Nitschke
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Torben Kimhofer
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Julien Wist
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
| | - Sze-How Bong
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Ruey Leng Loo
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Reika Masuda
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Sofina Begum
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
- Department
of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Toby Richards
- Division
of Surgery, Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Harry Perkins Building, Murdoch, Perth WA6150, Australia
| | - John C. Lindon
- Department
of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Wolfgang Bermel
- Bruker
Biospin GmbH, Silberstreifen, Ettlingen 76275, Germany
| | | | | | - Manfred Spraul
- Bruker
Biospin GmbH, Silberstreifen, Ettlingen 76275, Germany
| | - Elaine Holmes
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
- Department
of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Jeremy K. Nicholson
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
- Division
of Surgery, Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Harry Perkins Building, Murdoch, Perth WA6150, Australia
- Institute
of Global Health Innovation, Faculty of Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K.
| |
Collapse
|
133
|
Bittremieux W, Adams C, Laukens K, Dorrestein PC, Bandeira N. Open Science Resources for the Mass Spectrometry-Based Analysis of SARS-CoV-2. J Proteome Res 2021; 20:1464-1475. [PMID: 33605735 DOI: 10.1021/acs.jproteome.0c00929] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The SARS-CoV-2 virus is the causative agent of the 2020 pandemic leading to the COVID-19 respiratory disease. With many scientific and humanitarian efforts ongoing to develop diagnostic tests, vaccines, and treatments for COVID-19, and to prevent the spread of SARS-CoV-2, mass spectrometry research, including proteomics, is playing a role in determining the biology of this viral infection. Proteomics studies are starting to lead to an understanding of the roles of viral and host proteins during SARS-CoV-2 infection, their protein-protein interactions, and post-translational modifications. This is beginning to provide insights into potential therapeutic targets or diagnostic strategies that can be used to reduce the long-term burden of the pandemic. However, the extraordinary situation caused by the global pandemic is also highlighting the need to improve mass spectrometry data and workflow sharing. We therefore describe freely available data and computational resources that can facilitate and assist the mass spectrometry-based analysis of SARS-CoV-2. We exemplify this by reanalyzing a virus-host interactome data set to detect protein-protein interactions and identify host proteins that could potentially be used as targets for drug repurposing.
Collapse
Affiliation(s)
- Wout Bittremieux
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla 92093, California, United States.,Department of Computer Science, University of Antwerp, Antwerp 2020, Belgium
| | - Charlotte Adams
- Department of Computer Science, University of Antwerp, Antwerp 2020, Belgium
| | - Kris Laukens
- Department of Computer Science, University of Antwerp, Antwerp 2020, Belgium
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla 92093, California, United States
| | - Nuno Bandeira
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla 92093, California, United States.,Department of Computer Science and Engineering, University of California San Diego, La Jolla 92093, California, United States
| |
Collapse
|
134
|
Sindelar M, Stancliffe E, Schwaiger-Haber M, Anbukumar DS, Albrecht RA, Liu WC, Travis KA, García-Sastre A, Shriver LP, Patti GJ. Longitudinal Metabolomics of Human Plasma Reveals Robust Prognostic Markers of COVID-19 Disease Severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.02.05.21251173. [PMID: 33564793 PMCID: PMC7872388 DOI: 10.1101/2021.02.05.21251173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
There is an urgent need to identify which COVID-19 patients will develop life-threatening illness so that scarce medical resources can be optimally allocated and rapid treatment can be administered early in the disease course, when clinical management is most effective. To aid in the prognostic classification of disease severity, we performed untargeted metabolomics profiling of 341 patients with plasma samples collected at six longitudinal time points. Using the temporal metabolic profiles and machine learning, we then built a predictive model of disease severity. We determined that the levels of 25 metabolites measured at the time of hospital admission successfully predict future disease severity. Through analysis of longitudinal samples, we confirmed that these prognostic markers are directly related to disease progression and that their levels are restored to baseline upon disease recovery. Finally, we validated that these metabolites are also altered in a hamster model of COVID-19. Our results indicate that metabolic changes associated with COVID-19 severity can be effectively used to stratify patients and inform resource allocation during the pandemic.
Collapse
Affiliation(s)
- Miriam Sindelar
- Department of Chemistry, Washington University, St. Louis, MO
- Department of Medicine, Washington University, St. Louis, MO
- These authors contributed equally
| | - Ethan Stancliffe
- Department of Chemistry, Washington University, St. Louis, MO
- Department of Medicine, Washington University, St. Louis, MO
- These authors contributed equally
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Washington University, St. Louis, MO
- Department of Medicine, Washington University, St. Louis, MO
- These authors contributed equally
| | - Dhanalakshmi S. Anbukumar
- Department of Chemistry, Washington University, St. Louis, MO
- Department of Medicine, Washington University, St. Louis, MO
- These authors contributed equally
| | - Randy A. Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Wen-Chun Liu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York City, NY
- Current affiliation: Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York City, NY
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York City, NY
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Leah P. Shriver
- Department of Chemistry, Washington University, St. Louis, MO
- Department of Medicine, Washington University, St. Louis, MO
| | - Gary J. Patti
- Department of Chemistry, Washington University, St. Louis, MO
- Department of Medicine, Washington University, St. Louis, MO
- Siteman Cancer Center, Washington University, St. Louis, MO
| |
Collapse
|
135
|
Lodge S, Nitschke P, Kimhofer T, Coudert JD, Begum S, Bong SH, Richards T, Edgar D, Raby E, Spraul M, Schaefer H, Lindon JC, Loo RL, Holmes E, Nicholson JK. NMR Spectroscopic Windows on the Systemic Effects of SARS-CoV-2 Infection on Plasma Lipoproteins and Metabolites in Relation to Circulating Cytokines. J Proteome Res 2021; 20:1382-1396. [PMID: 33426894 PMCID: PMC7805607 DOI: 10.1021/acs.jproteome.0c00876] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 02/08/2023]
Abstract
To investigate the systemic metabolic effects of SARS-CoV-2 infection, we analyzed 1H NMR spectroscopic data on human blood plasma and co-modeled with multiple plasma cytokines and chemokines (measured in parallel). Thus, 600 MHz 1H solvent-suppressed single-pulse, spin-echo, and 2D J-resolved spectra were collected on plasma recorded from SARS-CoV-2 rRT-PCR-positive patients (n = 15, with multiple sampling timepoints) and age-matched healthy controls (n = 34, confirmed rRT-PCR negative), together with patients with COVID-19/influenza-like clinical symptoms who tested SARS-CoV-2 negative (n = 35). We compared the single-pulse NMR spectral data with in vitro diagnostic research (IVDr) information on quantitative lipoprotein profiles (112 parameters) extracted from the raw 1D NMR data. All NMR methods gave highly significant discrimination of SARS-CoV-2 positive patients from controls and SARS-CoV-2 negative patients with individual NMR methods, giving different diagnostic information windows on disease-induced phenoconversion. Longitudinal trajectory analysis in selected patients indicated that metabolic recovery was incomplete in individuals without detectable virus in the recovery phase. We observed four plasma cytokine clusters that expressed complex differential statistical relationships with multiple lipoproteins and metabolites. These included the following: cluster 1, comprising MIP-1β, SDF-1α, IL-22, and IL-1α, which correlated with multiple increased LDL and VLDL subfractions; cluster 2, including IL-10 and IL-17A, which was only weakly linked to the lipoprotein profile; cluster 3, which included IL-8 and MCP-1 and were inversely correlated with multiple lipoproteins. IL-18, IL-6, and IFN-γ together with IP-10 and RANTES exhibited strong positive correlations with LDL1-4 subfractions and negative correlations with multiple HDL subfractions. Collectively, these data show a distinct pattern indicative of a multilevel cellular immune response to SARS CoV-2 infection interacting with the plasma lipoproteome giving a strong and characteristic immunometabolic phenotype of the disease. We observed that some patients in the respiratory recovery phase and testing virus-free were still metabolically highly abnormal, which indicates a new role for these technologies in assessing full systemic recovery.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
| | - Jerome D. Coudert
- Centre for Molecular Medicine and Innovative
Therapeutics, Murdoch University, Harry Perkins Building,
Perth, Western Australia 6150, Australia
- Perron Institute for Neurological and
Translational Science, Nedlands, Western Australia 6009,
Australia
- School of Medicine, University of Notre
Dame, Fremantle, Western Australia 6160,
Australia
| | - Sofina Begum
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Section of Nutrition Research , Department of Metabolism,
Nutrition and Reproduction, Faculty of Medicine, Sir Alexander Fleming Building,
Imperial College London, London SW7 2AZ,
U.K.
| | - Sze-How Bong
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
| | - Toby Richards
- Division of Surgery, Medical School, Faculty of Health
and Medical Sciences, University of Western Australia, Harry
Perkins Building, Robert Warren Drive, Murdoch, Perth, Western Australia 6150,
Australia
| | - Dale Edgar
- Faculty of Health and Medical Sciences,
University of Western Australia, Harry Perkins Building,
Robert Warren Drive, Murdoch, Perth, Western Australia 6150,
Australia
| | - Edward Raby
- Department of Clinical Microbiology,
PathWest Laboratory Medicine WA, Murdoch, Perth, Western
Australia 6150, Australia
| | | | | | - John C. Lindon
- Division of Systems Medicine, Department of
Metabolism, Nutrition and Reproduction, Faculty of Medicine, Sir Alexander Fleming
Building, Imperial College London, London SW7 2AZ,
U.K.
| | - Ruey Leng Loo
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
- Section of Nutrition Research , Department of Metabolism,
Nutrition and Reproduction, Faculty of Medicine, Sir Alexander Fleming Building,
Imperial College London, London SW7 2AZ,
U.K.
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
- Division of Surgery, Medical School, Faculty of Health
and Medical Sciences, University of Western Australia, Harry
Perkins Building, Robert Warren Drive, Murdoch, Perth, Western Australia 6150,
Australia
- Institute of Global Health Innovation,
Imperial College London, Level 1, Faculty Building South
Kensington Campus, London SW7 2NA, U.K.
| |
Collapse
|
136
|
Lodge S, Nitschke P, Loo RL, Kimhofer T, Bong SH, Richards T, Begum S, Spraul M, Schaefer H, Lindon JC, Holmes E, Nicholson JK. Low Volume in Vitro Diagnostic Proton NMR Spectroscopy of Human Blood Plasma for Lipoprotein and Metabolite Analysis: Application to SARS-CoV-2 Biomarkers. J Proteome Res 2021; 20:1415-1423. [PMID: 33491459 PMCID: PMC7857136 DOI: 10.1021/acs.jproteome.0c00815] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Indexed: 11/29/2022]
Abstract
The utility of low sample volume in vitro diagnostic (IVDr) proton nuclear magnetic resonance (1H NMR) spectroscopic experiments on blood plasma for information recovery from limited availability or high value samples was exemplified using plasma from patients with SARS-CoV-2 infection and normal controls. 1H NMR spectra were obtained using solvent-suppressed 1D, spin-echo (CPMG), and 2-dimensional J-resolved (JRES) spectroscopy using both 3 mm outer diameter SampleJet NMR tubes (100 μL plasma) and 5 mm SampleJet NMR tubes (300 μL plasma) under in vitro diagnostic conditions. We noted near identical diagnostic models in both standard and low volume IVDr lipoprotein analysis (measuring 112 lipoprotein parameters) with a comparison of the two tubes yielding R2 values ranging between 0.82 and 0.99 for the 40 paired lipoprotein parameters samples. Lipoprotein measurements for the 3 mm tubes were achieved without time penalty over the 5 mm tubes as defined by biomarker recovery for SARS-CoV-2. Overall, biomarker pattern recovery for the lipoproteins was extremely similar, but there were some small positive offsets in the linear equations for several variables due to small shimming artifacts, but there was minimal degradation of the biological information. For the standard untargeted 1D, CPMG, and JRES NMR experiments on the same samples, the reduced signal-to-noise was more constraining and required greater scanning times to achieve similar differential diagnostic performance (15 min per sample per experiment for 3 mm 1D and CPMG, compared to 4 min for the 5 mm tubes). We conclude that the 3 mm IVDr method is fit-for-purpose for quantitative lipoprotein measurements, allowing the preparation of smaller volumes for high value or limited volume samples that is common in clinical studies. If there are no analytical time constraints, the lower volume experiments are equally informative for untargeted profiling.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Philipp Nitschke
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Ruey Leng Loo
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Sze-How Bong
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Toby Richards
- Division
of Surgery, Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Harry Perkins Building, Robert Warren
Drive, Murdoch, Perth, WA 6150, Australia
| | - Sofina Begum
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Division
of Systems Medicine, Department of Metabolism, Nutrition and Reproduction,
Sir Alexander Fleming Building, Imperial
College London, London SW7 2AZ, U.K.
| | | | | | - John C. Lindon
- Division
of Systems Medicine, Department of Metabolism, Nutrition and Reproduction,
Sir Alexander Fleming Building, Imperial
College London, London SW7 2AZ, U.K.
| | - Elaine Holmes
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Division
of Systems Medicine, Department of Metabolism, Nutrition and Reproduction,
Sir Alexander Fleming Building, Imperial
College London, London SW7 2AZ, U.K.
| | - Jeremy K. Nicholson
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Institute
of Global Health Innovation, Imperial College
London, Level 1, Faculty Building South Kensington Campus, London SW7 2NA, U.K.
| |
Collapse
|
137
|
Gray N, Lawler NG, Yang R, Morillon AC, Gay MC, Bong SH, Holmes E, Nicholson JK, Whiley L. A simultaneous exploratory and quantitative amino acid and biogenic amine metabolic profiling platform for rapid disease phenotyping via UPLC-QToF-MS. Talanta 2021; 223:121872. [DOI: 10.1016/j.talanta.2020.121872] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/26/2022]
|
138
|
Metabolomic/lipidomic profiling of COVID-19 and individual response to tocilizumab. PLoS Pathog 2021; 17:e1009243. [PMID: 33524041 PMCID: PMC7877736 DOI: 10.1371/journal.ppat.1009243] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/11/2021] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
The current pandemic emergence of novel coronavirus disease (COVID-19) poses a relevant threat to global health. SARS-CoV-2 infection is characterized by a wide range of clinical manifestations, ranging from absence of symptoms to severe forms that need intensive care treatment. Here, plasma-EDTA samples of 30 patients compared with age- and sex-matched controls were analyzed via untargeted nuclear magnetic resonance (NMR)-based metabolomics and lipidomics. With the same approach, the effect of tocilizumab administration was evaluated in a subset of patients. Despite the heterogeneity of the clinical symptoms, COVID-19 patients are characterized by common plasma metabolomic and lipidomic signatures (91.7% and 87.5% accuracy, respectively, when compared to controls). Tocilizumab treatment resulted in at least partial reversion of the metabolic alterations due to SARS-CoV-2 infection. In conclusion, NMR-based metabolomic and lipidomic profiling provides novel insights into the pathophysiological mechanism of human response to SARS-CoV-2 infection and to monitor treatment outcomes. The current COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is markedly affecting the world population. Here we report about the small-molecule profile of patients hospitalized during the first wave of the COVID-19 pandemic in Florence (Italy). Using magnetic resonance spectroscopy, we showed that the infection induces profound changes in the metabolome. The analysis of the specific metabolite changes and correlations with clinical data enabled the identification of potential biochemical determinants of the disease fingerprint. We also followed how metabolic alterations revert towards those of the control group upon treatment with tocilizumab, a recombinant humanized monoclonal antibody against the interleukin-6 receptor. These results open up possibilities for the monitoring of novel patients and their individual response to treatment.
Collapse
|
139
|
Altered high-density lipoprotein composition and functions during severe COVID-19. Sci Rep 2021; 11:2291. [PMID: 33504824 PMCID: PMC7841145 DOI: 10.1038/s41598-021-81638-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) pandemic is affecting millions of patients worldwide. The consequences of initial exposure to SARS-CoV-2 go beyond pulmonary damage, with a particular impact on lipid metabolism. Decreased levels in HDL-C were reported in COVID-19 patients. Since HDL particles display antioxidant, anti-inflammatory and potential anti-infectious properties, we aimed at characterizing HDL proteome and functionality during COVID-19 relative to healthy subjects. HDLs were isolated from plasma of 8 severe COVID-19 patients sampled at admission to intensive care unit (Day 1, D1) at D3 and D7, and from 16 sex- and age-matched healthy subjects. Proteomic analysis was performed by LC-MS/MS. The relative amounts of proteins identified in HDLs were compared between COVID-19 and controls. apolipoprotein A-I and paraoxonase 1 were confirmed by Western-blot analysis to be less abundant in COVID-19 versus controls, whereas serum amyloid A and alpha-1 antitrypsin were higher. HDLs from patients were less protective in endothelial cells stiumalted by TNFα (permeability, VE-cadherin disorganization and apoptosis). In these conditions, HDL inhibition of apoptosis was blunted in COVID-19 relative to controls. In conclusion, we show major changes in HDL proteome and decreased functionality in severe COVID-19 patients.
Collapse
|
140
|
Lionetto L, Ulivieri M, Capi M, De Bernardini D, Fazio F, Petrucca A, Pomes LM, De Luca O, Gentile G, Casolla B, Curto M, Salerno G, Schillizzi S, Torre MS, Santino I, Rocco M, Marchetti P, Aceti A, Ricci A, Bonfini R, Nicoletti F, Simmaco M, Borro M. Increased kynurenine-to-tryptophan ratio in the serum of patients infected with SARS-CoV2: An observational cohort study. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166042. [PMID: 33338598 PMCID: PMC7834629 DOI: 10.1016/j.bbadis.2020.166042] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/01/2020] [Accepted: 12/06/2020] [Indexed: 12/16/2022]
Abstract
Immune dysregulation is a hallmark of patients infected by SARS-CoV2 and the balance between immune reactivity and tolerance is a key determinant of all stages of infection, including the excessive inflammatory state causing the acute respiratory distress syndrome. The kynurenine pathway (KP) of tryptophan (Trp) metabolism is activated by pro-inflammatory cytokines and drives mechanisms of immune tolerance. We examined the state of activation of the KP by measuring the Kyn:Trp ratio in the serum of healthy subjects (n = 239), and SARS-CoV2-negative (n = 305) and -positive patients (n = 89). Patients were recruited at the Emergency Room of St. Andrea Hospital (Rome, Italy). Kyn and Trp serum levels were assessed by HPLC/MS-MS. Compared to healthy controls, both SARS-CoV2-negative and -positive patients showed an increase in the Kyn:Trp ratio. The increase was larger in SARS-CoV2-positive patients, with a significant difference between SARS-CoV2-positive and -negative patients. In addition, the increase was more prominent in males, and positively correlated with age and severity of SARS-CoV2 infection, categorized as follows: 1 = no need for intensive care unit (ICU); 2 ≤ 3 weeks spent in ICU; 3 ≥ 3 weeks spent in ICU; and 4 = death. The highest Kyn:Trp values were found in SARS-CoV2-positive patients with severe lymphopenia. These findings suggest that the Kyn:Trp ratio reflects the level of inflammation associated with SARS-CoV2 infection, and, therefore, might represent a valuable biomarker for therapeutic intervention.
Collapse
Affiliation(s)
- Luana Lionetto
- Laboratory of Clinical Biochemistry, Mass Spectrometry Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Martina Ulivieri
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, New York, NY, USA
| | - Matilde Capi
- Laboratory of Clinical Biochemistry, Mass Spectrometry Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Donatella De Bernardini
- Laboratory of Clinical Biochemistry, Mass Spectrometry Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Francesco Fazio
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, New York, NY, USA
| | - Andrea Petrucca
- Microbiology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Leda Marina Pomes
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University, Rome, Italy
| | - Ottavia De Luca
- Laboratory of Clinical Biochemistry, Advanced Molecular Diagnostic Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Giovanna Gentile
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University, Rome, Italy; Laboratory of Clinical Biochemistry, Advanced Molecular Diagnostic Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Barbara Casolla
- University of Lille, Inserm U1172, CHU Lille, Department of Neurology, Stroke Unit, Lille, France
| | - Martina Curto
- Department of Mental Health, ASL, Rome 3, Rome, Italy; International Mood & Psychotic Disorders Research Consortium, Mailman Research Center, Belmon, MA, USA
| | - Gerardo Salerno
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University, Rome, Italy
| | | | - Maria Simona Torre
- Laboratory of Clinical Biochemistry, Advanced Molecular Diagnostic Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Iolanda Santino
- Microbiology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Monica Rocco
- Department of Surgical and Medical Science and Translational Medicine, Anesthesia and Intensive Care, Sapienza University, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Antonio Aceti
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University, Rome, Italy; Infectious disease Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Alberto Ricci
- Department of Surgical and Medical Science and Translational Medicine, Anesthesia and Intensive Care, Sapienza University, Rome, Italy; Division of Pneumology, Sant'Andrea University Hospital, Rome, Italy
| | - Rita Bonfini
- Emergency Department, Sant'Andrea University Hospital, Rome, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - Maurizio Simmaco
- Laboratory of Clinical Biochemistry, Mass Spectrometry Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University, Rome, Italy; Laboratory of Clinical Biochemistry, Advanced Molecular Diagnostic Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Marina Borro
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University, Rome, Italy; Laboratory of Clinical Biochemistry, Advanced Molecular Diagnostic Unit, Sant'Andrea University Hospital, Rome, Italy.
| |
Collapse
|
141
|
Affiliation(s)
- Suman S. Thakur
- Proteomics and Cell Signaling, Lab
W110, Centre for Cellular & Molecular
Biology, Habsiguda, Uppal
Road, Hyderabad 500 007, Telangana, India
| |
Collapse
|
142
|
Loo RL, Lodge S, Kimhofer T, Bong SH, Begum S, Whiley L, Gray N, Lindon JC, Nitschke P, Lawler NG, Schäfer H, Spraul M, Richards T, Nicholson JK, Holmes E. Quantitative In-Vitro Diagnostic NMR Spectroscopy for Lipoprotein and Metabolite Measurements in Plasma and Serum: Recommendations for Analytical Artifact Minimization with Special Reference to COVID-19/SARS-CoV-2 Samples. J Proteome Res 2020; 19:4428-4441. [PMID: 32852212 PMCID: PMC7640974 DOI: 10.1021/acs.jproteome.0c00537] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Indexed: 12/14/2022]
Abstract
Quantitative nuclear magnetic resonance (NMR) spectroscopy of blood plasma is widely used to investigate perturbed metabolic processes in human diseases. The reliability of biochemical data derived from these measurements is dependent on the quality of the sample collection and exact preparation and analysis protocols. Here, we describe systematically, the impact of variations in sample collection and preparation on information recovery from quantitative proton (1H) NMR spectroscopy of human blood plasma and serum. The effects of variation of blood collection tube sizes and preservatives, successive freeze-thaw cycles, sample storage at -80 °C, and short-term storage at 4 and 20 °C on the quantitative lipoprotein and metabolite patterns were investigated. Storage of plasma samples at 4 °C for up to 48 h, freezing at -80 °C and blood sample collection tube choice have few and minor effects on quantitative lipoprotein profiles, and even storage at 4 °C for up to 168 h caused little information loss. In contrast, the impact of heat-treatment (56 °C for 30 min), which has been used for inactivation of SARS-CoV-2 and other viruses, that may be required prior to analytical measurements in low level biosecurity facilities induced marked changes in both lipoprotein and low molecular weight metabolite profiles. It was conclusively demonstrated that this heat inactivation procedure degrades lipoproteins and changes metabolic information in complex ways. Plasma from control individuals and SARS-CoV-2 infected patients are differentially altered resulting in the creation of artifactual pseudo-biomarkers and destruction of real biomarkers to the extent that data from heat-treated samples are largely uninterpretable. We also present several simple blood sample handling recommendations for optimal NMR-based biomarker discovery investigations in SARS CoV-2 studies and general clinical biomarker research.
Collapse
Affiliation(s)
- Ruey Leng Loo
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
| | - Samantha Lodge
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
| | - Sze-How Bong
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Sofina Begum
- Section
for Nutrition Research, Imperial College
London, Sir Alexander Fleming Building, South Kensington, London SW72AZ, U.K.
| | - Luke Whiley
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
- Perron
Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Nicola Gray
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
| | - John C. Lindon
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
- Department
of Metabolism, Nutrition and Reproduction, Imperial College London, Sir Alexander Fleming Building, London SW72AZ, U.K.
| | - Philipp Nitschke
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
| | - Nathan G. Lawler
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
| | | | - Manfred Spraul
- Biospin
GmbH, Silberstreifen, 76287 Rheinstetten, Germany
| | - Toby Richards
- Division
of Surgery, Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Harry Perkins Building, Robert Warren Drive, Murdoch, Perth, WA 6150, Australia
- Department
of Endocrinology and Diabetes, Fiona Stanley
Hospital, Harry Perkins
Building, Murdoch, Perth, WA 6150, Australia
| | - Jeremy K. Nicholson
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
- Division
of Surgery, Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Harry Perkins Building, Robert Warren Drive, Murdoch, Perth, WA 6150, Australia
- Institute
of Global Health Innovation, Imperial College
London, Level 1, Faculty Building, South Kensington Campus, London SW72NA, U.K.
| | - Elaine Holmes
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, Perth, WA 6150, Australia
- Section
for Nutrition Research, Imperial College
London, Sir Alexander Fleming Building, South Kensington, London SW72AZ, U.K.
| |
Collapse
|