1
|
Dutta M, Dolan KA, Amiar S, Bass EJ, Sultana R, Voth GA, Brohawn SG, Stahelin RV. Direct lipid interactions control SARS-CoV-2 M protein conformational dynamics and virus assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.620124. [PMID: 39574576 PMCID: PMC11580925 DOI: 10.1101/2024.11.04.620124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
M is the most abundant structural membrane protein in coronaviruses and is essential for the formation of infectious virus particles. SARS-CoV-2 M adopts two conformations, Mshort and Mlong, and regulated transition between states is hypothesized to coordinate viral assembly and budding. However, the factors that regulate M conformation and roles for each state are unknown. Here, we discover a direct M-sphingolipid interaction that controls M conformational dynamics and virus assembly. We show M binds Golgi-enriched anionic lipids including ceramide-1-phosphate (C1P). Molecular dynamics simulations show C1P interaction promotes a long to short transition and energetically stabilizes Mshort. Cryo-EM structures show C1P specifically binds Mshort at a conserved site bridging transmembrane and cytoplasmic regions. Disrupting Mshort-C1P interaction alters M subcellular localization, reduces interaction with Spike and E, and impairs subsequent virus-like particle cell entry. Together, these results show endogenous signaling lipids regulate M structure and support a model in which Mshort is stabilized in the early endomembrane system to organize other structural proteins prior to viral budding.
Collapse
Affiliation(s)
- Mandira Dutta
- Department of Chemistry, The University of Chicago, Chicago, IL 60637
| | - Kimberly A. Dolan
- Department of Molecular & Cell Biology, Department of Neuroscience, California Institute for Quantitative Biology (QB3), Biophysics Graduate Program, University of California Berkeley, Berkeley, California 94720, USA
| | - Souad Amiar
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana 47907
| | - Elijah J. Bass
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana 47907
| | - Rokaia Sultana
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana 47907
| | - Gregory A. Voth
- Department of Chemistry, The University of Chicago, Chicago, IL 60637
- Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics, and James Franck Institute, The University of Chicago, Chicago, IL 60637
| | - Stephen G. Brohawn
- Department of Molecular & Cell Biology, Department of Neuroscience, California Institute for Quantitative Biology (QB3), Biophysics Graduate Program, University of California Berkeley, Berkeley, California 94720, USA
| | - Robert V. Stahelin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
2
|
Ibáñez-Prada ED, Guerrero JL, Bustos IG, León L, Fuentes YV, Santamaría-Torres M, Restrepo-Martínez JM, Serrano-Mayorga CC, Mendez L, Gomez-Duque S, Santacruz CA, Conway-Morris A, Martín-Loeches I, Gonzalez-Juarbe N, Cala MP, Reyes LF. The unique metabolic and lipid profiles of patients with severe COVID-19 compared to severe community-acquired pneumonia: a potential prognostic and therapeutic target. Expert Rev Respir Med 2024; 18:815-829. [PMID: 39327745 DOI: 10.1080/17476348.2024.2409264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Compare the changes and differences in metabolome and lipidome profiles among severe COVID-19 and CAP patients with ARF to identify biomarkers that could be used for personalized diagnosis, prognosis, and treatment. RESEARCH DESIGN AND METHODS Plasma samples were taken at hospital admission (baseline) and on the 5th day of hospitalization (follow-up) and examined by RP-LC-QTOF-MS and HILIC-LC-QTOF-MS. RESULTS 127 patients, 17 with CAP and 110 with COVID-19, were included. The analysis revealed 87 altered metabolites, suggesting changes in the metabolism of arachidonic acid, glycerolipids, glycerophospholipids, linoleic acid, pyruvate, glycolysis, among others. Most of these metabolites are involved in inflammatory, hypoxic, and thrombotic processes. At baseline, the greatest differences were found in phosphatidylcholine (PC) 31:4 (p < 0.001), phosphoserine (PS) 34:3 (p < 0.001), and phosphatidylcholine (PC) 36:5 (p < 0.001), all of which were notably decreased in COVID-19 patients. At follow-up, the most dysregulated metabolites were monomethyl-phosphatidylethanolamine (PE-Nme) 40:5 (p < 0.001) and phosphatidylcholine (PC) 38:4 (p < 0.001). CONCLUSIONS Metabolic and lipidic alterations suggest inhibition of innate anti-inflammatory and anti-thrombotic mechanisms in COVID-19 patients, which might lead to increased viral proliferation, uncontrolled inflammation, and thrombi formation. Results provide novel targets for predictive biomarkers against CAP and COVID-19. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Elsa D Ibáñez-Prada
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
- Clínica Universidad de La Sabana Chía, Colombia
| | - Jose L Guerrero
- MetCore-Metabolomics Core Facility, Vice-Presidency of Research and Knowledge Creation, Universidad de Los Andes, Bogotá, Colombia
| | - Ingrid G Bustos
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
| | - Lizeth León
- MetCore-Metabolomics Core Facility, Vice-Presidency of Research and Knowledge Creation, Universidad de Los Andes, Bogotá, Colombia
| | - Yuli V Fuentes
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
| | - Mary Santamaría-Torres
- MetCore-Metabolomics Core Facility, Vice-Presidency of Research and Knowledge Creation, Universidad de Los Andes, Bogotá, Colombia
| | | | | | - Lina Mendez
- Clínica Universidad de La Sabana Chía, Colombia
| | - Salome Gomez-Duque
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
| | - Carlos A Santacruz
- Fundación Santa Fe de Bogotá, Bogotá, Colombia
- Critical Care Department, Instituto de Ensino e Pesquisa do Pará, Brasil - IEPPA, Brazil
| | - Andrew Conway-Morris
- Division of Anesthesia, Department of Medicine, University of Cambridge, Cambridge, UK
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ignacio Martín-Loeches
- Department of Clinical Medicine, St James's Hospital, Multidisciplinary Intensive Care Research Organization (MICRO), Dublin, Ireland
| | | | - Mónica P Cala
- MetCore-Metabolomics Core Facility, Vice-Presidency of Research and Knowledge Creation, Universidad de Los Andes, Bogotá, Colombia
| | - Luis Felipe Reyes
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
- Clínica Universidad de La Sabana Chía, Colombia
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Neuenschwander FC, Barnett-Griness O, Piconi S, Maor Y, Sprinz E, Assy N, Khmelnitskiy O, Lomakin NV, Goloshchekin BM, Nahorecka E, Joaquim Westheimer Calvacante A, Ivanova A, Vladimirovich Zhuravel S, Yurevna Trufanova G, Bonora S, Saffoury A, Mayo A, Shvarts YG, Rizzardini G, Sobroza de Mello R, Pilau J, Klinov A, Valente-Acosta B, Olegovich Burlaka O, Bakhtina N, Bar-Meir M, Nikolaevich Shishimorov I, Oñate-Gutierrez J, García Rincón CI, Ivanovna Martynenko T, Hajjar LA, Carolina Nazare de Mendonca Procopio A, Simon K, Gabriel Chaves Santiago W, Fronczak A, Roberto Hoffmann Filho C, Hussein O, Aleksandrovich Martynov V, Chichino G, Blewaska P, Wroblewski J, Saul Irizar Santana S, Felipe Ocampo Agudelo A, Barczyk A, Lask Gerlach R, Campbell E, Bibliowicz A, Fathi R, Anderson P, Raday G, Klein M, Fehrmann C, Eagle G, Ben-Yair VK, Levitt ML. Effect of Opaganib on Supplemental Oxygen and Mortality in Patients with Severe SARS-CoV-2 Based upon FIO 2 Requirements. Microorganisms 2024; 12:1767. [PMID: 39338442 PMCID: PMC11434591 DOI: 10.3390/microorganisms12091767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/22/2024] [Accepted: 08/12/2024] [Indexed: 09/30/2024] Open
Abstract
Once a patient has been diagnosed with severe COVID-19 pneumonia, treatment options have limited effectiveness. Opaganib is an oral treatment under investigation being evaluated for treatment of hospitalized patients with severe COVID-19 pneumonia. A randomized, placebo-controlled, double-blind phase 2/3 trial was conducted in 57 sites worldwide from August 2020 to July 2021. Patients received either opaganib (n = 230; 500 mg twice daily) or matching placebo (n = 233) for 14 days. The primary outcome was the proportion of patients no longer requiring supplemental oxygen by day 14. Secondary outcomes included changes in the World Health Organization Ordinal Scale for Clinical Improvement, viral clearance, intubation, and mortality at 28 and 42 days. Pre-specified primary and secondary outcome analyses did not demonstrate statistically significant benefit (except nominally for time to viral clearance). Post-hoc analysis revealed the fraction of inspired oxygen (FIO2) at baseline was prognostic for opaganib treatment responsiveness and corresponded to disease severity markers. Patients with FIO2 levels at or below the median value (≤60%) had better outcomes after opaganib treatment (n = 117) compared to placebo (n = 134). The proportion of patients with ≤60% FIO2 at baseline that no longer required supplemental oxygen (≥24 h) by day 14 of opaganib treatment increased (76.9% vs. 63.4%; nominal p-value = 0.033). There was a 62.6% reduction in intubation/mechanical ventilation (6.84% vs. 17.91%; nominal p-value = 0.012) and a clinically meaningful 62% reduction in mortality (5.98% vs. 16.7%; nominal p-value = 0.019) by day 42. No new safety concerns were observed. While the primary analyses were not statistically significant, post-hoc analysis suggests opaganib benefit for patients with severe COVID-19 requiring supplemental oxygen with an FIO2 of ≤60%. Further studies are warranted to prospectively confirm opaganib benefit in this subpopulation.
Collapse
Affiliation(s)
| | | | | | - Yasmin Maor
- Infectious Disease Unit, E. Wolfson Medical Center, Holon 58100, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv 6997801, Israel
| | - Eduardo Sprinz
- Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil
| | - Nimer Assy
- Western Galilee Medical Center, Nahariya 221001, Israel
| | - Oleg Khmelnitskiy
- Saint-Petersburg State Budget Healthcare Institution "City Pokrovskaya Hospital", Saint-Petersburg 199106, Russia
| | - Nikita V Lomakin
- Russian Medical Academy of Continuous Professional Education of the Ministry of Health of the Russian Federation, Moscow 121359, Russia
| | | | | | | | - Anastasia Ivanova
- State Budgetary Institution of Ryazan Region "Regional Clinical Hospital", Ryazan 390026, Russia
| | | | - Galina Yurevna Trufanova
- State Budgetary Healthcare Institution of the Tver Region "Regional Clinical Hospital", Tver 170100, Russia
| | | | | | - Ami Mayo
- Assuta Medical Center Ashdod, Ashdod 7747629, Israel
| | - Yury G Shvarts
- Clinical Hospital n.a-S.R. Mirotvortseva SSMU, Saratov 410012, Russia
| | | | - Rogerio Sobroza de Mello
- Hospital Nossa Senhora da Conceição de Tubarão, Centro de Pesquisas Clínicas do Hospital Nossa Senhora da Conceição, Tubarão 88701-160, Brazil
| | - Janaina Pilau
- Hospital de Clínicas de Passo Fundo, Cento de Pesquisa Clínica, Passo Fundo 99010-260, Brazil
| | - Alexey Klinov
- Kirovsk Interregional Hospital, Leningrad 187342, Russia
| | | | - Oleg Olegovich Burlaka
- Saint-Petersburg State Budgetary Healthcare Institution "City Aleksandrovskaya Hospital", Saint-Petersburg 193312, Russia
| | - Natalia Bakhtina
- State Regional Budgetary Healthcare Institution Murmansk Regional Clinical Hospital, Murmansk 183032, Russia
| | | | - Ivan Nikolaevich Shishimorov
- Department of Pediatrics and Neonatology, Institute of Medical and Physiotherapy, Federal State Budgetary Educational Institution of Higher Education Volgograd State Medical University, Volgograd 400087, Russia
| | | | | | | | - Ludhmila Abrahão Hajjar
- Instituto do Coração do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, Andar 05403-900, Brazil
| | | | - Krzysztof Simon
- Szpital Specjalistyczny im. Gromkowskiego, 51-149 Wrocław, Poland
| | | | - Adam Fronczak
- Centrum Onkologii w Lodzi, Oddzial COVID-19, 93-510 Lodz, Poland
| | | | | | | | | | | | - Jacek Wroblewski
- Szpital Wojewódzki im. Mikołaja Kopernika, 75-581 Koszalin, Poland
| | | | | | - Adam Barczyk
- Department of Pneumology, School of Medicine in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | | | | | | | - Reza Fathi
- RedHill Biopharma Ltd., Tel-Aviv 6473921, Israel
| | | | - Gilead Raday
- RedHill Biopharma Ltd., Tel-Aviv 6473921, Israel
| | | | - Clara Fehrmann
- CEEF Solutions Beaconsfield, Pointe-Claire, QC H9S 4L7, Canada
| | - Gina Eagle
- G.E.T. Pharma Consulting, LLC, Lumberville, PA 18933, USA
| | | | | |
Collapse
|
4
|
Dai J, Feng Y, Liao Y, Tan L, Sun Y, Song C, Qiu X, Ding C. Virus infection and sphingolipid metabolism. Antiviral Res 2024; 228:105942. [PMID: 38908521 DOI: 10.1016/j.antiviral.2024.105942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Cellular sphingolipids have vital roles in human virus replication and spread as they are exploited by viruses for cell entry, membrane fusion, genome replication, assembly, budding, and propagation. Intracellular sphingolipid biosynthesis triggers conformational changes in viral receptors and facilitates endosomal escape. However, our current understanding of how sphingolipids precisely regulate viral replication is limited, and further research is required to comprehensively understand the relationships between viral replication and endogenous sphingolipid species. Emerging evidence now suggests that targeting and manipulating sphingolipid metabolism enzymes in host cells is a promising strategy to effectively combat viral infections. Additionally, serum sphingolipid species and concentrations could function as potential serum biomarkers to help monitor viral infection status in different patients. In this work, we comprehensively review the literature to clarify how viruses exploit host sphingolipid metabolism to accommodate viral replication and disrupt host innate immune responses. We also provide valuable insights on the development and use of antiviral drugs in this area.
Collapse
Affiliation(s)
- Jun Dai
- Experimental Animal Center, Zunyi Medical University, Zunyi, 563099, China
| | - Yiyi Feng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; Laboratory of Veterinary Microbiology and Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning, 530004, Guangxi China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Lei Tan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yingjie Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Cuiping Song
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xusheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
5
|
Uranbileg B, Isago H, Nakayama H, Jubishi D, Okamoto K, Sakai E, Kubota M, Tsutsumi T, Moriya K, Kurano M. Comprehensive metabolic modulations of sphingolipids are promising severity indicators in COVID-19. FASEB J 2024; 38:e23827. [PMID: 39012295 DOI: 10.1096/fj.202401099r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has had a significant worldwide impact, affecting millions of people. COVID-19 is characterized by a heterogenous clinical phenotype, potentially involving hyperinflammation and prolonged tissue damage, although the exact underlying mechanisms are yet to be fully understood. Sphingolipid metabolites, which govern cell survival and proliferation, have emerged as key players in inflammatory signaling and cytokine responses. Given the complex metabolic pathway of sphingolipids, this study aimed to understand their potential role in the pathogenesis of COVID-19. We conducted a comprehensive examination of sphingolipid modulations across groups classified based on disease severity, incorporating a time-course in serum and urine samples. Several sphingolipids, including sphingosine, lactosylceramide, and hexosylceramide, emerged as promising indicators of COVID-19 severity, as validated by correlation analyses conducted on both serum and urine samples. Other sphingolipids, such as sphingosine 1-phosphate, ceramides, and deoxy-dihydroceramides, decreased in both COVID-19 patients and individuals with non-COVID infectious diseases. This suggests that these sphingolipids are not specifically associated with COVID-19 but rather with pathological conditions caused by infectious diseases. Our analysis of urine samples revealed elevated levels of various sphingolipids, with changes dependent on disease severity, potentially highlighting the acute kidney injury associated with COVID-19. This study illuminates the intricate relationship between disturbed sphingolipid metabolism, COVID-19 severity, and clinical factors. These findings provide valuable insights into the broader landscape of inflammatory diseases.
Collapse
Affiliation(s)
- Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideaki Isago
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hitoshi Nakayama
- Laboratory of Biochemistry, Faculty of Health Care and Nursing, Juntendo University, Chiba, Japan
- Institute for Environmental and Gender-specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan
| | - Daisuke Jubishi
- Department of Infectious Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Koh Okamoto
- Department of Infectious Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eri Sakai
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Nihon Waters K.K., Tokyo, Japan
| | | | - Takeya Tsutsumi
- Department of Infectious Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kyoji Moriya
- Department of Infectious Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Loveday EK, Welhaven H, Erdogan AE, Hain K, Chang CB, June RK, Taylor MP. Starve a cold or feed a fever? Identifying cellular metabolic changes following infection and exposure to SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595410. [PMID: 38826440 PMCID: PMC11142155 DOI: 10.1101/2024.05.22.595410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Viral infections induce major shifts in cellular metabolism elicited by active viral replication and antiviral responses. For the virus, harnessing cellular metabolism and evading changes that limit replication are essential for productive viral replication. In contrast, the cellular response to infection disrupts metabolic pathways to prevent viral replication and promote an antiviral state in the host cell and neighboring bystander cells. This competition between the virus and cell results in measurable shifts in cellular metabolism that differ depending on the virus, cell type, and extracellular environment. The resulting metabolic shifts can be observed and analyzed using global metabolic profiling techniques to identify pathways that are critical for either viral replication or cellular defense. SARS-CoV-2 is a respiratory virus that can exhibit broad tissue tropism and diverse, yet inconsistent, symptomatology. While the factors that determine the presentation and severity of SARS-CoV-2 infection remain unclear, metabolic syndromes are associated with more severe manifestations of SARS-CoV-2 disease. Despite these observations a critical knowledge gap remains between cellular metabolic responses and SARS-CoV-2 infection. Using a well-established untargeted metabolomics analysis workflow, we compared SARS-CoV-2 infection of human lung carcinoma cells. We identified significant changes in metabolic pathways that correlate with either productive or non-productive viral infection. This information is critical for characterizing the factors that contribute to SARS-CoV-2 replication that could be targeted for therapeutic interventions to limit viral disease.
Collapse
Affiliation(s)
- Emma Kate Loveday
- Center for Biofilm Engineering, Montana State University, Bozeman MT 59717
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
| | - Hope Welhaven
- Chemistry and Biochemistry, Montana State University, Bozeman MT 59717
| | - Ayten Ebru Erdogan
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
| | - Kyle Hain
- Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| | - Connie B. Chang
- Center for Biofilm Engineering, Montana State University, Bozeman MT 59717
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman MT 59717
| | - Matthew P. Taylor
- Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| |
Collapse
|
7
|
Li C, Wu K, Yang R, Liao M, Li J, Zhu Q, Zhang J, Zhang X. Comprehensive analysis of immunogenic cell death-related gene and construction of prediction model based on WGCNA and multiple machine learning in severe COVID-19. Sci Rep 2024; 14:8450. [PMID: 38600309 PMCID: PMC11006847 DOI: 10.1038/s41598-024-59117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/08/2024] [Indexed: 04/12/2024] Open
Abstract
The death of coronavirus disease 2019 (COVID-19) is primarily due to from critically ill patients, especially from ARDS complications caused by SARS-CoV-2. Therefore, it is essential to contribute an in-depth understanding of the pathogenesis of the disease and to identify biomarkers for predicting critically ill patients at the molecular level. Immunogenic cell death (ICD), as a specific variant of regulatory cell death driven by stress, can induce adaptive immune responses against cell death antigens in the host. Studies have confirmed that both innate and adaptive immune pathways are involved in the pathogenesis of SARS-CoV-2 infection. However, the role of ICD in the pathogenesis of severe COVID-19 has rarely been explored. In this study, we systematically evaluated the role of ICD-related genes in COVID-19. We conducted consensus clustering, immune infiltration analysis, and functional enrichment analysis based on ICD differentially expressed genes. The results showed that immune infiltration characteristics were altered in severe and non-severe COVID-19. In addition, we used multiple machine learning methods to screen for five risk genes (KLF5, NSUN7, APH1B, GRB10 and CD4), which are used to predict COVID-19 severity. Finally, we constructed a nomogram to predict the risk of severe COVID-19 based on the classification and recognition model, and validated the model with external data sets. This study provides a valuable direction for the exploration of the pathogenesis and progress of COVID-19, and helps in the early identification of severe cases of COVID-19 to reduce mortality.
Collapse
Affiliation(s)
- Chunyu Li
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Ke Wu
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Rui Yang
- Department of Internal Medicine, Guiyang First People's Hospital, Guiyang, 550004, Guizhou, China
| | - Minghua Liao
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jun Li
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Qian Zhu
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jiayi Zhang
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Xianming Zhang
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
8
|
Bernardo RA, Roque JV, de Oliveira Júnior CI, Lima NM, Machado LS, Duarte GRM, Costa NL, Sorgi CA, Soares FFL, Vaz BG, Chaves AR. Exploring salivary lipid profile changes in COVID-19 patients: Insights from mass spectrometry analysis. Talanta 2024; 269:125522. [PMID: 38091738 DOI: 10.1016/j.talanta.2023.125522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
The most common COVID-19 testing relies on the use of nasopharyngeal swabs. However, this sampling step is very uncomfortable and is one of the biggest challenges regarding population testing. In the present study, the use of saliva as an alternative sample for COVID-19 diagnosis was investigated. Therefore, high-resolution mass spectrometry analysis and chemometric approaches were applied to salivary lipid extracts. Two data organizations were used: classical MS data and pseudo-MS image datasets. The latter transformed MS data into pseudo-images, simplifying data interpretation. Classification models achieved high accuracy, with pseudo-MS image data performing exceptionally well. PLS-DA with OPSDA successfully separated COVID-19 and healthy groups, serving as a potential diagnostic tool. The most important lipids for COVID-19 classification were elucidated and include sphingolipids, ceramides, phospholipids, and glycerolipids. These lipids play a crucial role in viral replication and the inflammatory response. While pseudo-MS image data excelled in classification, it lacked the ability to annotate important variables, which was performed using classical MS data. These findings have the potential to improve clinical diagnosis using rapid, non-invasive testing methods and accurate high-volume results.
Collapse
Affiliation(s)
- Ricardo A Bernardo
- Instituto de Química, Universidade Federal de Goiás, 74690-900, Goiânia, GO, Brazil; Departamento de Química, Universidade Federal do Paraná, 81531-980, Curitiba, PR, Brazil.
| | - Jussara V Roque
- Instituto de Química, Universidade Federal de Goiás, 74690-900, Goiânia, GO, Brazil
| | - Charles I de Oliveira Júnior
- Instituto de Química, Universidade Federal de Goiás, 74690-900, Goiânia, GO, Brazil; Departamento de Química, Universidade Federal de Jataí, 75804-020, Jataí, GO, Brazil
| | | | - Lucas Santos Machado
- Instituto de Química, Universidade Federal de Goiás, 74690-900, Goiânia, GO, Brazil
| | | | - Nádia L Costa
- Faculdade de Odontologia, Universidade Federal de Goiás, 74605-020, Goiânia, GO, Brazil
| | - Carlos A Sorgi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14015-130, Ribeirão Preto, SP, Brazil
| | - Frederico F L Soares
- Departamento de Química, Universidade Federal do Paraná, 81531-980, Curitiba, PR, Brazil
| | - Boniek G Vaz
- Instituto de Química, Universidade Federal de Goiás, 74690-900, Goiânia, GO, Brazil
| | - Andréa R Chaves
- Instituto de Química, Universidade Federal de Goiás, 74690-900, Goiânia, GO, Brazil; Departamento de Química, Universidade Federal de Jataí, 75804-020, Jataí, GO, Brazil.
| |
Collapse
|
9
|
D’Avila H, Lima CNR, Rampinelli PG, Mateus LCO, de Sousa Silva RV, Correa JR, de Almeida PE. Lipid Metabolism Modulation during SARS-CoV-2 Infection: A Spotlight on Extracellular Vesicles and Therapeutic Prospects. Int J Mol Sci 2024; 25:640. [PMID: 38203811 PMCID: PMC10778989 DOI: 10.3390/ijms25010640] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) have a significant impact on the pathophysiological processes associated with various diseases such as tumors, inflammation, and infection. They exhibit molecular, biochemical, and entry control characteristics similar to viral infections. Viruses, on the other hand, depend on host metabolic machineries to fulfill their biosynthetic requirements. Due to potential advantages such as biocompatibility, biodegradation, and efficient immune activation, EVs have emerged as potential therapeutic targets against the SARS-CoV-2 infection. Studies on COVID-19 patients have shown that they frequently have dysregulated lipid profiles, which are associated with an increased risk of severe repercussions. Lipid droplets (LDs) serve as organelles with significant roles in lipid metabolism and energy homeostasis as well as having a wide range of functions in infections. The down-modulation of lipids, such as sphingolipid ceramide and eicosanoids, or of the transcriptional factors involved in lipogenesis seem to inhibit the viral multiplication, suggesting their involvement in the virus replication and pathogenesis as well as highlighting their potential as targets for drug development. Hence, this review focuses on the role of modulation of lipid metabolism and EVs in the mechanism of immune system evasion during SARS-CoV-2 infection and explores the therapeutic potential of EVs as well as application for delivering therapeutic substances to mitigate viral infections.
Collapse
Affiliation(s)
- Heloisa D’Avila
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | | | - Pollianne Garbero Rampinelli
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - Laiza Camila Oliveira Mateus
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - Renata Vieira de Sousa Silva
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - José Raimundo Correa
- Laboratory of Microscopy and Microanalysis, University of Brasília, Brasília 70910-900, Brazil;
| | - Patrícia Elaine de Almeida
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| |
Collapse
|
10
|
Hou J, Wei Y, Zou J, Jaffery R, Sun L, Liang S, Zheng N, Guerrero AM, Egan NA, Bohat R, Chen S, Zheng C, Mao X, Yi SS, Chen K, McGrail DJ, Sahni N, Shi PY, Chen Y, Xie X, Peng W. Integrated multi-omics analyses identify anti-viral host factors and pathways controlling SARS-CoV-2 infection. Nat Commun 2024; 15:109. [PMID: 38168026 PMCID: PMC10761986 DOI: 10.1038/s41467-023-44175-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Host anti-viral factors are essential for controlling SARS-CoV-2 infection but remain largely unknown due to the biases of previous large-scale studies toward pro-viral host factors. To fill in this knowledge gap, we perform a genome-wide CRISPR dropout screen and integrate analyses of the multi-omics data of the CRISPR screen, genome-wide association studies, single-cell RNA-Seq, and host-virus proteins or protein/RNA interactome. This study uncovers many host factors that are currently underappreciated, including the components of V-ATPases, ESCRT, and N-glycosylation pathways that modulate viral entry and/or replication. The cohesin complex is also identified as an anti-viral pathway, suggesting an important role of three-dimensional chromatin organization in mediating host-viral interaction. Furthermore, we discover another anti-viral regulator KLF5, a transcriptional factor involved in sphingolipid metabolism, which is up-regulated, and harbors genetic variations linked to COVID-19 patients with severe symptoms. Anti-viral effects of three identified candidates (DAZAP2/VTA1/KLF5) are confirmed individually. Molecular characterization of DAZAP2/VTA1/KLF5-knockout cells highlights the involvement of genes related to the coagulation system in determining the severity of COVID-19. Together, our results provide further resources for understanding the host anti-viral network during SARS-CoV-2 infection and may help develop new countermeasure strategies.
Collapse
Affiliation(s)
- Jiakai Hou
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Yanjun Wei
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Zou
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Roshni Jaffery
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Long Sun
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Shaoheng Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Computer Science, Rice University, Houston, TX, USA
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ningbo Zheng
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Ashley M Guerrero
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Nicholas A Egan
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Ritu Bohat
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Si Chen
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Caishang Zheng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - S Stephen Yi
- Department of Oncology, Livestrong Cancer Institutes, and Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Interdisciplinary Life Sciences Graduate Programs (ILSGP) and Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel J McGrail
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Nidhi Sahni
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences, The University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Center for Structural Biology & Molecular Biophysics, The University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Translational Science, The University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Drug Discovery, The University of Texas Medical Branch, Galveston, TX, USA.
| | - Yiwen Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Quantitative Sciences Program, MD Anderson Cancer Center, UT Health Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Xuping Xie
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Drug Discovery, The University of Texas Medical Branch, Galveston, TX, USA.
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| |
Collapse
|
11
|
Chen S, Liang J, Chen D, Huang Q, Sun K, Zhong Y, Lin B, Kong J, Sun J, Gong C, Wang J, Gao Y, Zhang Q, Sun H. Cerebrospinal fluid metabolomic and proteomic characterization of neurologic post-acute sequelae of SARS-CoV-2 infection. Brain Behav Immun 2024; 115:209-222. [PMID: 37858739 DOI: 10.1016/j.bbi.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/08/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023] Open
Abstract
The mechanism by which SARS-CoV-2 causes neurological post-acute sequelae of SARS-CoV-2 (neuro-PASC) remains unclear. Herein, we conducted proteomic and metabolomic analyses of cerebrospinal fluid (CSF) samples from 21 neuro-PASC patients, 45 healthy volunteers, and 26 inflammatory neurological diseases patients. Our data showed 69 differentially expressed metabolites and six differentially expressed proteins between neuro-PASC patients and healthy individuals. Elevated sphinganine and ST1A1, sphingolipid metabolism disorder, and attenuated inflammatory responses may contribute to the occurrence of neuro-PASC, whereas decreased levels of 7,8-dihydropterin and activation of steroid hormone biosynthesis may play a role in the repair process. Additionally, a biomarker cohort consisting of sphinganine, 7,8-dihydroneopterin, and ST1A1 was preliminarily demonstrated to have high value in diagnosing neuro-PASC. In summary, our study represents the first attempt to integrate the diagnostic benefits of CSF with the methodological advantages of multi-omics, thereby offering valuable insights into the pathogenesis of neuro-PASC and facilitating the work of neuroscientists in disclosing different neurological dimensions associated with COVID-19.
Collapse
Affiliation(s)
- Shilan Chen
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jianhao Liang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Dingqiang Chen
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qiyuan Huang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Kaijian Sun
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuxia Zhong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Baojia Lin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jingjing Kong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jiaduo Sun
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Chengfang Gong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jun Wang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ya Gao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Qingguo Zhang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Goracci L, Petito E, Di Veroli A, Falcinelli E, Bencivenga C, Giglio E, Becattini C, De Robertis E, Vaudo G, Gresele P. A platelet lipidomics signature in patients with COVID-19. Platelets 2023; 34:2200847. [PMID: 37114418 DOI: 10.1080/09537104.2023.2200847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Ischemic cardiovascular and venous thromboembolic events are a frequent cause of death in severe COVID-19 patients. Platelet activation plays a key role in these complications, however platelet lipidomics have not been studied yet. The aim of our pilot investigation was to perform a preliminary study of platelet lipidomics in COVID-19 patients compared to healthy subjects. Lipid extraction and identification of ultrapurified platelets from eight hospitalized COVID-19 patients and eight age- and sex-matched healthy controls showed a lipidomic pattern almost completely separating COVID-19 patients from healthy controls. In particular, a significant decrease of ether phospholipids and increased levels of ganglioside GM3 were observed in platelets from COVID-19 patients. In conclusion, our study shows for the first time that platelets from COVID-19 patients display a different lipidomics signature distinguishing them from healthy controls, and suggests that altered platelet lipid metabolism may play a role in viral spreading and in the thrombotic complications of COVID-19.
Collapse
Affiliation(s)
- Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Eleonora Petito
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Alessandra Di Veroli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Emanuela Falcinelli
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Caterina Bencivenga
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Elisa Giglio
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Cecilia Becattini
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Edoardo De Robertis
- Division of Anaesthesia, Analgesia, and Intensive Care, University of Perugia, Perugia, Italy
| | - Gaetano Vaudo
- Unit of Internal Medicine, Terni University Hospital, Terni, Italy
| | - Paolo Gresele
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
13
|
Cesar-Silva D, Pereira-Dutra FS, Giannini ALM, Maya-Monteiro CM, de Almeida CJG. Lipid compartments and lipid metabolism as therapeutic targets against coronavirus. Front Immunol 2023; 14:1268854. [PMID: 38106410 PMCID: PMC10722172 DOI: 10.3389/fimmu.2023.1268854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/24/2023] [Indexed: 12/19/2023] Open
Abstract
Lipids perform a series of cellular functions, establishing cell and organelles' boundaries, organizing signaling platforms, and creating compartments where specific reactions occur. Moreover, lipids store energy and act as secondary messengers whose distribution is tightly regulated. Disruption of lipid metabolism is associated with many diseases, including those caused by viruses. In this scenario, lipids can favor virus replication and are not solely used as pathogens' energy source. In contrast, cells can counteract viruses using lipids as weapons. In this review, we discuss the available data on how coronaviruses profit from cellular lipid compartments and why targeting lipid metabolism may be a powerful strategy to fight these cellular parasites. We also provide a formidable collection of data on the pharmacological approaches targeting lipid metabolism to impair and treat coronavirus infection.
Collapse
Affiliation(s)
- Daniella Cesar-Silva
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Filipe S. Pereira-Dutra
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ana Lucia Moraes Giannini
- Laboratory of Functional Genomics and Signal Transduction, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarissa M. Maya-Monteiro
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Laboratory of Endocrinology and Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Cecília Jacques G. de Almeida
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Wang B, Yang W, Tong Y, Sun M, Quan S, Zhu J, Zhang Q, Qin Z, Ni Y, Zhao Y, Wang K, Zhang C, Zhang Y, Wang Z, Song Z, Liu H, Fang H, Kong Z, Ding C, Guo W. Integrative proteomics and metabolomics study reveal enhanced immune responses by COVID-19 vaccine booster shot against Omicron SARS-CoV-2 infection. J Med Virol 2023; 95:e29219. [PMID: 37966997 DOI: 10.1002/jmv.29219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
Since its outbreak in late 2021, the Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been widely reported to be able to evade neutralizing antibodies, becoming more transmissible while causing milder symptoms than previous SARS-CoV-2 strains. Understanding the underlying molecular changes of Omicron SARS-CoV-2 infection and corresponding host responses are important to the control of Omicron COVID-19 pandemic. In this study, we report an integrative proteomics and metabolomics investigation of serum samples from 80 COVID-19 patients infected with Omicron SARS-CoV-2, as well as 160 control serum samples from 80 healthy individuals and 80 patients who had flu-like symptoms but were negative for SARS-CoV-2 infection. The multiomics results indicated that Omicron SARS-CoV-2 infection caused significant changes to host serum proteome and metabolome comparing to the healthy controls and patients who had flu-like symptoms without COVID-19. Protein and metabolite changes also pointed to liver dysfunctions and potential damage to other host organs by Omicron SARS-CoV-2 infection. The Omicron COVID-19 patients could be roughly divided into two subgroups based on their proteome differences. Interestingly, the subgroup who mostly had received full vaccination with booster shot had fewer coughing symptom, changed sphingomyelin lipid metabolism, and stronger immune responses including higher numbers of lymphocytes, monocytes, neutrophils, and upregulated proteins related to CD4+ T cells, CD8+ effector memory T cells (Tem), and conventional dendritic cells, revealing beneficial effects of full COVID-19 vaccination against Omicron SARS-CoV-2 infection through molecular changes.
Collapse
Affiliation(s)
- Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
- Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yexin Tong
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Mingjun Sun
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Sheng Quan
- Calibra Lab at DIAN Diagnostics, Hangzhou, Zhejiang, China
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jing Zhu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianwen Zhang
- Calibra Lab at DIAN Diagnostics, Hangzhou, Zhejiang, China
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhaoyu Qin
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yanxia Ni
- Calibra Lab at DIAN Diagnostics, Hangzhou, Zhejiang, China
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Ying Zhao
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kouqiong Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
- Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, China
| | - Yichi Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenxin Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenju Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
| | - Huafen Liu
- Calibra Lab at DIAN Diagnostics, Hangzhou, Zhejiang, China
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hao Fang
- Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai, China
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziqing Kong
- Calibra Lab at DIAN Diagnostics, Hangzhou, Zhejiang, China
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, Zhejiang, China
- Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
- Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Khan RJ, Single SL, Simmons CS, Athar M, Liu Y, Bodduluri S, Benson PV, Goliwas KF, Deshane JS. Altered sphingolipid pathway in SARS-CoV-2 infected human lung tissue. Front Immunol 2023; 14:1216278. [PMID: 37868972 PMCID: PMC10585362 DOI: 10.3389/fimmu.2023.1216278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction The SARS-CoV-2 mediated COVID-19 pandemic has impacted millions worldwide. Hyper-inflammatory processes, including cytokine storm, contribute to long-standing tissue injury and damage in COVID-19. The metabolism of sphingolipids as regulators of cell survival, differentiation, and proliferation has been implicated in inflammatory signaling and cytokine responses. Sphingosine-kinase-1 (SK1) and ceramide-synthase-2 (CERS2) generate metabolites that regulate the anti- and pro-apoptotic processes, respectively. Alterations in SK1 and CERS2 expression may contribute to the inflammation and tissue damage during COVID-19. The central objective of this study is to evaluate structural changes in the lung post-SARS-CoV-2 infection and to investigate whether the sphingolipid rheostat is altered in response to SARS-CoV-2 infection. Methods Central and peripheral lung tissues from COVID-19+ or control autopsies and resected lung tissue from COVID-19 convalescents were subjected to histologic evaluation of airspace and collagen deposisiton, and immunohistochemical evaluation of SK1 and CERS2. Results Here, we report significant reduction in air space and increase in collagen deposition in lung autopsy tissues from patients who died from COVID-19 (COVID-19+) and COVID-19 convalescent individuals. SK1 expression increased in the lungs of COVID-19+ autopsies and COVID-19 convalescent lung tissue compared to controls and was mostly associated with Type II pneumocytes and alveolar macrophages. No significant difference in CERS2 expression was noted. SARS-CoV-2 infection upregulates SK1 and increases the ratio of SK1 to CERS2 expression in lung tissues of COVID-19 autopsies and COVID-19 convalescents. Discussion These data suggest an alteration in the sphingolipid rheostat in lung tissue during COVID-19, suggesting a potential contribution to the inflammation and tissue damage associated with viral infection.
Collapse
Affiliation(s)
- Rabisa J. Khan
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, United States
| | - Sierra L. Single
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christopher S. Simmons
- University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, United States
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuelong Liu
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sandeep Bodduluri
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Paul V. Benson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kayla F. Goliwas
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jessy S. Deshane
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
16
|
Fenizia S, Gaggini M, Vassalle C. The Sphingolipid-Signaling Pathway as a Modulator of Infection by SARS-CoV-2. Curr Issues Mol Biol 2023; 45:7956-7973. [PMID: 37886946 PMCID: PMC10605018 DOI: 10.3390/cimb45100503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Ceramides and other related sphingolipids, important cellular components linked to metabolic homeostasis and cardiometabolic diseases, have been found to be involved in different steps of the SARS-CoV-2 life cycle. Hence, changes in their physiological levels are identified as predictors of COVID-19 severity and prognosis, as well as potential therapeutic targets. In this review, an overview of the SARS-CoV-2 life cycle is given, followed by a description of the sphingolipid metabolism and its role in viral infection, with a particular focus on those steps required to finalize the viral life cycle. Furthermore, the use and development of pharmaceutical strategies to target sphingolipids to prevent and treat severe and long-term symptoms of infectious diseases, particularly COVID-19, are reviewed herein. Finally, research perspectives and current challenges in this research field are highlighted. Although many aspects of sphingolipid metabolism are not fully known, this review aims to highlight how the discovery and use of molecules targeting sphingolipids with reliable and selective properties may offer new therapeutic alternatives to infectious and other diseases, including COVID-19.
Collapse
Affiliation(s)
- Simona Fenizia
- Istituto di Fisiologia Clinica, Italian National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Melania Gaggini
- Fondazione CNR-Regione Toscana G. Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
| | - Cristina Vassalle
- Fondazione CNR-Regione Toscana G. Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
17
|
Abdel-Megied AM, Monreal IA, Zhao L, Apffel A, Aguilar HC, Jones JW. Characterization of the cellular lipid composition during SARS-CoV-2 infection. Anal Bioanal Chem 2023; 415:5269-5279. [PMID: 37438564 PMCID: PMC10981079 DOI: 10.1007/s00216-023-04825-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/14/2023]
Abstract
Emerging and re-emerging zoonotic viral diseases continue to significantly impact public health. Of particular interest are enveloped viruses (e.g., SARS-CoV-2, the causative pathogen of COVID-19), which include emerging pathogens of highest concern. Enveloped viruses contain a viral envelope that encapsulates the genetic material and nucleocapsid, providing structural protection and functional bioactivity. The viral envelope is composed of a coordinated network of glycoproteins and lipids. The lipid composition of the envelope consists of lipids preferentially appropriated from host cell membranes. Subsequently, changes to the host cell lipid metabolism and an accounting of what lipids are changed during viral infection provide an opportunity to fingerprint the host cell's response to the infecting virus. To address this issue, we comprehensively characterized the lipid composition of VeroE6-TMPRSS2 cells infected with SARS-CoV-2. Our approach involved using an innovative solid-phase extraction technique to efficiently extract cellular lipids combined with liquid chromatography coupled to high-resolution tandem mass spectrometry. We identified lipid changes in cells exposed to SARS-CoV-2, of which the ceramide to sphingomyelin ratio was most prominent. The identification of a lipid profile (i.e., lipid fingerprint) that is characteristic of cellular SARS-CoV-2 infection lays the foundation for targeting lipid metabolism pathways to further understand how enveloped viruses infect cells, identifying opportunities to aid antiviral and vaccine development.
Collapse
Affiliation(s)
- Ahmed M Abdel-Megied
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Room N721, Baltimore, MD, 21201, USA
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Kafr El-Sheikh University, Kafr El-Sheikh City, Egypt
| | - Isaac A Monreal
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | | | | | - Hector C Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Room N721, Baltimore, MD, 21201, USA.
| |
Collapse
|
18
|
Toro DM, da Silva-Neto PV, de Carvalho JCS, Fuzo CA, Pérez MM, Pimentel VE, Fraga-Silva TFC, Oliveira CNS, Caruso GR, Vilela AFL, Nobre-Azevedo P, Defelippo-Felippe TV, Argolo JGM, Degiovani AM, Ostini FM, Feitosa MR, Parra RS, Vilar FC, Gaspar GG, da Rocha JJR, Feres O, Costa GP, Maruyama SRC, Russo EMS, Fernandes APM, Santos IKFM, Malheiro A, Sadikot RT, Bonato VLD, Cardoso CRB, Dias-Baruffi M, Trapé ÁA, Faccioli LH, Sorgi CA. Plasma Sphingomyelin Disturbances: Unveiling Its Dual Role as a Crucial Immunopathological Factor and a Severity Prognostic Biomarker in COVID-19. Cells 2023; 12:1938. [PMID: 37566018 PMCID: PMC10417089 DOI: 10.3390/cells12151938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 08/12/2023] Open
Abstract
SARS-CoV-2 infection triggers distinct patterns of disease development characterized by significant alterations in host regulatory responses. Severe cases exhibit profound lung inflammation and systemic repercussions. Remarkably, critically ill patients display a "lipid storm", influencing the inflammatory process and tissue damage. Sphingolipids (SLs) play pivotal roles in various cellular and tissue processes, including inflammation, metabolic disorders, and cancer. In this study, we employed high-resolution mass spectrometry to investigate SL metabolism in plasma samples obtained from control subjects (n = 55), COVID-19 patients (n = 204), and convalescent individuals (n = 77). These data were correlated with inflammatory parameters associated with the clinical severity of COVID-19. Additionally, we utilized RNAseq analysis to examine the gene expression of enzymes involved in the SL pathway. Our analysis revealed the presence of thirty-eight SL species from seven families in the plasma of study participants. The most profound alterations in the SL species profile were observed in patients with severe disease. Notably, a predominant sphingomyelin (SM d18:1) species emerged as a potential biomarker for COVID-19 severity, showing decreased levels in the plasma of convalescent individuals. Elevated SM levels were positively correlated with age, hospitalization duration, clinical score, and neutrophil count, as well as the production of IL-6 and IL-8. Intriguingly, we identified a putative protective effect against disease severity mediated by SM (d18:1/24:0), while ceramide (Cer) species (d18:1/24:1) and (d18:1/24:0)were associated with increased risk. Moreover, we observed the enhanced expression of key enzymes involved in the SL pathway in blood cells from severe COVID-19 patients, suggesting a primary flow towards Cer generation in tandem with SM synthesis. These findings underscore the potential of SM as a prognostic biomarker for COVID-19 and highlight promising pharmacological targets. By targeting sphingolipid pathways, novel therapeutic strategies may emerge to mitigate the severity of COVID-19 and improve patient outcomes.
Collapse
Affiliation(s)
- Diana Mota Toro
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Pedro V. da Silva-Neto
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Jonatan C. S. de Carvalho
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Carlos A. Fuzo
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Malena M. Pérez
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Vinícius E. Pimentel
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Thais F. C. Fraga-Silva
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Camilla N. S. Oliveira
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Glaucia R. Caruso
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Adriana F. L. Vilela
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Pedro Nobre-Azevedo
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Thiago V. Defelippo-Felippe
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Jamille G. M. Argolo
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto–EERP, University of São Paulo–USP, Ribeirão Preto 14040-902, SP, Brazil; (J.G.M.A.); (A.P.M.F.)
| | - Augusto M. Degiovani
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto 14085-000, SP, Brazil; (A.M.D.); (F.M.O.)
| | - Fátima M. Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto 14085-000, SP, Brazil; (A.M.D.); (F.M.O.)
| | - Marley R. Feitosa
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Rogerio S. Parra
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Fernando C. Vilar
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil;
| | - Gilberto G. Gaspar
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil;
| | - José J. R. da Rocha
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
| | - Omar Feres
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Gabriel P. Costa
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo–USP, Ribeirão Preto 14040-900, SP, Brazil; (G.P.C.); (Á.A.T.)
| | - Sandra R. C. Maruyama
- Department of Genetics and Evolution, Center for Biological and Health Sciences, Federal University of São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil;
| | - Elisa M. S. Russo
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Ana Paula M. Fernandes
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto–EERP, University of São Paulo–USP, Ribeirão Preto 14040-902, SP, Brazil; (J.G.M.A.); (A.P.M.F.)
| | - Isabel K. F. M. Santos
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Adriana Malheiro
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Ruxana T. Sadikot
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Vânia L. D. Bonato
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Cristina R. B. Cardoso
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Marcelo Dias-Baruffi
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Átila A. Trapé
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo–USP, Ribeirão Preto 14040-900, SP, Brazil; (G.P.C.); (Á.A.T.)
| | - Lúcia H. Faccioli
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Carlos A. Sorgi
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | | |
Collapse
|
19
|
Lodge S, Lawler NG, Gray N, Masuda R, Nitschke P, Whiley L, Bong SH, Yeap BB, Dwivedi G, Spraul M, Schaefer H, Gil-Redondo R, Embade N, Millet O, Holmes E, Wist J, Nicholson JK. Integrative Plasma Metabolic and Lipidomic Modelling of SARS-CoV-2 Infection in Relation to Clinical Severity and Early Mortality Prediction. Int J Mol Sci 2023; 24:11614. [PMID: 37511373 PMCID: PMC10380980 DOI: 10.3390/ijms241411614] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/10/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
An integrative multi-modal metabolic phenotyping model was developed to assess the systemic plasma sequelae of SARS-CoV-2 (rRT-PCR positive) induced COVID-19 disease in patients with different respiratory severity levels. Plasma samples from 306 unvaccinated COVID-19 patients were collected in 2020 and classified into four levels of severity ranging from mild symptoms to severe ventilated cases. These samples were investigated using a combination of quantitative Nuclear Magnetic Resonance (NMR) spectroscopy and Mass Spectrometry (MS) platforms to give broad lipoprotein, lipidomic and amino acid, tryptophan-kynurenine pathway, and biogenic amine pathway coverage. All platforms revealed highly significant differences in metabolite patterns between patients and controls (n = 89) that had been collected prior to the COVID-19 pandemic. The total number of significant metabolites increased with severity with 344 out of the 1034 quantitative variables being common to all severity classes. Metabolic signatures showed a continuum of changes across the respiratory severity levels with the most significant and extensive changes being in the most severely affected patients. Even mildly affected respiratory patients showed multiple highly significant abnormal biochemical signatures reflecting serious metabolic deficiencies of the type observed in Post-acute COVID-19 syndrome patients. The most severe respiratory patients had a high mortality (56.1%) and we found that we could predict mortality in this patient sub-group with high accuracy in some cases up to 61 days prior to death, based on a separate metabolic model, which highlighted a different set of metabolites to those defining the basic disease. Specifically, hexosylceramides (HCER 16:0, HCER 20:0, HCER 24:1, HCER 26:0, HCER 26:1) were markedly elevated in the non-surviving patient group (Cliff's delta 0.91-0.95) and two phosphoethanolamines (PE.O 18:0/18:1, Cliff's delta = -0.98 and PE.P 16:0/18:1, Cliff's delta = -0.93) were markedly lower in the non-survivors. These results indicate that patient morbidity to mortality trajectories is determined relatively soon after infection, opening the opportunity to select more intensive therapeutic interventions to these "high risk" patients in the early disease stages.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Nathan G. Lawler
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Nicola Gray
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Reika Masuda
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
| | - Philipp Nitschke
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
| | - Luke Whiley
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Sze-How Bong
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
| | - Bu B. Yeap
- Medical School, University of Western Australia, Perth, WA 6150, Australia; (B.B.Y.); (G.D.)
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, WA 6150, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, WA 6150, Australia; (B.B.Y.); (G.D.)
- Department of Cardiology, Fiona Stanley Hospital, Perth, WA 6150, Australia
| | | | | | - Rubén Gil-Redondo
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain; (R.G.-R.); (N.E.); (O.M.)
| | - Nieves Embade
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain; (R.G.-R.); (N.E.); (O.M.)
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain; (R.G.-R.); (N.E.); (O.M.)
| | - Elaine Holmes
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for 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, UK
| | - Julien Wist
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| | - Jeremy K. Nicholson
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Institute of Global Health Innovation, Faculty of Medicine, Imperial College London, Faculty Building, South Kensington Campus, London SW7 2NA, UK
| |
Collapse
|
20
|
Chen P, Wu M, He Y, Jiang B, He ML. Metabolic alterations upon SARS-CoV-2 infection and potential therapeutic targets against coronavirus infection. Signal Transduct Target Ther 2023; 8:237. [PMID: 37286535 DOI: 10.1038/s41392-023-01510-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/18/2023] [Accepted: 05/19/2023] [Indexed: 06/09/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by coronavirus SARS-CoV-2 infection has become a global pandemic due to the high viral transmissibility and pathogenesis, bringing enormous burden to our society. Most patients infected by SARS-CoV-2 are asymptomatic or have mild symptoms. Although only a small proportion of patients progressed to severe COVID-19 with symptoms including acute respiratory distress syndrome (ARDS), disseminated coagulopathy, and cardiovascular disorders, severe COVID-19 is accompanied by high mortality rates with near 7 million deaths. Nowadays, effective therapeutic patterns for severe COVID-19 are still lacking. It has been extensively reported that host metabolism plays essential roles in various physiological processes during virus infection. Many viruses manipulate host metabolism to avoid immunity, facilitate their own replication, or to initiate pathological response. Targeting the interaction between SARS-CoV-2 and host metabolism holds promise for developing therapeutic strategies. In this review, we summarize and discuss recent studies dedicated to uncovering the role of host metabolism during the life cycle of SARS-CoV-2 in aspects of entry, replication, assembly, and pathogenesis with an emphasis on glucose metabolism and lipid metabolism. Microbiota and long COVID-19 are also discussed. Ultimately, we recapitulate metabolism-modulating drugs repurposed for COVID-19 including statins, ASM inhibitors, NSAIDs, Montelukast, omega-3 fatty acids, 2-DG, and metformin.
Collapse
Affiliation(s)
- Peiran Chen
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China
| | - Mandi Wu
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China
| | - Yaqing He
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China
| | - Binghua Jiang
- Cell Signaling and Proteomic Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China.
| |
Collapse
|
21
|
Lu S, Zhou Y, Hu Y, Wang J, Li H, Lin Y, Wang D, Xian J, Zhao S, Ma J, Zhu Z, Yang S, Meng Q, Kang Y, Chen B, Li W. Metatranscriptomic analysis revealed Prevotella as a potential biomarker of oropharyngeal microbiomes in SARS-CoV-2 infection. Front Cell Infect Microbiol 2023; 13:1161763. [PMID: 37333851 PMCID: PMC10272425 DOI: 10.3389/fcimb.2023.1161763] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/09/2023] [Indexed: 06/20/2023] Open
Abstract
Background and objectives Disease severity and prognosis of coronavirus disease 2019 (COVID-19) disease with other viral infections can be affected by the oropharyngeal microbiome. However, limited research had been carried out to uncover how these diseases are differentially affected by the oropharyngeal microbiome of the patient. Here, we aimed to explore the characteristics of the oropharyngeal microbiota of COVID-19 patients and compare them with those of patients with similar symptoms. Methods COVID-19 was diagnosed in patients through the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Characterization of the oropharyngeal microbiome was performed by metatranscriptomic sequencing analyses of oropharyngeal swab specimens from 144 COVID-19 patients, 100 patients infected with other viruses, and 40 healthy volunteers. Results The oropharyngeal microbiome diversity in patients with SARS-CoV-2 infection was different from that of patients with other infections. Prevotella and Aspergillus could play a role in the differentiation between patients with SARS-CoV-2 infection and patients with other infections. Prevotella could also influence the prognosis of COVID-19 through a mechanism that potentially involved the sphingolipid metabolism regulation pathway. Conclusion The oropharyngeal microbiome characterization was different between SARS-CoV-2 infection and infections caused by other viruses. Prevotella could act as a biomarker for COVID-19 diagnosis and of host immune response evaluation in SARS-CoV-2 infection. In addition, the cross-talk among Prevotella, SARS-CoV-2, and sphingolipid metabolism pathways could provide a basis for the precise diagnosis, prevention, control, and treatment of COVID-19.
Collapse
Affiliation(s)
- Sifen Lu
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongzhao Zhou
- Department of Integrated Care Management Center, Frontier Science Center of Disease Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ya Hu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Wang
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Honghao Li
- Department of Hospital Management, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Lin
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Denian Wang
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinghong Xian
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Shengmei Zhao
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Jinmin Ma
- Beijing Genomics Institution (BGI)-PathoGenesis Pharmaceutical Technology, Beijing Genomics Institution (BGI)-Shenzhen, Shenzhen, China
| | - Zhongyi Zhu
- Beijing Genomics Institution (BGI)-PathoGenesis Pharmaceutical Technology, Beijing Genomics Institution (BGI)-Shenzhen, Shenzhen, China
| | - Shengying Yang
- Department of Computer and Software, Jincheng College of Chengdu, Chengdu, China
| | - Qinghui Meng
- Beijing Milu Ecological Research Center, Beijing Research Institute of Science and Technology, Beijing, China
| | - Yulin Kang
- Institute of Environmental Information, Chinese Research academy of Environmental Sciences, Beijing, China
| | - Bojiang Chen
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weimin Li
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Integrated Care Management Center, Frontier Science Center of Disease Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Zhu H, Byrnes C, Lee YT, Tuymetova G, Duffy HBD, Bakir JY, Pettit SN, Angina J, Springer DA, Allende ML, Kono M, Proia RL. SARS-CoV-2 ORF3a expression in brain disrupts the autophagy-lysosomal pathway, impairs sphingolipid homeostasis, and drives neuropathogenesis. FASEB J 2023; 37:e22919. [PMID: 37071464 DOI: 10.1096/fj.202300149r] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/10/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes injury to multiple organ systems, including the brain. SARS-CoV-2's neuropathological mechanisms may include systemic inflammation and hypoxia, as well as direct cell damage resulting from viral infections of neurons and glia. How the virus directly causes injury to brain cells, acutely and over the long term, is not well understood. In order to gain insight into this process, we studied the neuropathological effects of open reading frame 3a (ORF3a), a SARS-CoV-2 accessory protein that is a key pathological factor of the virus. Forced ORF3a brain expression in mice caused the rapid onset of neurological impairment, neurodegeneration, and neuroinflammation-key neuropathological features found in coronavirus disease (COVID-19, which is caused by SARS-CoV-2 infection). Furthermore, ORF3a expression blocked autophagy progression in the brain and caused the neuronal accumulation of α-synuclein and glycosphingolipids, all of which are linked to neurodegenerative disease. Studies with ORF3-expressing HeLa cells confirmed that ORF3a disrupted the autophagy-lysosomal pathway and blocked glycosphingolipid degradation, resulting in their accumulation. These findings indicate that, in the event of neuroinvasion by SARS-CoV-2, ORF3a expression in brain cells may drive neuropathogenesis and be an important mediator of both short- and long-term neurological manifestations of COVID-19.
Collapse
Affiliation(s)
- Hongling Zhu
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Colleen Byrnes
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Y Terry Lee
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Galina Tuymetova
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Hannah B D Duffy
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Jenna Y Bakir
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Sydney N Pettit
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Jabili Angina
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Danielle A Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria L Allende
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Mari Kono
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Richard L Proia
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
23
|
Avraham R, Melamed S, Achdout H, Erez N, Israeli O, Barlev-Gross M, Pasmanik-Chor M, Paran N, Israely T, Vitner EB. Antiviral activity of glucosylceramide synthase inhibitors in alphavirus infection of the central nervous system. Brain Commun 2023; 5:fcad086. [PMID: 37168733 PMCID: PMC10165247 DOI: 10.1093/braincomms/fcad086] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 12/23/2022] [Accepted: 03/23/2023] [Indexed: 05/13/2023] Open
Abstract
Virus-induced CNS diseases impose a considerable human health burden worldwide. For many viral CNS infections, neither antiviral drugs nor vaccines are available. In this study, we examined whether the synthesis of glycosphingolipids, major membrane lipid constituents, could be used to establish an antiviral therapeutic target. We found that neuroinvasive Sindbis virus altered the sphingolipid levels early after infection in vitro and increased the levels of gangliosides GA1 and GM1 in the sera of infected mice. The alteration in the sphingolipid levels appears to play a role in neuroinvasive Sindbis virus replication, as treating infected cells with UDP-glucose ceramide glucosyltransferase (UGCG) inhibitors reduced the replication rate. Moreover, the UGCG inhibitor GZ-161 increased the survival rates of Sindbis-infected mice, most likely by reducing the detrimental immune response activated by sphingolipids in the brains of Sindbis virus-infected mice. These findings suggest a role for glycosphingolipids in the host immune response against neuroinvasive Sindbis virus and suggest that UGCG inhibitors should be further examined as antiviral therapeutics for viral infections of the CNS.
Collapse
Affiliation(s)
- Roy Avraham
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Hagit Achdout
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Noam Erez
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Ofir Israeli
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Moria Barlev-Gross
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George S. Wise Faculty of Life Science, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Tomer Israely
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Einat B Vitner
- Correspondence to: Einat B. Vitner Department of Infectious Diseases Israel Institute for Biological Research P.O.B 19, 7410001 Ness-Ziona, Israel E-mail:
| |
Collapse
|
24
|
Trivedi VS, Magnusen AF, Rani R, Marsili L, Slavotinek AM, Prows DR, Hopkin RJ, McKay MA, Pandey MK. Targeting the Complement-Sphingolipid System in COVID-19 and Gaucher Diseases: Evidence for a New Treatment Strategy. Int J Mol Sci 2022; 23:14340. [PMID: 36430817 PMCID: PMC9695449 DOI: 10.3390/ijms232214340] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2)-induced disease (COVID-19) and Gaucher disease (GD) exhibit upregulation of complement 5a (C5a) and its C5aR1 receptor, and excess synthesis of glycosphingolipids that lead to increased infiltration and activation of innate and adaptive immune cells, resulting in massive generation of pro-inflammatory cytokines, chemokines and growth factors. This C5a-C5aR1-glycosphingolipid pathway- induced pro-inflammatory environment causes the tissue damage in COVID-19 and GD. Strikingly, pharmaceutically targeting the C5a-C5aR1 axis or the glycosphingolipid synthesis pathway led to a reduction in glycosphingolipid synthesis and innate and adaptive immune inflammation, and protection from the tissue destruction in both COVID-19 and GD. These results reveal a common involvement of the complement and glycosphingolipid systems driving immune inflammation and tissue damage in COVID-19 and GD, respectively. It is therefore expected that combined targeting of the complement and sphingolipid pathways could ameliorate the tissue destruction, organ failure, and death in patients at high-risk of developing severe cases of COVID-19.
Collapse
Affiliation(s)
- Vyoma Snehal Trivedi
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Albert Frank Magnusen
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Reena Rani
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Luca Marsili
- Department of Neurology, James J. and Joan A. Gardner Center for Parkinson’s Disease and Movement Disorders, University of Cincinnati, 3113 Bellevue Ave, Cincinnati, OH 45219, USA
| | - Anne Michele Slavotinek
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Daniel Ray Prows
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Robert James Hopkin
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Mary Ashley McKay
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| |
Collapse
|
25
|
Hu Q, Liu B, Fan Y, Zheng Y, Wen F, Yu U, Wang W. Multi-omics association analysis reveals interactions between the oropharyngeal microbiome and the metabolome in pediatric patients with influenza A virus pneumonia. Front Cell Infect Microbiol 2022; 12:1011254. [PMID: 36389138 PMCID: PMC9651038 DOI: 10.3389/fcimb.2022.1011254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
Children are at high risk for influenza A virus (IAV) infections, which can develop into severe illnesses. However, little is known about interactions between the microbiome and respiratory tract metabolites and their impact on the development of IAV pneumonia in children. Using a combination of liquid chromatography tandem mass spectrometry (LC-MS/MS) and 16S rRNA gene sequencing, we analyzed the composition and metabolic profile of the oropharyngeal microbiota in 49 pediatric patients with IAV pneumonia and 42 age-matched healthy children. The results indicate that compared to healthy children, children with IAV pneumonia exhibited significant changes in the oropharyngeal macrobiotic structure (p = 0.001), and significantly lower microbial abundance and diversity (p < 0.05). These changes came with significant disturbances in the levels of oropharyngeal metabolites. Intergroup differences were observed in 204 metabolites mapped to 36 metabolic pathways. Significantly higher levels of sphingolipid (sphinganine and phytosphingosine) and propanoate (propionic acid and succinic acid) metabolism were observed in patients with IAV pneumonia than in healthy controls. Using Spearman’s rank-correlation analysis, correlations between IAV pneumonia-associated discriminatory microbial genera and metabolites were evaluated. The results indicate significant correlations and consistency in variation trends between Streptococcus and three sphingolipid metabolites (phytosphingosine, sphinganine, and sphingosine). Besides these three sphingolipid metabolites, the sphinganine-to-sphingosine ratio and the joint analysis of the three metabolites indicated remarkable diagnostic efficacy in children with IAV pneumonia. This study confirmed significant changes in the characteristics and metabolic profile of the oropharyngeal microbiome in pediatric patients with IAV pneumonia, with high synergy between the two factors. Oropharyngeal sphingolipid metabolites may serve as potential diagnostic biomarkers of IAV pneumonia in children.
Collapse
Affiliation(s)
- Qian Hu
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen, China
| | - Baiming Liu
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yanqun Fan
- Department of Trans-omics Research, Biotree Metabolomics Technology Research Center, Shanghai, China
| | - Yuejie Zheng
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen, China
| | - Feiqiu Wen
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Uet Yu
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
- *Correspondence: Wenjian Wang, ; Uet Yu,
| | - Wenjian Wang
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen, China
- *Correspondence: Wenjian Wang, ; Uet Yu,
| |
Collapse
|
26
|
Hou J, Wei Y, Zou J, Jaffery R, Liang S, Zheng C, Chen K, Shi PY, Chen Y, Xie X, Peng W. Integrated multi-omics analyses identify key anti-viral host factors and pathways controlling SARS-CoV-2 infection. RESEARCH SQUARE 2022:rs.3.rs-1910932. [PMID: 36032971 PMCID: PMC9413708 DOI: 10.21203/rs.3.rs-1910932/v1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Host anti-viral factors are essential for controlling SARS-CoV-2 infection but remain largely unknown due to the biases of previous large-scale studies toward pro-viral host factors. To fill in this knowledge gap, we performed a genome-wide CRISPR dropout screen and integrated analyses of the multi-omics data of the CRISPR screen, genome-wide association studies, single-cell RNA-seq, and host-virus proteins or protein/RNA interactome. This study has uncovered many host factors that were missed by previous studies, including the components of V-ATPases, ESCRT, and N-glycosylation pathways that modulated viral entry and/or replication. The cohesin complex was also identified as a novel anti-viral pathway, suggesting an important role of three-dimensional chromatin organization in mediating host-viral interaction. Furthermore, we discovered an anti-viral regulator KLF5, a transcriptional factor involved in sphingolipid metabolism, which was up-regulated and harbored genetic variations linked to the COVID-19 patients with severe symptoms. Our results provide a resource for understanding the host anti-viral network during SARS-CoV-2 infection and may help develop new countermeasure strategies.
Collapse
Affiliation(s)
| | - Yanjun Wei
- The University of Texas MD Anderson Cancer Center
| | - Jing Zou
- University of Texas Medical Branch
| | | | | | | | - Ken Chen
- The University of Texas MD Anderson Cancer Center
| | | | - Yiwen Chen
- The University of Texas MD Anderson Cancer Center
| | | | | |
Collapse
|
27
|
Smith CD, Maines LW, Keller SN, Katz Ben-Yair V, Fathi R, Plasse TF, Levitt ML. Recent Progress in the Development of Opaganib for the Treatment of Covid-19. Drug Des Devel Ther 2022; 16:2199-2211. [PMID: 35855741 PMCID: PMC9288228 DOI: 10.2147/dddt.s367612] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/02/2022] [Indexed: 12/15/2022] Open
Abstract
The Covid-19 pandemic driven by the SARS-CoV-2 virus continues to exert extensive humanitarian and economic stress across the world. Although antivirals active against mild disease have been identified recently, new drugs to treat moderate and severe Covid-19 patients are needed. Sphingolipids regulate key pathologic processes, including viral proliferation and pathologic host inflammation. Opaganib (aka ABC294640) is a first-in-class clinical drug targeting sphingolipid metabolism for the treatment of cancer and inflammatory diseases. Recent work demonstrates that opaganib also has antiviral activity against several viruses including SARS-CoV-2. A recently completed multinational Phase 2/3 clinical trial of opaganib in patients hospitalized with Covid-19 demonstrated that opaganib can be safely administered to these patients, and more importantly, resulted in a 62% decrease in mortality in a large subpopulation of patients with moderately severe Covid-19. Furthermore, acceleration of the clearance of the virus was observed in opaganib-treated patients. Understanding the biochemical mechanism for the anti-SARS-CoV-2 activity of opaganib is essential for optimizing Covid-19 treatment protocols. Opaganib inhibits three key enzymes in sphingolipid metabolism: sphingosine kinase-2 (SK2); dihydroceramide desaturase (DES1); and glucosylceramide synthase (GCS). Herein, we describe a tripartite model by which opaganib suppresses infection and replication of SARS-CoV-2 by inhibiting SK2, DES1 and GCS. The potential impact of modulation of sphingolipid signaling on multi-organ dysfunction in Covid-19 patients is also discussed.
Collapse
Affiliation(s)
- Charles D Smith
- Apogee Biotechnology Corporation, Hummelstown, PA, USA
- Correspondence: Charles D Smith, Apogee Biotechnology Corporation, 1214 Research Blvd, Suite 2015, Hummelstown, PA, 17036, USA, Tel +1 843 814 9257, Email
| | - Lynn W Maines
- Apogee Biotechnology Corporation, Hummelstown, PA, USA
| | | | | | | | | | | |
Collapse
|
28
|
Farley SE, Kyle JE, Leier HC, Bramer LM, Weinstein JB, Bates TA, Lee JY, Metz TO, Schultz C, Tafesse FG. A global lipid map reveals host dependency factors conserved across SARS-CoV-2 variants. Nat Commun 2022; 13:3487. [PMID: 35715395 PMCID: PMC9203258 DOI: 10.1038/s41467-022-31097-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 06/01/2022] [Indexed: 12/31/2022] Open
Abstract
A comprehensive understanding of host dependency factors for SARS-CoV-2 remains elusive. Here, we map alterations in host lipids following SARS-CoV-2 infection using nontargeted lipidomics. We find that SARS-CoV-2 rewires host lipid metabolism, significantly altering hundreds of lipid species to effectively establish infection. We correlate these changes with viral protein activity by transfecting human cells with each viral protein and performing lipidomics. We find that lipid droplet plasticity is a key feature of infection and that viral propagation can be blocked by small-molecule glycerolipid biosynthesis inhibitors. We find that this inhibition was effective against the main variants of concern (alpha, beta, gamma, and delta), indicating that glycerolipid biosynthesis is a conserved host dependency factor that supports this evolving virus.
Collapse
Affiliation(s)
- Scotland E Farley
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Jennifer E Kyle
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, USA
| | - Hans C Leier
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Lisa M Bramer
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, USA
| | - Jules B Weinstein
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Timothy A Bates
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Joon-Yong Lee
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, USA
| | - Thomas O Metz
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, USA
| | - Carsten Schultz
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Fikadu G Tafesse
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
29
|
Liu P, Hu D, Yuan L, Lian Z, Yao X, Zhu Z, Li X. Metabolomics Analysis of PK-15 Cells with Pseudorabies Virus Infection Based on UHPLC-QE-MS. Viruses 2022; 14:v14061158. [PMID: 35746630 PMCID: PMC9229976 DOI: 10.3390/v14061158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 12/02/2022] Open
Abstract
Viruses depend on the metabolic mechanisms of the host to support viral replication. We utilize an approach based on ultra-high-performance liquid chromatography/Q Exactive HF-X Hybrid Quadrupole-Orbitrap Mass (UHPLC-QE-MS) to analyze the metabolic changes in PK-15 cells induced by the infections of the pseudorabies virus (PRV) variant strain and Bartha K61 strain. Infections with PRV markedly changed lots of metabolites, when compared to the uninfected cell group. Additionally, most of the differentially expressed metabolites belonged to glycerophospholipid metabolism, sphingolipid metabolism, purine metabolism, and pyrimidine metabolism. Lipid metabolites account for the highest proportion (around 35%). The results suggest that those alterations may be in favor of virion formation and genome amplification to promote PRV replication. Different PRV strains showed similar results. An understanding of PRV-induced metabolic reprogramming will provide valuable information for further studies on PRV pathogenesis and the development of antiviral therapy strategies.
Collapse
Affiliation(s)
- Panrao Liu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (P.L.); (D.H.); (L.Y.); (Z.L.); (X.Y.); (Z.Z.)
| | - Danhe Hu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (P.L.); (D.H.); (L.Y.); (Z.L.); (X.Y.); (Z.Z.)
| | - Lili Yuan
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (P.L.); (D.H.); (L.Y.); (Z.L.); (X.Y.); (Z.Z.)
| | - Zhengmin Lian
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (P.L.); (D.H.); (L.Y.); (Z.L.); (X.Y.); (Z.Z.)
| | - Xiaohui Yao
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (P.L.); (D.H.); (L.Y.); (Z.L.); (X.Y.); (Z.Z.)
| | - Zhenbang Zhu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (P.L.); (D.H.); (L.Y.); (Z.L.); (X.Y.); (Z.Z.)
| | - Xiangdong Li
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (P.L.); (D.H.); (L.Y.); (Z.L.); (X.Y.); (Z.Z.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: ; Tel.: +86-514-8797-9036
| |
Collapse
|
30
|
Network Pharmacology Study to Elucidate the Key Targets of Underlying Antihistamines against COVID-19. Curr Issues Mol Biol 2022; 44:1597-1609. [PMID: 35723367 PMCID: PMC9164076 DOI: 10.3390/cimb44040109] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/02/2022] [Accepted: 04/06/2022] [Indexed: 12/19/2022] Open
Abstract
Antihistamines have potent efficacy to alleviate COVID-19 (Coronavirus disease 2019) symptoms such as anti-inflammation and as a pain reliever. However, the pharmacological mechanism(s), key target(s), and drug(s) are not documented well against COVID-19. Thus, we investigated to decipher the most significant components and how its research methodology was utilized by network pharmacology. The list of 32 common antihistamines on the market were retrieved via drug browsing databases. The targets associated with the selected antihistamines and the targets that responded to COVID-19 infection were identified by the Similarity Ensemble Approach (SEA), SwissTargetPrediction (STP), and PubChem, respectively. We described bubble charts, the Pathways-Targets-Antihistamines (PTA) network, and the protein–protein interaction (PPI) network on the RPackage via STRING database. Furthermore, we utilized the AutoDock Tools software to perform molecular docking tests (MDT) on the key targets and drugs to evaluate the network pharmacological perspective. The final 15 targets were identified as core targets, indicating that Neuroactive ligand–receptor interaction might be the hub-signaling pathway of antihistamines on COVID-19 via bubble chart. The PTA network was constructed by the RPackage, which identified 7 pathways, 11 targets, and 30 drugs. In addition, GRIN2B, a key target, was identified via topological analysis of the PPI network. Finally, we observed that the GRIN2B-Loratidine complex was the most stable docking score with −7.3 kcal/mol through molecular docking test. Our results showed that Loratadine might exert as an antagonist on GRIN2B via the neuroactive ligand–receptor interaction pathway. To sum up, we elucidated the most potential antihistamine, a key target, and a key pharmacological pathway as alleviating components against COVID-19, supporting scientific evidence for further research.
Collapse
|