1
|
de Bruin ACM, Lamers MM, Haagmans BL, Leijten LM, Fouchier RAM, Richard M. Long-term culture of chicken tracheal organoids for the purpose of avian influenza virus research. Virol J 2025; 22:99. [PMID: 40234888 PMCID: PMC11998437 DOI: 10.1186/s12985-025-02714-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
There is an increasing need for reproducible long-term in vitro primary cell culture systems that are representative of the avian respiratory tract to study pathogens like highly pathogenic avian influenza viruses (HPAIVs), which threaten poultry, wildlife, and human health. Self-renewing organoid cultures allow for long-term culture, due to the presence of tissue-resident stem cells, and can approximate the in vivo cellular diversity and organization of tissues. Efforts to establish avian organoid cultures have been limited to the intestinal tract. Here, we describe the isolation and long-term culture of chicken tracheal organoids (CTOs). The CTO cultures were passaged for three to four months and cryopreserved at different stages. Mucociliary differentiation of CTOs was promoted by culture at air-liquid-interface, after which the pseudostratified epithelial cell layer of the avian trachea was recapitulated, including ciliated, goblet, and basal cells. Inoculation of CTO-derived 2D cultures with low pathogenic avian influenza viruses (LPAIVs) and HPAIVs showed that the appropriate receptors, as confirmed by virus histochemistry, and proteases to sustain multi-cycle replication of LPAIVs were expressed and that HPAIVs preferentially disseminated to the endothelium of epithelial/endothelial co-culture systems. Taken together, CTOs represent a useful tool for research on the avian respiratory tract, and their application will generate new insights into host-pathogen interactions, including HPAIV tropism.
Collapse
Affiliation(s)
- Anja C M de Bruin
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
- Institute of Virology, Medical University Innsbruck, Innsbruck, Austria
| | - Mart M Lamers
- Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore, Singapore
| | - Bart L Haagmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lonneke M Leijten
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mathilde Richard
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Chen Z, Yang Z, Rao L, Li C, Zang N, Liu E. Human adenovirus type 7 (HAdV-7) infection induces pulmonary vascular endothelial injury through the activation of endothelial autophagy. Respir Res 2024; 25:425. [PMID: 39633448 PMCID: PMC11619570 DOI: 10.1186/s12931-024-03025-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND HAdV-7 is a prevalent pathogen that can cause severe pneumonia in children. Previous studies have shown a significant increase in serum levels of vascular permeability factor (VPF/VEGF) and viral load in pediatric patients with fatal HAdV-7 infection, suggesting potential damage to the pulmonary vascular endothelium. Further research is necessary to elucidate the underlying mechanism. METHODS The human lung microvascular endothelial cell line-5a and human CD46 mice were used for in vitro and in vivo experiments, respectively. RNA-seq was employed for correlative omics analysis. Viral infection and copy status were examined using transmission electron microscopy to observe virus particles, immunofluorescence to detect the viral protein Hexon, and qPCR to assess HAdV-7 fiber gene copies. Various methods, including ELISAs for VEGF and other injury markers, the CCK8 assay for cell viability, and flow cytometry for endothelium numbers, were employed to evaluate endothelial damage. Acute lung injury severity was evaluated by scoring pathological inflammation and measuring pulmonary vascular permeability. Autophagy activation was assessed by observing autophagosomes and validating marker proteins. RESULTS GSEA analysis showed significant enrichment of gene sets related to endothelial functions (barrier, defense, and regeneration) and ALI in the HAdV-7-infected group. GO analysis indicated an enrichment of autophagy-related pathways linked to cell death. Subsequently, successful signs of HAdV-7 infection and replication were observed in the endothelium, including cytopathic effects, intracellular virions, and increased HAdV-7 fiber gene copies. Endothelial injury, including mitochondrial damage, decreased endothelium, and elevated levels of endothelial injury markers such as VEGF, sICAM-1, sVCAM-1, E-selectin, ESM1, MCP1, and IL1β were observed after HAdV-7 infection. Additionally, evidence of leaky lung blood vessels and ALI was observed, including progressive weight loss, elevated pulmonary vascular permeability, and severe lung consolidation. Furthermore, HAdV-7 infection induced autophagosome formation in the endothelium and triggered complete cell autophagy. Importantly, inhibiting autophagic flux reduced VEGF levels and other endothelial injury markers, decreased viral load, improved cell survival rate, alleviated pulmonary vessel leakage, and mitigated lung inflammation. CONCLUSIONS HAdV-7 successfully infects pulmonary vascular endothelium and replicates effectively, causing injury to the endothelium, high VEGF expression and viral load in the serum, as well as ALI/ARDS. Autophagy inhibitors can alleviate endothelial injury, inhibit viral replication, relieve leakage from the vasculature, and reduce lung inflammation.
Collapse
Affiliation(s)
- Zhihe Chen
- Department of Respiratory Medicine Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, 400014, China
- Pediatric Department, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Zhongying Yang
- Department of Respiratory Medicine Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, 400014, China
| | - Lifen Rao
- Department of Respiratory Medicine Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, 400014, China
| | - Changgen Li
- Department of Respiratory Medicine Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, 400014, China
| | - Na Zang
- Department of Respiratory Medicine Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, 400014, China.
| | - Enmei Liu
- Department of Respiratory Medicine Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, 400014, China.
| |
Collapse
|
3
|
Li K, Feng KC, Simon M, Fu Y, Galanakis D, Mueller S, Rafailovich MH. Molecular Basis for Surface-Initiated Non-Thrombin-Generated Clot Formation Following Viral Infection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30703-30714. [PMID: 38848451 DOI: 10.1021/acsami.4c02918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
In this paper, we propose a model that connects two standard inflammatory responses to viral infection, namely, elevation of fibrinogen and the lipid drop shower, to the initiation of non-thrombin-generated clot formation. In order to understand the molecular basis for the formation of non-thrombin-generated clots following viral infection, human epithelial and Madin-Darby Canine Kidney (MDCK, epithelial) cells were infected with H1N1, OC43, and adenovirus, and conditioned media was collected, which was later used to treat human umbilical vein endothelial cells and human lung microvascular endothelial cells. After direct infection or after exposure to conditioned media from infected cells, tissue surfaces of both epithelial and endothelial cells, exposed to 8 mg/mL fibrinogen, were observed to initiate fibrillogenesis in the absence of thrombin. No fibers were observed after direct viral exposure of the endothelium or when the epithelium cells were exposed to SARS-CoV-2 isolated spike proteins. Heating the conditioned media to 60 °C had no effect on fibrillogenesis, indicating that the effect was not enzymatic but rather associated with relatively thermally stable inflammatory factors released soon after viral infection. Spontaneous fibrillogenesis had previously been reported and interpreted as being due to the release of the alpha C domains due to strong interactions of the interior of the fibrinogen molecule in contact with hydrophobic material surfaces rather than cleavage of the fibrinopeptides. Contact angle goniometry and immunohistochemistry were used to demonstrate that the lipids produced within the epithelium and released in the conditioned media, probably after the death of infected epithelial cells, formed a hydrophobic residue responsible for fibrillogenesis. Hence, the standard inflammatory response constitutes the ideal conditions for surface-initiated clot formation.
Collapse
Affiliation(s)
- Kao Li
- School of Biomedicine and Nursing, Shandong Institute of Petroleum and Chemical Technology, Dongying 257061, Shandong, China
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Kuan-Che Feng
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Marcia Simon
- Department of Oral Biology and Pathology, Stony Brook University Medical Center, Stony Brook, New York 11794, United States
| | - Yuyang Fu
- Dongying Stem Cell Bank Medical Technology Co., Ltd., Dongying 257000, Shandong, China
| | - Dennis Galanakis
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, New York 11720, United States
| | | | - Miriam H Rafailovich
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
4
|
Chenane HR, Lingas G, Menidjel R, Laouenan C, Tubiana S, Descamps D, Le Hingrat Q, Abel L, Guedj J, Malhotra S, Kumar-Singh S, Visseaux B, Ghosn J, Charpentier C, Lebourgeois S. High sera levels of SARS-CoV-2 N antigen are associated with death in hospitalized COVID-19 patients. J Med Virol 2023; 95:e29247. [PMID: 38009713 DOI: 10.1002/jmv.29247] [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: 04/19/2023] [Revised: 09/05/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023]
Abstract
The presence of free severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid-antigen in sera (N-antigenemia) has been shown in COVID-19 patients. However, the link between the quantitative levels of N-antigenemia and COVID-19 disease severity is not entirely understood. To assess the dynamics and clinical association of N-antigen sera levels with disease severity in COVID-19 patients, we analyzed data from patients included in the French COVID cohort, with at least one sera sample between January and September 2020. We assessed N-antigenemia levels and anti-N IgG titers, and patient outcomes was classified in two groups, survival or death. In samples collected within 8 days since symptom onset, we observed that deceased patients had a higher positivity rate (93% vs. 81%; p < 0.001) and higher median levels of predicted N-antigenemia (2500 vs. 1200 pg/mL; p < 0.001) than surviving patients. Predicted time to N-antigen clearance in sera was prolonged in deceased patients compared to survivors (23.3 vs 19.3 days; p < 0.0001). In a subset of patients with both sera and nasopharyngeal (NP) swabs, predicted time to N-antigen clearance in sera was prolonged in deceased patients (p < 0.001), whereas NP viral load clearance did not differ between the groups (p = 0.07). Our results demonstrate a strong relationship between N-antigenemia levels and COVID-19 severity on a prospective cohort.
Collapse
Affiliation(s)
| | | | - Reyene Menidjel
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
| | - Cédric Laouenan
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Centre d'Investigations cliniques-Epidémiologie Clinique 1425, Hôpital Bichat, Paris, France
| | - Sarah Tubiana
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Centre d'Investigations cliniques-Epidémiologie Clinique 1425, Hôpital Bichat, Paris, France
| | - Diane Descamps
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Service de Virologie, Hôpital Bichat, Paris, France
| | - Quentin Le Hingrat
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Service de Virologie, Hôpital Bichat, Paris, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Jérémie Guedj
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
| | - Surbhi Malhotra
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Molecular Pathology group, Cell Biology & Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Benoit Visseaux
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
| | - Jade Ghosn
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Service de Maladies Infectieuses et Tropicales, Hôpital Bichat, Paris, France
| | - Charlotte Charpentier
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Service de Virologie, Hôpital Bichat, Paris, France
| | | |
Collapse
|
5
|
Bogot NR, Steiner R, Helviz Y, Weiss C, Cherniavsky K, Pichkhadze O, Ball L, Frank Y, Levin P, Pelosi P, Benjaminov O, Einav S. Distribution of Aeration and Pulmonary Blood Volume in Healthy, ARDS and COVID-19 Lungs: A Dual-Energy Computed Tomography Retrospective Cohort Study. Acad Radiol 2023; 30:2548-2556. [PMID: 36966073 PMCID: PMC10035816 DOI: 10.1016/j.acra.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023]
Abstract
RATIONALE AND OBJECTIVES Few reports have studied lung aeration and perfusion in normal lungs, COVID-19, and ARDS from other causes (NC-ARDS) using dual-energy computed tomography pulmonary angiograms (DE-CTPA). To describe lung aeration and blood-volume distribution using DE-CTPAs of patients with NC-ARDS, COVID-19, and controls with a normal DE-CTPA ("healthy lungs"). We hypothesized that each of these conditions has unique ranges of aeration and pulmonary blood volumes. MATERIALS AND METHODS This retrospective, single-center study of DE-CTPAs included patients with COVID-19, NC-ARDS (Berlin criteria), and controls. Patients with macroscopic pulmonary embolisms were excluded. The outcomes studied were the (1) lung blood-volume in areas with different aeration levels (normal, ground glass opacities [GGO], consolidated lung) and (2) aeration/blood-volume ratios. RESULTS Included were 20 patients with COVID-19 (10 milds, 10 moderate-severe), six with NC-ARDS, and 12 healthy-controls. Lung aeration was lowest in patients with severe COVID-19 24% (IQR13%-31%) followed by those with NC-ARDS 40%(IQR21%-46%). Blood-volume in GGO was lowest in patients with COVID-19 [moderate-severe:-28.6 (IQR-33.1-23.2); mild: -30.1 (IQR-33.3-23.4)] and highest in normally aerated areas in NC-ARDS -37.4 (IQR-52.5-30.2-) and moderate-severe COVID-19 -33.5(IQR-44.2-28.5). The median aeration/blood-volume ratio was lowest in severe COVID-19 but some values overlapped with those observed among patients with NC-ARDS. CONCLUSION Severe COVID-19 disease is associated with low total aerated lung volume and blood-volume in areas with GGO and overall aeration/blood volume ratios, and with high blood volume in normal lung areas. In this hypothesis-generating study, these findings were most pronounced in severe COVID disease. Larger studies are needed to confirm these preliminary findings.
Collapse
Affiliation(s)
| | | | - Yigal Helviz
- The Intensive Care Unit, Jerusalem, Israel; The Shaare Zedek Medical Center, Hebrew University Faculty of Medicine, Samuel Byte 12, Jerusalem, Israel
| | - Chedva Weiss
- The Intensive Care Unit, Jerusalem, Israel; Department of pediatrics, Hadassah Medical Center Jerusalem, Jerusalem, Israel
| | | | | | - Lorenzo Ball
- The Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy; Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Yigal Frank
- Department of Radiology(,) Jerusalem, Israel
| | - Philip Levin
- The Intensive Care Unit, Jerusalem, Israel; The Shaare Zedek Medical Center, Hebrew University Faculty of Medicine, Samuel Byte 12, Jerusalem, Israel
| | - Paolo Pelosi
- The Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy; Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | | | - Sharon Einav
- The Intensive Care Unit, Jerusalem, Israel; The Shaare Zedek Medical Center, Hebrew University Faculty of Medicine, Samuel Byte 12, Jerusalem, Israel.
| |
Collapse
|
6
|
Li K, Huang Z, Liu C, Xu Y, Chen W, Shi L, Li C, Zhou F, Zhou F. Transcriptomic analysis of human pulmonary microvascular endothelial cells treated with LPS. Cell Signal 2023; 111:110870. [PMID: 37633475 DOI: 10.1016/j.cellsig.2023.110870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Acute respiratory distress syndrome (ARDS) has a rapid onset and progression, which lead to the severity and complexity of the primary disease and significantly increase the fatality rate of patients. Transcriptomics provides some ideas for clarifying the mechanism of ARDS, exploring prevention and treatment targets, and searching for related specific markers. In this study, RNA-Seq technology was used to observe the gene expression of human pulmonary microvascular endothelial cells (PMVECs) induced by LPS, and to excavate the key genes and signaling pathways in ARDS process. A total of 2300 up-regulated genes were detected, and a corresponding 1696 down-regulated genes were screened. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and protein-protein interaction (PPI) were also used for functional annotation of key genes. TFDP1 was identified as a cell cycle-dependent differentially expressed gene, and its reduced expression was verified in LPS-treated PMVECs and lung tissues of CLP-induced mice. In addition, the inhibition of TFDP1 on inflammation and apoptosis, and the promotion of proliferation were confirmed. The decreased expression of E2F1, Rb, CDK1 and the activation of MAPK signaling pathway were substantiated in the in vivo and in vitro models of ARDS. Moreover, SREBF1 has been demonstrated to be involved in cell cycle arrest in PMVECs by inhibiting CDK1. Our study shows that transcriptomics combined with basic research can broaden the investigation of ARDS mechanisms and may provide a basis for future mechanistic innovations.
Collapse
Affiliation(s)
- Kaili Li
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China.
| | - Zuotian Huang
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, 400030 Chongqing Municipality, China
| | - Chang Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China.
| | - Yuanyuan Xu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Wei Chen
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Lu Shi
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Can Li
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Fawei Zhou
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Fachun Zhou
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China; Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China.
| |
Collapse
|
7
|
Guo TJF, Singhera GK, Leung JM, Dorscheid DR. Airway Epithelial-Derived Immune Mediators in COVID-19. Viruses 2023; 15:1655. [PMID: 37631998 PMCID: PMC10458661 DOI: 10.3390/v15081655] [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: 06/13/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The airway epithelium, which lines the conducting airways, is central to the defense of the lungs against inhaled particulate matter and pathogens such as SARS-CoV-2, the virus that causes COVID-19. Recognition of pathogens results in the activation of an innate and intermediate immune response which involves the release of cytokines and chemokines by the airway epithelium. This response can inhibit further viral invasion and influence adaptive immunity. However, severe COVID-19 is characterized by a hyper-inflammatory response which can give rise to clinical presentations including lung injury and lead to acute respiratory distress syndrome, viral pneumonia, coagulopathy, and multi-system organ failure. In response to SARS-CoV-2 infection, the airway epithelium can mount a maladaptive immune response which can delay viral clearance, perpetuate excessive inflammation, and contribute to the pathogenesis of severe COVID-19. In this article, we will review the barrier and immune functions of the airway epithelium, how SARS-CoV-2 can interact with the epithelium, and epithelial-derived cytokines and chemokines and their roles in COVID-19 and as biomarkers. Finally, we will discuss these immune mediators and their potential as therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Tony J. F. Guo
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
| | - Gurpreet K. Singhera
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Janice M. Leung
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Delbert R. Dorscheid
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
8
|
Balistrieri A, Makino A, Yuan JXJ. Pathophysiology and pathogenic mechanisms of pulmonary hypertension: role of membrane receptors, ion channels, and Ca 2+ signaling. Physiol Rev 2023; 103:1827-1897. [PMID: 36422993 PMCID: PMC10110735 DOI: 10.1152/physrev.00030.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/11/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The pulmonary circulation is a low-resistance, low-pressure, and high-compliance system that allows the lungs to receive the entire cardiac output. Pulmonary arterial pressure is a function of cardiac output and pulmonary vascular resistance, and pulmonary vascular resistance is inversely proportional to the fourth power of the intraluminal radius of the pulmonary artery. Therefore, a very small decrease of the pulmonary vascular lumen diameter results in a significant increase in pulmonary vascular resistance and pulmonary arterial pressure. Pulmonary arterial hypertension is a fatal and progressive disease with poor prognosis. Regardless of the initial pathogenic triggers, sustained pulmonary vasoconstriction, concentric vascular remodeling, occlusive intimal lesions, in situ thrombosis, and vascular wall stiffening are the major and direct causes for elevated pulmonary vascular resistance in patients with pulmonary arterial hypertension and other forms of precapillary pulmonary hypertension. In this review, we aim to discuss the basic principles and physiological mechanisms involved in the regulation of lung vascular hemodynamics and pulmonary vascular function, the changes in the pulmonary vasculature that contribute to the increased vascular resistance and arterial pressure, and the pathogenic mechanisms involved in the development and progression of pulmonary hypertension. We focus on reviewing the pathogenic roles of membrane receptors, ion channels, and intracellular Ca2+ signaling in pulmonary vascular smooth muscle cells in the development and progression of pulmonary hypertension.
Collapse
Affiliation(s)
- Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- Harvard University, Cambridge, Massachusetts
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
9
|
Kim Y, Bae CR, Kim D, Kim H, Lee S, Zhang H, Noh M, Kim YM, Mochizuki N, Kwon YG. Efficacy of CU06-1004 via regulation of inflammation and endothelial permeability in LPS-induced acute lung injury. J Inflamm (Lond) 2023; 20:13. [PMID: 37024954 PMCID: PMC10078077 DOI: 10.1186/s12950-023-00338-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a life-threatening condition that fundamentally results from inflammation and edema in the lung. There are no effective treatments available for clinical use. Previously, we found that as a leakage blocker CU06-1004 prevents endothelial barrier disruption and enhances endothelial cell survival under inflammatory conditions. In this study, we aimed to elucidate the effect of CU06-1004 in terms of prevention of inflammation and endothelial dysfunction in an ALI mouse model. METHODS An ALI model was established that included intraperitoneal administration of LPS. Following LPS administration, survival rates and lung wet/dry ratios were assessed. Histological analysis was performed using hematoxylin and eosin staining. Scanning electron microscopy was used to examine alveolar and capillary morphology. Cytokines such as IL-1β, IL-6, and TNF-α were analyzed using an ELISA assay of bronchoalveolar lavage fluid (BALF) and serum. Neutrophil infiltration was observed in BALF using Wright-Giemsa staining, and myeloperoxidase (MPO) activity was assessed. Pulmonary vascular leakage was confirmed using Evans-blue dye, and the expression of junctional proteins was evaluated using immunofluorescent staining. Expression of adhesion molecules was observed using immunofluorescence staining. NF-κB activation was determined using immunohistochemistry and western blot analysis. RESULTS Survival rates and pulmonary edema were ameliorated with CU06-1004 treatment. Administration of CU06-1004 normalized histopathological changes induced by LPS, and alveolar-capillary wall thickening was reduced. Compared with the LPS-challenged group, after CU06-1004 treatment, the infiltration of immune cells was decreased in the BALF, and MPO activity in lung tissue was reduced. Similarly, in the CU06-1004 treatment group, pro-inflammatory cytokines were significantly inhibited in both BALF and serum. Evans-blue leakage was reduced, and the expression of junctional proteins was recovered in the CU06-1004 group. Adhesion molecules were downregulated and NF-κB activation was inhibited after CU06-1004 treatment. CONCLUSIONS These results suggested that CU06-1004 had a therapeutic effect against LPS-induced ALI via alleviation of the inflammatory response and protection of vascular integrity.
Collapse
Affiliation(s)
- Yeomyeong Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Cho-Rong Bae
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Dongyeop Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyejeong Kim
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Sunghye Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Haiying Zhang
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Minyoung Noh
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe- shimmachi, Suita, Osaka, 564-8565, Japan
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
10
|
Wu Y, Song X, Li P, Wang Z, Zhao Z, Zhang T. Highly pathogenic porcine reproductive and respiratory syndrome virus-induced inflammatory response in porcine pulmonary microvascular endothelial cells and effects of herbal ingredients on main inflammatory molecules. Int Immunopharmacol 2023; 118:110012. [PMID: 36958210 DOI: 10.1016/j.intimp.2023.110012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/25/2023]
Abstract
The role of microvascular endothelial cells (MVECs) in viral infection has received increasing attention. Our previous study demonstrated the susceptibility of porcine pulmonary MVECs to highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV), while their responses to the viral infection remain unclear. This study aimed to understand effects of the HP-PRRSV infection on functions of porcine pulmonary MVECs and the intervention effects of Chinese herbal ingredients on them. Highly purified porcine pulmonary MVECs were separated using CD31-immunomagnetic beads and infected with HP-PRRSV JXA1 and HN strain. The virus particles in cells and the ultrastructural pathological changes of cells were revealed by transmission electron microscopy. High-throughput transcriptome sequencing indicated that 104 and 228 genes were differentially expressed at 36 h post-infection, respectively, including many inflammatory molecules such as interleukins, chemokines, and adhesion molecules. The expression kinetics of HP-PRRSV-induced IL-1α, IL-6, IL-8, and VCAM-1 were characterized at the mRNA and protein levels. Luteolin significantly down-regulated HP-PRRSV-induced increase of the four molecules at both levels, and glycyrrhetinic acid and baicalin reduced that of IL-6 and VCAM-1. Our results suggest that porcine pulmonary MVECs play important roles in the inflammatory lung injury caused by HP-PRRSV infection and that herbal ingredients have potential regulatory effects on the HP-PRRSV-induced dysfunction of MVECs.
Collapse
Affiliation(s)
- Yanmei Wu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Xiaoxiao Song
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Peishan Li
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Zhaoli Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China
| | - Zhanzhong Zhao
- Laboratory of Pharmacobiology, Institute of Animal Science, Chinese Academy of Agricultural Science, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Tao Zhang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, No. 7 Beinong Road, Beijing 102206, China.
| |
Collapse
|
11
|
Treating COVID-19: Targeting the Host Response, Not the Virus. Life (Basel) 2023; 13:life13030712. [PMID: 36983871 PMCID: PMC10054780 DOI: 10.3390/life13030712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/19/2023] [Accepted: 01/31/2023] [Indexed: 03/09/2023] Open
Abstract
In low- and middle-income countries (LMICs), inexpensive generic drugs like statins, ACE inhibitors, and ARBs, especially if used in combination, might be the only practical way to save the lives of patients with severe COVID-19. These drugs will already be available in all countries on the first pandemic day. Because they target the host response to infection instead of the virus, they could be used to save lives during any pandemic. Observational studies show that inpatient statin treatment reduces 28–30-day mortality but randomized controlled trials have failed to show this benefit. Combination treatment has been tested for antivirals and dexamethasone but, with the exception of one observational study in Belgium, not for inexpensive generic drugs. Future pandemic research must include testing combination generic drug treatments that could be used in LMICs.
Collapse
|
12
|
Konijnenberg LSF, Luiken TTJ, Veltien A, Uthman L, Kuster CTA, Rodwell L, de Waard GA, Kea-Te Lindert M, Akiva A, Thijssen DHJ, Nijveldt R, van Royen N. Imatinib attenuates reperfusion injury in a rat model of acute myocardial infarction. Basic Res Cardiol 2023; 118:2. [PMID: 36639597 PMCID: PMC9839396 DOI: 10.1007/s00395-022-00974-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023]
Abstract
Following an acute myocardial infarction, reperfusion of an occluded coronary artery is often accompanied by microvascular injury, leading to worse long-term prognosis. Experimental studies have revealed the potential of tyrosine-kinase inhibitor imatinib to reduce vascular leakage in various organs. Here, we examined the potential of imatinib to attenuate microvascular injury in a rat model of myocardial reperfusion injury. Isolated male Wistar rat hearts (n = 20) in a Langendorff system and male Wistar rats (n = 37) in an in vivo model were randomly assigned to imatinib or placebo and subjected to ischaemia and reperfusion. Evans-blue/Thioflavin-S/TTC staining and Cardiac Magnetic Resonance Imaging were performed to assess the extent of reperfusion injury. Subsequently, in vivo hearts were perfused ex vivo with a vascular leakage tracer and fluorescence and electron microscopy were performed. In isolated rat hearts, imatinib reduced global infarct size, improved end-diastolic pressure, and improved rate pressure product recovery compared to placebo. In vivo, imatinib reduced no-reflow and infarct size with no difference between imatinib and placebo for global cardiac function. In addition, imatinib showed lower vascular resistance, higher coronary flow, and less microvascular leakage in the affected myocardium. At the ultrastructural level, imatinib showed higher preserved microvascular integrity compared to placebo. We provide evidence that low-dose imatinib can reduce microvascular injury and accompanying myocardial infarct size in a rat model of acute myocardial infarction. These data warrant future work to examine the potential of imatinib to reduce reperfusion injury in patients with acute myocardial infarction.
Collapse
Affiliation(s)
- Lara S F Konijnenberg
- Department of Cardiology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Tom T J Luiken
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andor Veltien
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Laween Uthman
- Department of Cardiology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carolien T A Kuster
- Department of Cardiology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Laura Rodwell
- Department of Epidemiology and Biostatistics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Guus A de Waard
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Mariska Kea-Te Lindert
- Department of Cell Biology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Electron Microscopy Center, Radboudumc Technology Center Microscopy, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Biochemistry, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anat Akiva
- Electron Microscopy Center, Radboudumc Technology Center Microscopy, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Biochemistry, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robin Nijveldt
- Department of Cardiology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
13
|
Athale J, Gallagher J, Busch LM. Management of Severe and Critical COVID-19 Infection with Immunotherapies. Infect Dis Clin North Am 2022; 36:761-775. [PMID: 36328635 PMCID: PMC9293954 DOI: 10.1016/j.idc.2022.07.002] [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] [Indexed: 01/31/2023]
Abstract
Following the reduction in mortality demonstrated by dexamethasone treatment in severe COVID-19, many targeted immunotherapies have been investigated. Thus far, inhibition of IL-6 and JAK pathways have the most robust data and have been granted Emergency Use Authorization for treatment of severe disease. However, it must be noted that critically ill patients comprised a relatively small proportion of most of the trials of COVID-19 therapeutics, despite bearing a disproportionate burden of morbidity and mortality. Furthermore, the rapidity and fluidity with which clinical trials have been conducted in the pandemic setting have contributed to difficulty in extrapolating available trial data to critically ill patients. The exclusion of many patients requiring invasive mechanical ventilation, preponderance of ordinal scale based endpoints, and frequent lack of blinding are particular challenges. More data is needed to identify beneficial treatments in the complex milieu of critical illness from COVID-19 infection.
Collapse
Affiliation(s)
- Janhavi Athale
- Critical Care Medicine Department, Mayo Clinic, Phoenix, AZ, USA
| | - Jolie Gallagher
- Department of Pharmacy, Emory University Hospital, Atlanta, GA, USA
| | - Lindsay M Busch
- Division of Infectious Diseases, Emory University School of Medicine, 101 Woodruff Memorial Building, Suite 2101, Atlanta, GA 30322, USA; Emory Critical Care Center, Atlanta, GA, USA.
| |
Collapse
|
14
|
Lean FZX, Núñez A, Spiro S, Priestnall SL, Vreman S, Bailey D, James J, Wrigglesworth E, Suarez-Bonnet A, Conceicao C, Thakur N, Byrne AMP, Ackroyd S, Delahay RJ, van der Poel WHM, Brown IH, Fooks AR, Brookes SM. Differential susceptibility of SARS-CoV-2 in animals: Evidence of ACE2 host receptor distribution in companion animals, livestock and wildlife by immunohistochemical characterisation. Transbound Emerg Dis 2022; 69:2275-2286. [PMID: 34245662 PMCID: PMC8447087 DOI: 10.1111/tbed.14232] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022]
Abstract
Angiotensin converting enzyme 2 (ACE2) is a host cell membrane protein (receptor) that mediates the binding of coronavirus, most notably SARS coronaviruses in the respiratory and gastrointestinal tracts. Although SARS-CoV-2 infection is mainly confined to humans, there have been numerous incidents of spillback (reverse zoonoses) to domestic and captive animals. An absence of information on the spatial distribution of ACE2 in animal tissues limits our understanding of host species susceptibility. Here, we describe the distribution of ACE2 using immunohistochemistry (IHC) on histological sections derived from carnivores, ungulates, primates and chiroptera. Comparison of mink (Neovison vison) and ferret (Mustela putorius furo) respiratory tracts showed substantial differences, demonstrating that ACE2 is present in the lower respiratory tract of mink but not ferrets. The presence of ACE2 in the respiratory tract in some species was much more restricted as indicated by limited immunolabelling in the nasal turbinate, trachea and lungs of cats (Felis catus) and only the nasal turbinate in the golden Syrian hamster (Mesocricetus auratus). In the lungs of other species, ACE2 could be detected on the bronchiolar epithelium of the sheep (Ovis aries), cattle (Bos taurus), European badger (Meles meles), cheetah (Acinonyx jubatus), tiger and lion (Panthera spp.). In addition, ACE2 was present in the nasal mucosa epithelium of the serotine bat (Eptesicus serotinus) but not in pig (Sus scrofa domestica), cattle or sheep. In the intestine, ACE2 immunolabelling was seen on the microvillus of enterocytes (surface of intestine) across various taxa. These results provide anatomical evidence of ACE2 expression in a number of species which will enable further understanding of host susceptibility and tissue tropism of ACE2 receptor-mediated viral infection.
Collapse
Affiliation(s)
- Fabian Z X Lean
- Department of Pathology and Animal Sciences, Animal and Plant Health Agency (APHA), Addlestone, Surrey, UK
| | - Alejandro Núñez
- Department of Pathology and Animal Sciences, Animal and Plant Health Agency (APHA), Addlestone, Surrey, UK
| | - Simon Spiro
- Wildlife Health Services, Zoological Society of London, London, UK
| | - Simon L Priestnall
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, North Mymms, UK
| | - Sandra Vreman
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | | | - Joe James
- Department of Virology, APHA, Addlestone, Surrey, UK
| | | | - Alejandro Suarez-Bonnet
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, North Mymms, UK
| | | | | | | | - Stuart Ackroyd
- Department of Pathology and Animal Sciences, Animal and Plant Health Agency (APHA), Addlestone, Surrey, UK
| | | | | | - Ian H Brown
- Department of Virology, APHA, Addlestone, Surrey, UK
| | | | | |
Collapse
|
15
|
Xue Y, Liu C, Andrews G, Wang J, Ge Y. Recent advances in carbon quantum dots for virus detection, as well as inhibition and treatment of viral infection. NANO CONVERGENCE 2022; 9:15. [PMID: 35366117 PMCID: PMC8976173 DOI: 10.1186/s40580-022-00307-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/17/2022] [Indexed: 05/28/2023]
Abstract
In the last decade, carbon quantum dots (CQDs), as a novel class of carbon-based nanomaterials, have received increasing attention due to their distinct properties. CQDs are ultimately small nanoparticles with an average size below 10 nm, possessing high water solubility, alluring photoluminescence, photostability, excellent biocompatibility, low/none toxicity, environmental friendliness, and high sustainability, etc. In history, there are intermittent threats from viruses to humans, animals and plants worldwide, resulting in enormous crises and impacts on our life, environment, economy and society. Some recent studies have unveiled that certain types of CQDs exhibited high and potent antiviral activities against various viruses such as human coronavirus, arterivirus, norovirus and herpesvirus. Moreover, they have been successfully explored and developed for different virus detections including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This article exclusively overviews and discusses the recent progress of designing, synthesizing, modifying/functionalizing and developing CQDs towards effective virus detection as well as the inhibition and treatment of viral infection. Their mechanisms and applications against various pathogenic viruses are addressed. The latest outcomes for combating the coronavirus disease 2019 (COVID-19) utilizing CQDs are also highlighted. It can be envisaged that CQDs could further benefit the development of virus detectors and antiviral agents with added broad-spectrum activity and cost-effective production.
Collapse
Affiliation(s)
- Yuxiang Xue
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, EH9 3HL, UK
| | - Chenchen Liu
- Department of Metabolism, Digestion and Reproductive, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Gavin Andrews
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, UK
| | - Jinyan Wang
- College of Basic Medical Science, China Medical University, Shenyang, 110122, China
| | - Yi Ge
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, UK.
| |
Collapse
|
16
|
Bernal-Bello D, Morales-Ortega A, Isabel Farfán-Sedano A, de Tena JG, Martín-López JVS. Imatinib in COVID-19: hope and caution. THE LANCET RESPIRATORY MEDICINE 2021; 9:938-939. [PMID: 34147143 PMCID: PMC8409974 DOI: 10.1016/s2213-2600(21)00266-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/19/2022]
Affiliation(s)
- David Bernal-Bello
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid 28942, Spain.
| | - Alejandro Morales-Ortega
- Department of Internal Medicine, Hospital Universitario de Fuenlabrada, Madrid 28942, Spain; Department of Medicine, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | | | | | | |
Collapse
|
17
|
Abstract
Increased mortality in COVID-19 cases is often associated with microvascular complications. We have recently shown that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein promotes an inflammatory cytokine interleukin 6 (IL-6)/IL-6R-induced trans signaling response and alarmin secretion. Virus-infected or spike-transfected human epithelial cells exhibited an increase in senescence, with a release of senescence-associated secretory phenotype (SASP)-related inflammatory molecules. Introduction of the bromodomain-containing protein 4 (BRD4) inhibitor AZD5153 to senescent epithelial cells reversed this effect and reduced SASP-related inflammatory molecule release in TMNK-1 or EAhy926 (representative human endothelial cell lines), when cells were exposed to cell culture medium (CM) derived from A549 cells expressing SARS-CoV-2 spike protein. Cells also exhibited a senescence phenotype with enhanced p16, p21, and senescence-associated β-galactosidase (SA-β-Gal) expression and triggered SASP pathways. Inhibition of IL-6 trans signaling by tocilizumab and inhibition of inflammatory receptor signaling by the Bruton’s tyrosine kinase (BTK) inhibitor zanubrutinib, prior to exposure of CM to endothelial cells, inhibited p21 and p16 induction. We also observed an increase in reactive oxygen species (ROS) in A549 spike-transfected and endothelial cells exposed to spike-transfected CM. ROS generation in endothelial cell lines was reduced after treatment with tocilizumab and zanubrutinib. Cellular senescence was associated with an increased level of the endothelial adhesion molecules vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1), which have in vitro leukocyte attachment potential. Inhibition of senescence or SASP function prevented VCAM-1/ICAM-1 expression and leukocyte attachment. Taken together, we identified that human endothelial cells exposed to cell culture supernatant derived from SARS-CoV-2 spike protein expression displayed cellular senescence markers, leading to enhanced leukocyte adhesion. IMPORTANCE The present study was aimed at examining the underlying mechanism of extrapulmonary manifestations of SARS-CoV-2 spike protein-associated pathogenesis, with the notion that infection of the pulmonary epithelium can lead to mediators that drive endothelial dysfunction. We utilized SARS-CoV-2 spike protein expression in cultured human hepatocytes (Huh7.5) and pneumocytes (A549) to generate conditioned culture medium (CM). Endothelial cell lines (TMNK-1 or EAhy926) treated with CM exhibited an increase in cellular senescence markers by a paracrine mode and led to leukocyte adhesion. Overall, the link between these responses in endothelial cell senescence and a potential contribution to microvascular complication in productively SARS-CoV-2-infected humans is implicated. Furthermore, the use of inhibitors (BTK, IL-6, and BRD4) showed a reverse effect in the senescent cells. These results may support the selection of potential adjunct therapeutic modalities to impede SARS-CoV-2-associated pathogenesis.
Collapse
|