1
|
Mishra V, Agrawal S, Malik D, Mishra D, Bhavya B, Pathak E, Mishra R. Targeting Matrix Metalloproteinase-1, Matrix Metalloproteinase-7, and Serine Protease Inhibitor E1: Implications in preserving lung vascular endothelial integrity and immune modulation in COVID-19. Int J Biol Macromol 2025; 306:141602. [PMID: 40024412 DOI: 10.1016/j.ijbiomac.2025.141602] [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: 01/03/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND SARS-CoV-2 disrupts lung vascular endothelial integrity, contributing to severe COVID-19 complications. However, the molecular mechanisms driving endothelial dysfunction remain underexplored, and targeted therapeutic strategies are lacking. OBJECTIVE This study investigates Naringenin-7-O-glucoside (N7G) as a multi-target therapeutic candidate for modulating vascular integrity and immune response by inhibiting MMP1, MMP7, and SERPINE1-key regulators of extracellular matrix (ECM) remodeling and inflammation. METHODS & RESULTS RNA-seq analysis of COVID-19 lung tissues identified 17 upregulated N7G targets, including MMP1, MMP7, and SERPINE1, with the latter exhibiting the highest expression. PPI network analysis linked these targets to ECM degradation, IL-17, HIF-1, and AGE-RAGE signaling pathways, and endothelial dysfunction. Disease enrichment associated these genes with idiopathic pulmonary fibrosis and asthma. Molecular docking, 200 ns MD simulations (triplicate), and MMGBSA calculations confirmed N7G's stable binding affinity to MMP1, MMP7, and SERPINE1. Immune profiling revealed increased neutrophils and activated CD4+ T cells, alongside reduced mast cells, NK cells, and naïve B cells, indicating immune dysregulation. Correlation analysis linked MMP1, MMP7, and SERPINE1 to distinct immune cell populations, supporting N7G's immunomodulatory role. CONCLUSION These findings suggest that N7G exhibits multi-target therapeutic potential by modulating vascular integrity, ECM remodeling, and immune dysregulation, positioning it as a promising candidate for mitigating COVID-19-associated endothelial dysfunction.
Collapse
Affiliation(s)
- Vibha Mishra
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Shivangi Agrawal
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Divya Malik
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Divya Mishra
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Bhavya Bhavya
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Ekta Pathak
- Institute of Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Rajeev Mishra
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India.
| |
Collapse
|
2
|
Jiang J, Wu Q, Rajasekaran S, Wu R. MMP3 at the crossroads: Linking molecular pathways to disease diagnosis and therapy. Pharmacol Res 2025; 216:107750. [PMID: 40311957 DOI: 10.1016/j.phrs.2025.107750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/03/2025]
Abstract
Matrix metalloproteinase 3 (MMP-3) is a multifaceted enzyme that plays a critical role in the regulation of extracellular matrix (ECM) dynamics, influencing both normal physiological and pathological processes. In addition to its established role in ECM degradation, MMP-3 is gaining recognition for modulating cellular behaviors such as inflammation, migration, and proliferation. Recent research has uncovered its capacity to activate latent signaling molecules, release growth factors from the ECM and interact with various cell surface receptors, linking MMP-3 to the progression of various diseases, including inflammatory diseases, infection diseases, cardiovascular diseases, neurodegenerative disorders, and cancer. The review provides an overview of MMP-3's molecular regulation, emphasizing the mechanisms controlling its expression and activity. We discuss MMP3's involvement in both ECM-dependent and independent pathways, and its potential as a diagnostic, prognostic biomarker in various diseases. Additionally, we explore therapeutic strategies targeting MMP-3, summarizing ongoing efforts to develop specific inhibitors and modulate its activity in different pathologic conditions. Through this review, we aim to consolidate the diverse functions of MMP-3 and provide new insights into future research directions, particularly in translating these findings into clinical applications.
Collapse
Affiliation(s)
- Jing Jiang
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Binzhou Medical University, Yantai, China
| | - Qiong Wu
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Snekha Rajasekaran
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Rongxue Wu
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
3
|
Hong J, Guo Z, Huang X, Wu P, Chen X, Liu X, Yang J, Lai Y. Pharmacological mechanisms of probenecid for SARS-CoV-2 and RSV co-infection: findings of system pharmacology, molecular docking, molecular dynamics simulation, and structure-activity relationship. Front Microbiol 2025; 16:1552603. [PMID: 40371107 PMCID: PMC12075369 DOI: 10.3389/fmicb.2025.1552603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Background The clinical consequences of the co-infection with novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory syncytial virus (RSV) are not optimistic. Nevertheless, there is currently no approved therapeutic regimen specifically targeting SARS-CoV-2/RSV co-infection, with existing monotherapies showing limited efficacy. According to recent studies, probenecid has both anti-SARS-CoV-2 and anti-RSV effects. Therefore, as one probable molecular candidate for the co-infection with SARS-CoV-2 and RSV, probenecid was researched in this exploration. Methods Using systems pharmacology and bioinformatics, we characterized the targets associated with probenecid for the treatment of SARS-CoV-2/RSV co-infection, focusing on their biological functions, mechanisms and binding activities. To further validate these findings, we conducted molecular docking, MD simulations, electrostatic potential mapping, and SAR analysis to explore the binding interactions between probenecid and the identified core targets. Results We identified 141 targets that overlapped with the co-infection and probenecid, and used these shared targets to construct a protein-protein interaction (PPI) network. Subsequently, we obtained the top 16 hub targets of probenecid for SARS-CoV-2/RSV co-infection, namely, AKT1, ALB, EGFR, CASP3, CTNNB1, SRC, HSP90AA1, and so on. According to the enrichment analysis, probenecid might affect inflammation, immunity, oxidative stress, and virus defenses; Toll-like receptor, TNF, IL-17, NOD-like receptor, cytokine-cytokine receptor, among others. Additionally, based on molecular docking analysis, probenecid is effectively bound to the targets related to the SARS-CoV-2/RSV co-infection. Meanwhile, according to molecular dynamics (MD) simulations and structure-activity relationship (SAR) analysis, we speculated that SRC and HSP90AA1 are more likely to be the target proteins of probenecid than the other proteins. Conclusion Our findings from systems pharmacology and bioinformatics analysis indicate that immune and inflammatory responses play a pivotal role in the therapeutic effects of probenecid. Infectious disease-related pathways also contribute significantly to its effectiveness in treating SARS-CoV-2/RSV co-infection. Further validation was conducted through molecular docking, MD simulations, electrostatic potential mapping, and SAR analysis. These analyses suggest that SRC and HSP90AA1 are the potential binding targets of probenecid. This study provides valuable preliminary insights into the molecular mechanisms of probenecid. It establishes a strong foundation for future research to explore its potential as a therapeutic strategy for SARS-CoV-2/RSV co-infection.
Collapse
Affiliation(s)
- Junbin Hong
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhendong Guo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - XiaoMei Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinying Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyi Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinghua Yang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanni Lai
- School of Medicine and Health, Shunde Polytechnic, Foshan, China
| |
Collapse
|
4
|
Giannattasio A, Castaldo A, Grieco M, Gelzo M, Cernera G, Castaldo G, Tipo V. Serum HDL and their subfractions are impaired in multisystem inflammatory syndrome in children (MIS-C). J Transl Med 2025; 23:99. [PMID: 39838487 PMCID: PMC11752960 DOI: 10.1186/s12967-025-06123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Multisystem inflammatory syndrome in children (MIS-C) is a severe post-COVID condition due to a delayed hyperimmune response to SARS-CoV-2. High-density lipoproteins (HDL) are pivotal players in inflammatory and immune modulation through the remodeling of their subfractions. METHODS This study aimed to evaluate serum levels of cholesterol, HDL, and HDL subfractions (HDL-SUB) to define their role in the pathogenesis of MIS-C and their potential use as biomarkers of this condition. We analyzed serum cholesterol, HDL and HDL-SUB (by capillary electrophoresis) in relation to serum values of biomarkers of inflammation and endothelial damage (by microfluidic immunoassays) in 48 patients with MIS-C at hospital admission and in 48 age- and sex-matched healthy controls. RESULTS Serum cholesterol, as well as HDL, were significantly lower in MIS-C patients than controls. Serum cholesterol was inversely correlated with all biomarkers of inflammation, confirming the impact of cytokines on reverse cholesterol transport, whereas HDL values were inversely correlated with serum biomarkers of endothelial damage, suggesting a role of HDL in endothelial damage in MIS-C patients. Furthermore, we found a remodeling of HDL-SUB with a more pronounced decrease in small HDL that have anti-inflammatory activity. CONCLUSIONS These data confirm the severe impairment of reverse cholesterol transport in MIS-C and indicate serum HDL and HDL-SUB as potential useful diagnostic biomarkers of MIS-C.
Collapse
Affiliation(s)
| | - Alice Castaldo
- SC di Pneumologia e UTSIR, AORN Santobono-Pausilipon, Naples, Italy
- Dipartimento di Scienze Mediche Traslazionali, Sezione di Pediatria, Università di Napoli Federico II, Naples, Italy
| | - Michela Grieco
- Dipartimento di Pediatria Generale e d'Urgenza, AORN Santobono-Pausilipon, Naples, Italy
| | - Monica Gelzo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Via Pansini 5, 80131, Naples, Italy.
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
| | - Gustavo Cernera
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Via Pansini 5, 80131, Naples, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
| | - Giuseppe Castaldo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Via Pansini 5, 80131, Naples, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
| | - Vincenzo Tipo
- Dipartimento di Pediatria Generale e d'Urgenza, AORN Santobono-Pausilipon, Naples, Italy
| |
Collapse
|
5
|
Zhang S, Wang N, Ma H, Jing L. A stable rat model of high altitude pulmonary edema established by hypobaric hypoxia combined diurnal temperature fluctuation and exercise. Biochem Biophys Res Commun 2025; 744:151193. [PMID: 39706055 DOI: 10.1016/j.bbrc.2024.151193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/20/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Hypobaric hypoxia (HH) is regarded as the main cause of high-altitude pulmonary edema (HAPE), however, the effect of diurnal temperature fluctuation and exercise has been overlooked. The aim of current study was to elucidate the role of diurnal temperature fluctuation and exercise in the development of HAPE and establish a reliable experimental rat model. Male SPF Wistar rats were assigned to control group (1400 m, 25 °C) and five model groups: Model Ⅰ group (6000 m, 25 °C), Model Ⅱ group (6000 m, 2 °C), Model Ⅲ group (6000 m, 12 °C/2 °C light/dark cycle), Model IV group (6000 m, 2 °C, and exercise) and Model V group (6000 m, 12 °C/2 °C light/dark cycle, and exercise). After exposure for 72 h, the blood and lung tissues were collected for further research. The rats in Model I group did not show signs of HAPE. Compared with Model I group, the rats in Model II and Model III groups were suffered from more damage, evidence by enhanced oxidative stress and inflammatory reaction, but still did not show signs of HAPE. Model IV and Model V could induce HAPE, display the obvious pathological changes and edema, more serious oxidative stress and inflammatory reaction in lung tissues, suggesting that the key role of exercise in the development of HAPE. The rats in the Model V group showed the best performance in terms of modeling indicators, indicating that diurnal temperature fluctuation could further aggravate the degree of lung edema. In summary, HH combined with diurnal temperature fluctuation and exercise is a stable and reliable modeling method for HAPE, which can be used for subsequent research on the prevention and treatment of HAPE.
Collapse
Affiliation(s)
- Shuyu Zhang
- Department of Pharmacy, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, 730050, People's Republic of China
| | - Ning Wang
- Department of Pharmacy, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, 730050, People's Republic of China
| | - Huiping Ma
- Department of Pharmacy, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, 730050, People's Republic of China.
| | - Linlin Jing
- Department of Pharmacy, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, 730050, People's Republic of China; Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China.
| |
Collapse
|
6
|
Kamdar A, Sykes R, Thomson CR, Mangion K, Ang D, Lee MAW, Van Agtmael T, Berry C. Vascular fibrosis and extracellular matrix remodelling in post-COVID 19 conditions. INFECTIOUS MEDICINE 2024; 3:100147. [PMID: 39649442 PMCID: PMC11621938 DOI: 10.1016/j.imj.2024.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/02/2024] [Accepted: 09/23/2024] [Indexed: 12/10/2024]
Abstract
Causal associations between viral infections and acute myocardial injury are not fully understood, with mechanisms potentially involving direct cardiovascular involvement or systemic inflammation. This review explores plausible mechanisms of vascular fibrosis in patients with post-COVID-19 syndrome, focusing on extracellular matrix remodelling. Despite global attention, significant mechanistic or translational breakthroughs in the management of post-viral syndromes remain limited. No effective pharmacological or non-pharmacological interventions are currently available for patients experiencing persistent symptoms following COVID-19 infection. The substantial expansion of scientific knowledge resulting from collaborative efforts by medical experts, scientists, and government organisations in undertaking COVID-19 research could inform treatment strategies for other post-viral syndromes and respiratory illnesses. There is a critical need for clinical trials to evaluate potential therapeutic candidates, providing evidence to guide treatment decisions for post-COVID-19 syndromes.
Collapse
Affiliation(s)
- Anna Kamdar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Robert Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Cameron R. Thomson
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK
| | - Daniel Ang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Michelle AW Lee
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Tom Van Agtmael
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK
| |
Collapse
|
7
|
Bai H, Zhang S, Huang J, Diao K, Li C, Wang M. Unraveling the pathogenic interplay between SARS-CoV-2 and polycystic ovary syndrome using bioinformatics and experimental validation. Sci Rep 2024; 14:22934. [PMID: 39358491 PMCID: PMC11448505 DOI: 10.1038/s41598-024-74347-y] [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: 05/04/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
The prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) among polycystic ovary syndrome (PCOS) is significantly higher than in the general population. However, the mechanisms underlying this remain obscure. This study aimed to explore the mechanisms by identifying the genetic signature of SARS-CoV-2 infection in PCOS. In the present study, a total of 27 common differentially expressed genes (DEGs) were selected for subsequent analyses. Functional analyses showed that immunity and hormone-related pathways collectively participated in the development and progression of PCOS and SARS-CoV-2 infection. Under these, 7 significant hub genes were identified, including S100A9, MMP9, TLR2, THBD, ITGB2, ICAM1, and CD86 by using the algorithm in Cytoscape. Furthermore, hub gene expression was confirmed in the validation set, PCOS clinical samples, and mouse model. Immune microenvironment analysis with the CIBERSORTx database demonstrated that the hub genes were significantly correlated with T cells, dendritic cells, mast cells, B cells, NK cells, and eosinophils and positively correlated with immune scores. Among the hub genes, S100A9, MMP9, THBD, ITGB2, CD86, and ICAM1 demonstrated potential as possible diagnostic markers for COVID-19 and PCOS. In addition, we established the interaction networks of ovary-specific genes, transcription factors, miRNAs, drugs, and chemical compounds with hub genes with NetworkAnalyst. This work uncovered the common pathogenesis and genetic signature of PCOS and SARS-CoV-2 infection, which might provide a theoretical basis and innovative ideas for further mechanistic research and drug discovery of the comorbidity of the two diseases.
Collapse
Affiliation(s)
- Hai Bai
- Institute of Applied Biotechnology, College of Agronomy and Life Science, Shanxi Datong University, Datong, 037009, Shanxi, PR China
| | - Shanshan Zhang
- School of Biological Science, Jining Medical University, Rizhao, 276826, Shandong, PR China
| | - Jing Huang
- Department of Medical Informatics Engineering, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China
| | - Kangyang Diao
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China
| | - Cui Li
- Department of Physiology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China.
- National Experimental Teaching Demonstration Center for Basic Medicine, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China.
| | - Mingming Wang
- Department of Physiology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China.
- National Experimental Teaching Demonstration Center for Basic Medicine, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China.
| |
Collapse
|
8
|
Virtanen MI, Iversen MH, Patel DM, Brinchmann MF. Daily crowding stress has limited, yet detectable effects on skin and head kidney gene expression in surgically tagged atlantic salmon (Salmo salar). FISH & SHELLFISH IMMUNOLOGY 2024; 152:109794. [PMID: 39089638 DOI: 10.1016/j.fsi.2024.109794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/12/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
To ensure welfare-friendly and effective internal tagging, the tagging process should not cause a long-term burden on individuals given that tagged fish serve as representatives for the entire population in telemetry applications. To some extent, stress is inevitable within regular aquaculture practices, and thus, the consequences of long-term stress should be described in terms of their effects on internal tagging. In fish, stressors activate the Hypothalamus-Pituitary-Interrenal (HPI) and Brain-Sympathetic-Chromaffin Cell (BSC) axes, leading to neuroimmunoendocrine communication and paracrine interactions among stress hormones. The interrelation between wound healing and stress is complex, owing to their shared components, pathways, and energy demands. This study assessed 14 genes (mmp9, mmp13, il-2, il-4, il-8a, il-10, il-12, il-17d, il-1b, tnfa, ifng, leg-3, igm, and crh) in the skin (1.5 cm from the wound) and head kidney over eight weeks. These genes, associated with cell signaling in immunity, wound healing, and stress, have previously been identified as influenced and regulated by these processes. Half of a group of Atlantic salmon (n = 90) with surgically implanted dummy smart-tags were exposed to daily crowding stress. The goal was to investigate how this gene panel responds to a wound alone and then to the combined effects of wounding and daily crowding stress. Our observations indicate that chronic stress impacts inflammation and impedes wound healing, as seen through the expression of matrix metalloproteinases genes in the skin but not in the head kidney. This difference is likely due to the ongoing internal wound repair, in contrast to the externally healed wound incision. Cytokine expression, when significant in the skin, was mainly downregulated in both treatments compared to control values, particularly in the study's first half. Conversely, the head kidney showed initial cytokine downregulation followed by upregulation. Across all weeks observed and combining both tissues, the significantly expressed gene differences were 12 % between the Wound and Stress+ groups, 28 % between Wound and Control, and 25 % between Stress+ and Control. Despite significant fluctuations in cytokines, sustained variations across multiple weeks are only evident in a few select genes. Furthermore, Stress+ individuals demonstrated the most cytokine correlations within the head kidney, which may suggest that chronic stress affects cytokine expression. This investigation unveils that the presence of stress and prolonged activation of the HPI axis in an eight weeklong study has limited yet detectable effects on the selected gene expression within immunity, wound healing, and stress, with notable tissue-specific differences.
Collapse
|
9
|
da Silva GB, Manica D, da Silva AP, Valcarenghi E, Donassolo SR, Kosvoski GC, Mingoti MED, Gavioli J, Cassol JV, Hanauer MC, Hellmann MB, Marafon F, Bertollo AG, de Medeiros J, Cortez AD, Réus GZ, de Oliveira GG, Ignácio ZM, Bagatini MD. Peripheral biomarkers as a predictor of poor prognosis in severe cases of COVID-19. Am J Med Sci 2024; 368:122-135. [PMID: 38636654 DOI: 10.1016/j.amjms.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/29/2023] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
We evaluated glycemia and triglyceride, hepatic, muscular, and renal damage markers, redox profile, and leptin and ghrelin hormone levels in COVID-19 patients. We also conducted statistical analysis to verify the potential of biomarkers to predict poor prognosis and the correlation between them in severe cases. We assessed glycemia and the levels of triglycerides, hepatic, muscular, and renal markers in automatized biochemical analyzer. The leptin and ghrelin hormones were assessed by the ELISA assay. Severe cases presented high glycemia and triglyceride levels. Hepatic, muscular, and renal biomarkers were altered in severe patients. Oxidative stress status was found in severe COVID-19 patients. Severe cases also had increased levels of leptin. The ROC curves indicated many biomarkers as poor prognosis predictors in severe cases. The Spearman analysis showed that biomarkers correlate between themselves. Patients with COVID-19 showed significant dysregulation in the levels of several peripheral biomarkers. We bring to light that a robust panel of peripheral biomarkers and hormones predict poor prognosis in severe cases of COVID-19 and biomarkers correlate with each other. Early monitoring of these biomarkers may lead to appropriate clinical interventions in patients infected by SARS-CoV2.
Collapse
Affiliation(s)
- Gilnei B da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Daiane Manica
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, SC, Brazil
| | - Alana P da Silva
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, SC, Brazil
| | - Eduarda Valcarenghi
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Sabine R Donassolo
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Greicy C Kosvoski
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Maiqueli E D Mingoti
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Jullye Gavioli
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Joana V Cassol
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Marceli C Hanauer
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Mariélly B Hellmann
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Filomena Marafon
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Amanda G Bertollo
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Jesiel de Medeiros
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Arthur D Cortez
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Gislaine Z Réus
- Postgraduate Program in Health Sciences, Translational Psychiatry Laboratory, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Gabriela G de Oliveira
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Zuleide M Ignácio
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Margarete D Bagatini
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, SC, Brazil; Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
10
|
Zhang Y, Zhang Z, Mo Y, Zhang Y, Yuan J, Zhang Q. MMP-3 mediates copper oxide nanoparticle-induced pulmonary inflammation and fibrosis. J Nanobiotechnology 2024; 22:428. [PMID: 39030581 PMCID: PMC11264740 DOI: 10.1186/s12951-024-02707-x] [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: 05/02/2024] [Accepted: 07/05/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND The increasing production and usage of copper oxide nanoparticles (Nano-CuO) raise human health concerns. Previous studies have demonstrated that exposure to Nano-CuO could induce lung inflammation, injury, and fibrosis. However, the potential underlying mechanisms are still unclear. Here, we proposed that matrix metalloproteinase-3 (MMP-3) might play an important role in Nano-CuO-induced lung inflammation, injury, and fibrosis. RESULTS Exposure of mice to Nano-CuO caused acute lung inflammation and injury in a dose-dependent manner, which was reflected by increased total cell number, neutrophil count, macrophage count, lactate dehydrogenase (LDH) activity, and CXCL1/KC level in bronchoalveolar lavage fluid (BALF) obtained on day 3 post-exposure. The time-response study showed that Nano-CuO-induced acute lung inflammation and injury appeared as early as day 1 after exposure, peaked on day 3, and ameliorated over time. However, even on day 42 post-exposure, the LDH activity and macrophage count were still higher than those in the control group, suggesting that Nano-CuO caused chronic lung inflammation. The Nano-CuO-induced pulmonary inflammation was further confirmed by H&E staining of lung sections. Trichrome staining showed that Nano-CuO exposure caused pulmonary fibrosis from day 14 to day 42 post-exposure with an increasing tendency over time. Increased hydroxyproline content and expression levels of fibrosis-associated proteins in mouse lungs were also observed. In addition, Nano-CuO exposure induced MMP-3 overexpression and increased MMP-3 secretion in mouse lungs. Knocking down MMP-3 in mouse lungs significantly attenuated Nano-CuO-induced acute and chronic lung inflammation and fibrosis. Moreover, Nano-CuO exposure caused sustained production of cleaved osteopontin (OPN) in mouse lungs, which was also significantly decreased by knocking down MMP-3. CONCLUSIONS Our results demonstrated that short-term Nano-CuO exposure caused acute lung inflammation and injury, while long-term exposure induced chronic pulmonary inflammation and fibrosis. Knocking down MMP-3 significantly ameliorated Nano-CuO-induced pulmonary inflammation, injury, and fibrosis, and also attenuated Nano-CuO-induced cleaved OPN level. Our study suggests that MMP-3 may play important roles in Nano-CuO-induced pulmonary inflammation and fibrosis via cleavage of OPN and may provide a further understanding of the mechanisms underlying Nano-CuO-induced pulmonary toxicity.
Collapse
Affiliation(s)
- Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
- Beijing Key Laboratory of Occupational Safety and Health, Institute of Urban Safety and Environmental Science, Beijing Academy of Science and Technology, Beijing, 100054, China
| | - Zhenyu Zhang
- Department of Emergency, Xiang'An Hospital of Xiamen University, Xiamen, 361104, Fujian, China
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Yue Zhang
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA.
| |
Collapse
|
11
|
Ahmed G, Abdelgadir Y, Abdelghani A, Simpson P, Barbeau J, Basel D, Barrios CS, Smith BA, Schilter KF, Udani R, Reddi HV, Willoughby RE. Reduction in ACE2 expression in peripheral blood mononuclear cells during COVID-19 - implications for post COVID-19 conditions. BMC Infect Dis 2024; 24:663. [PMID: 38956476 PMCID: PMC11221185 DOI: 10.1186/s12879-024-09321-0] [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: 09/20/2023] [Accepted: 04/14/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Severe COVID-19 is uncommon, restricted to 19% of the total population. In response to the first virus wave (alpha variant of SARS-CoV-2), we investigated whether a biomarker indicated severity of disease and, in particular, if variable expression of angiotensin converting enzyme 2 (ACE2) in blood might clarify this difference in risk and of post COVID -19 conditions (PCC). METHODS The IRB-approved study compared patients hospitalized with severe COVID-19 to healthy controls. Severe infection was defined requiring oxygen or increased oxygen need from baseline at admission with positive COVID-19 PCR. A single blood sample was obtained from patients within a day of admission. ACE2 RNA expression in blood cells was measured by an RT-PCR assay. Plasma ACE1 and ACE2 enzyme activities were quantified by fluorescent peptides. Plasma TIMP-1, PIIINP and MMP-9 antigens were quantified by ELISA. Data were entered into REDCap and analyzed using STATA v 14 and GraphPad Prism v 10. RESULTS Forty-eight patients and 72 healthy controls were recruited during the pandemic. ACE2 RNA expression in peripheral blood mononuclear cells (PBMC) was rarely detected acutely during severe COVID-19 but common in controls (OR for undetected ACE2: 12.4 [95% CI: 2.62-76.1]). ACE2 RNA expression in PBMC did not determine plasma ACE1 and ACE2 activity, suggesting alternative cell-signaling pathways. Markers of fibrosis (TIMP-1 and PIIINP) and vasculopathy (MMP-9) were additionally elevated. ACE2 RNA expression during severe COVID-19 often responded within hours to convalescent plasma. Analogous to oncogenesis, we speculate that potent, persistent, cryptic processes following COVID-19 (the renin-angiotensin system (RAS), fibrosis and vasculopathy) initiate or promote post-COVID-19 conditions (PCC) in susceptible individuals. CONCLUSIONS This work elucidates biological and temporal plausibility for ACE2, TIMP1, PIIINP and MMP-9 in the pathogenesis of PCC. Intersection of these independent systems is uncommon and may in part explain the rarity of PCC.
Collapse
Affiliation(s)
- Gulrayz Ahmed
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | - Pippa Simpson
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jody Barbeau
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Donald Basel
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | | | - Rupa Udani
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Honey V Reddi
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Rodney E Willoughby
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
- Pediatric Infectious Diseases, C450, Medical College of Wisconsin, PO Box 1997, Milwaukee, WI 53201-1997, USA.
| |
Collapse
|
12
|
Uno K, Hasan A, Nakayama EE, Rahim R, Harada H, Kaneko M, Hashimoto S, Tanaka T, Matsumoto H, Fujimiya H, Shioda T, Rahman M, Yoshizaki K. Predictive biomarkers of COVID-19 prognosis identified in Bangladesh patients and validated in Japanese cohorts. Sci Rep 2024; 14:12713. [PMID: 38830928 PMCID: PMC11148188 DOI: 10.1038/s41598-024-63184-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Despite high vaccination rates globally, countries are still grappling with new COVID infections, and patients diagnosed as mild dying at home during outpatient treatment. Hence, this study aim to identify, then validate, biomarkers that could predict if newly infected COVID-19 patients would subsequently require hospitalization or could recover safely with medication as outpatients. Serum cytokine/chemokine data from 129 COVID-19 patients within 7 days after the onset of symptoms in Bangladesh were used as training data. The majority of patients were infected with the Omicron variant and over 88% were vaccinated. Patients were divided into those with mild symptoms who recovered, and those who deteriorated to moderate or severe illness. Using the Lasso method, 15 predictive markers were identified and used to classify patients into these two groups. The biomarkers were then validated in a cohort of 194 Covid patients in Japan with a predictive accuracy that exceeded 80% for patients infected with Delta and Omicron variants, and 70% for Wuhan and Alpha variants. In an environment of widespread vaccination, these biomarkers could help medical practitioners determine if newly infected COVID-19 patients will improve and can be managed on an out-patient basis, or if they will deteriorate and require hospitalization.
Collapse
Affiliation(s)
- Kazuko Uno
- IFN and Host-Defense Research Laboratory, Louis Pasteur Center for Medical Research, Kyoto, Kyoto, 606-8225, Japan.
| | - Abu Hasan
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka, 1229, Bangladesh
| | - Emi E Nakayama
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0781, Japan
| | - Rummana Rahim
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka, 1229, Bangladesh
| | | | | | - Shoji Hashimoto
- Osaka Prefectural Hospital Organization Osaka Habikino Medical Center, Habikino, Osaka, 583‑8588, Japan
| | - Toshio Tanaka
- Kinki Central Hospital, Itami, Hyogo, 664-8533, Japan
| | - Hisatake Matsumoto
- Trauma and Acute Critical Care Center, Osaka University, Suita, Osaka, 565‑0871, Japan
| | | | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0781, Japan
| | - Mizanur Rahman
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka, 1229, Bangladesh
| | - Kazuyuki Yoshizaki
- Department of Organic Fine Chemicals, Institute of Scientific and Industry Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
13
|
Toczyłowski K, Lewandowski D, Martonik D, Moniuszko-Malinowska A, Kruszewska E, Parfieniuk-Kowerda A, Flisiak R, Sulik A. Differential Inflammatory Responses in Adult and Pediatric COVID-19 Patients: Implications for Long-Term Consequences and Anti-Inflammatory Treatment. Med Sci Monit 2024; 30:e944052. [PMID: 38816982 PMCID: PMC11149468 DOI: 10.12659/msm.944052] [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: 02/04/2024] [Accepted: 03/21/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND COVID-19 manifests with varying degrees of severity across different age groups; adults typically experience more severe symptoms than children. Matrix metalloproteinases (MMPs), known for their role in tissue remodeling and immune responses, may contribute to the pathophysiological disparities observed between these groups. We sought to delineate differences in serum MMP profiles between adult and pediatric COVID-19 patients, assess the influence of anti-inflammatory treatment on MMP levels, and examine potential implications for long-term consequences. MATERIAL AND METHODS Serum samples from adult and pediatric COVID-19 patients, alongside controls, were analyzed for MMP-1, MMP-2, MMP-3, MMP-7, MMP-8, MMP-9, MMP-10, MMP-12, MMP-13, EMMPRIN, TNF-alpha, TIMP-1, TIMP-2, TIMP-3, and TIMP-4. A subset of adult patients received treatment with glucocorticoids, tocilizumab, and convalescent plasma, and MMP levels were compared with those of untreated patients. RESULTS Elevated levels of MMP-1, MMP-7, TIMP-1, and TIMP-2 were observed in adult and pediatric patients. Adult patients displayed higher concentrations of MMP-3, MMP-8, MMP-9, TNF-alpha, and TIMP-4 than children. Post-treatment reduction in MMP-1, MMP-8, MMP-9 levels was observed, with median decreases from 21% to 70%. MMP-3 and MMP-7 remained largely unchanged, and MMP-2 concentrations increased after treatment. Notably, anti-inflammatory treatment correlated with reduced post-treatment MMP levels, suggesting potential therapeutic benefit. CONCLUSIONS Distinctive inflammatory responses in COVID-19 were evident between adults and children. While certain MMPs exhibited post-treatment reduction, the persistence of elevated levels raises concerns about potential long-term consequences, including lung fibrosis. Our findings emphasize the need for personalized treatment strategies and further investigation into the dynamics of MMP regulation in COVID-19.
Collapse
Affiliation(s)
- Kacper Toczyłowski
- Department of Pediatric Infectious Diseases, Medical University of Białystok, Białystok, Poland
| | - Dawid Lewandowski
- Department of Pediatric Infectious Diseases, Medical University of Białystok, Białystok, Poland
| | - Diana Martonik
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Białystok, Białystok, Poland
| | - Ewelina Kruszewska
- Department of Infectious Diseases and Neuroinfections, Medical University of Białystok, Białystok, Poland
| | - Anna Parfieniuk-Kowerda
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland
| | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland
| | - Artur Sulik
- Department of Pediatric Infectious Diseases, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
14
|
Devaux Y, Zhang L, Lumley AI, Karaduzovic-Hadziabdic K, Mooser V, Rousseau S, Shoaib M, Satagopam V, Adilovic M, Srivastava PK, Emanueli C, Martelli F, Greco S, Badimon L, Padro T, Lustrek M, Scholz M, Rosolowski M, Jordan M, Brandenburger T, Benczik B, Agg B, Ferdinandy P, Vehreschild JJ, Lorenz-Depiereux B, Dörr M, Witzke O, Sanchez G, Kul S, Baker AH, Fagherazzi G, Ollert M, Wereski R, Mills NL, Firat H. Development of a long noncoding RNA-based machine learning model to predict COVID-19 in-hospital mortality. Nat Commun 2024; 15:4259. [PMID: 38769334 PMCID: PMC11106268 DOI: 10.1038/s41467-024-47557-1] [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: 12/11/2023] [Accepted: 04/03/2024] [Indexed: 05/22/2024] Open
Abstract
Tools for predicting COVID-19 outcomes enable personalized healthcare, potentially easing the disease burden. This collaborative study by 15 institutions across Europe aimed to develop a machine learning model for predicting the risk of in-hospital mortality post-SARS-CoV-2 infection. Blood samples and clinical data from 1286 COVID-19 patients collected from 2020 to 2023 across four cohorts in Europe and Canada were analyzed, with 2906 long non-coding RNAs profiled using targeted sequencing. From a discovery cohort combining three European cohorts and 804 patients, age and the long non-coding RNA LEF1-AS1 were identified as predictive features, yielding an AUC of 0.83 (95% CI 0.82-0.84) and a balanced accuracy of 0.78 (95% CI 0.77-0.79) with a feedforward neural network classifier. Validation in an independent Canadian cohort of 482 patients showed consistent performance. Cox regression analysis indicated that higher levels of LEF1-AS1 correlated with reduced mortality risk (age-adjusted hazard ratio 0.54, 95% CI 0.40-0.74). Quantitative PCR validated LEF1-AS1's adaptability to be measured in hospital settings. Here, we demonstrate a promising predictive model for enhancing COVID-19 patient management.
Collapse
Affiliation(s)
- Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg.
| | - Lu Zhang
- Bioinformatics Platform, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Andrew I Lumley
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | | | - Vincent Mooser
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Simon Rousseau
- The Meakins-Christie Laboratories at the Research Institute of the McGill University Heath Centre Research Institute, & Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Muhammad Shoaib
- Luxembourg Center for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - Venkata Satagopam
- Luxembourg Center for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - Muhamed Adilovic
- Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, England, UK
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Lina Badimon
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU); CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU); CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Mitja Lustrek
- Department of Intelligent Systems, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Markus Scholz
- Group Genetical Statistics and Biomathematical Modelling, Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Maciej Rosolowski
- Group Genetical Statistics and Biomathematical Modelling, Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Marko Jordan
- Department of Intelligent Systems, Jozef Stefan Institute, Ljubljana, Slovenia
| | | | - Bettina Benczik
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bence Agg
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Peter Ferdinandy
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Jörg Janne Vehreschild
- Medical Department 2 (Hematology/Oncology and Infectious Diseases), Center for Internal Medicine, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | | | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany; German Centre of Cardiovascular Research (DZHK), Greifswald, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | | | - Andy H Baker
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland
- CARIM Institute and Department of Pathology, University of Maastricht, Maastricht, The Netherlands
| | - Guy Fagherazzi
- Deep Digital Phenotyping Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis (ORCA), University of Southern Denmark, Odense, Denmark
| | - Ryan Wereski
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Nicholas L Mills
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
15
|
Wong M, Gain C, Sharma MB, Fotooh Abadi L, Hugo C, Vassilopoulos H, Daskou M, Fishbein GA, Kelesidis T. Severe Acute Respiratory Syndrome Coronavirus 2 Infection Alters Mediators of Lung Tissue Remodeling In Vitro and In Vivo. J Infect Dis 2024; 229:1372-1381. [PMID: 38109685 DOI: 10.1093/infdis/jiad536] [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] [Accepted: 11/27/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Altered mediators of airway tissue remodeling such as matrix metalloproteinases (MMPs) in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may contribute to morbidity in coronavirus disease 2019 (COVID-19); however, the differential impact of SARS-CoV-2 variants of concern (VOCs) on MMPs is unknown. METHODS Using both in vitro human airway cell culture model and in vivo transgenic mouse model of SARS-CoV-2 infection, we studied the differential effect of SARS-CoV-2 VOCs on expression of key MMPs and inflammatory mediators in airway cells and tissues. RESULTS The most consistent findings with all SARS-CoV-2 variants in infected compared to uninfected human bronchial epithelial cell air-liquid interface cultures were the SARS-CoV-2-induced increases in MMP-12 and tissue inhibitor of MMPs. Infection with both SARS-CoV-2 wild type and SARS-CoV-2 Delta variant over 3 days postinfection (dpi) and with Beta variant over 7 dpi increased lung tissue levels of MMP-9 compared to uninfected mice. Overall, SARS-CoV-2 variants had differential dose-dependent impact on secretion of MMP-1, MMP-2, MMP-9, and MMP-12 that varied at the protein versus the gene level and in the early noninflammatory compared to late inflammatory phase of infection. CONCLUSIONS We provide novel mechanistic insight that the differential impact of SARS-CoV-2 variants on severity of COVID-19 may partially be attributed to unique changes in MMPs.
Collapse
Affiliation(s)
- Michael Wong
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Chandrima Gain
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Madhav B Sharma
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Leila Fotooh Abadi
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
- Division of Infectious Diseases, Department of Medicine, University of Texas Southwestern, Dallas
| | - Cristelle Hugo
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Hariclea Vassilopoulos
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Maria Daskou
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Gregory A Fishbein
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles
| | - Theodoros Kelesidis
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
- Division of Infectious Diseases, Department of Medicine, University of Texas Southwestern, Dallas
| |
Collapse
|
16
|
Samara TD, Wartono M, Kosasih A, Alvina A. Association of Body Mass Index with Matrix Metalloproteinase-9, Tissue Inhibitor of Metalloproteinase-1, and Interleukin-6 Based on Blood Pressure. Rep Biochem Mol Biol 2024; 13:137-143. [PMID: 39582824 PMCID: PMC11580125 DOI: 10.61186/rbmb.13.1.137] [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: 03/08/2024] [Accepted: 06/09/2024] [Indexed: 11/26/2024]
Abstract
Background A high body mass index (BMI) is known to be associated with high blood pressure. Levels of matrix metalloproteinase-9 (MMP-9), tissue inhibitor of metalloproteinase-1 (TIMP-1), and interleukin-6 (IL-6) are also increased in hypertensive patients. The aim of this study was to determine the correlation of BMI with MMP-9, TIMP-1, and IL-6 based on blood pressure. Methods The study design was cross-sectional with subjects aged >= 36 years, male and female and divided into 3 groups: group with normal blood pressure (NBP), group with controlled hypertension (CHT), and group with uncontrolled hypertension (UcHT). Height, weight, and blood pressure were measured, as well as serum levels of MMP-9, TIMP-1 and IL-6 using the ELISA method. The correlation was considered significant at p-value of < 0.05. Results The BMI in group UcHT was higher than in the other groups. There was a positive correlation between BMI and MMP-9; BMI and TIMP-1; and BMI and IL-6 (r=0.480, p=0.007; r=0.620; p=0.000; r=374, p=0.042 respectively) in group UcHT. Conclusions An increase in BMI is accompanied by an increase in levels of MMP-9, TIMP-1, and IL-6 in group UcHT, signifying that it is necessary to control BMI to maintain stable levels of MMP-9, TIMP-1, and IL-6, thereby keeping blood pressure under control.
Collapse
Affiliation(s)
- Tjam Diana Samara
- Department of Anatomy, Faculty of Medicine, Universitas Trisakti, Jakarta, Indonesia.
| | - Magdalena Wartono
- Department of Anatomy, Faculty of Medicine, Universitas Trisakti, Jakarta, Indonesia.
| | - Adrianus Kosasih
- Department of Internal Medicine, Faculty of Medicine, Universitas Trisakti, Jakarta, Indonesia.
| | - Alvina Alvina
- Department of Clinical Pathology, Faculty of Medicine, Universitas Trisakti, Jakarta, Indonesia.
- Laboratory of Sumber Waras Hospital, Jakarta, Indonesia.
| |
Collapse
|
17
|
Takashima Y, Inaba T, Matsuyama T, Yoshii K, Tanaka M, Matsumoto K, Sudo K, Tokuda Y, Omi N, Nakano M, Nakaya T, Fujita N, Sotozono C, Sawa T, Tashiro K, Ohta B. Potential marker subset of blood-circulating cytokines on hematopoietic progenitor-to-Th1 pathway in COVID-19. Front Med (Lausanne) 2024; 11:1319980. [PMID: 38476443 PMCID: PMC10927758 DOI: 10.3389/fmed.2024.1319980] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/31/2024] [Indexed: 03/14/2024] Open
Abstract
In this study, we analyzed a relatively large subset of proteins, including 109 kinds of blood-circulating cytokines, and precisely described a cytokine storm in the expression level and the range of fluctuations during hospitalization for COVID-19. Of the proteins analyzed in COVID-19, approximately 70% were detected with Bonferroni-corrected significant differences in comparison with disease severity, clinical outcome, long-term hospitalization, and disease progression and recovery. Specifically, IP-10, sTNF-R1, sTNF-R2, sCD30, sCD163, HGF, SCYB16, IL-16, MIG, SDF-1, and fractalkine were found to be major components of the COVID-19 cytokine storm. Moreover, the 11 cytokines (i.e., SDF-1, SCYB16, sCD30, IL-11, IL-18, IL-8, IFN-γ, TNF-α, sTNF-R2, M-CSF, and I-309) were associated with the infection, mortality, disease progression and recovery, and long-term hospitalization. Increased expression of these cytokines could be explained in sequential pathways from hematopoietic progenitor cell differentiation to Th1-derived hyperinflammation in COVID-19, which might also develop a novel strategy for COVID-19 therapy with recombinant interleukins and anti-chemokine drugs.
Collapse
Affiliation(s)
- Yasuo Takashima
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tohru Inaba
- Department of Infection Control and Laboratory Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tasuku Matsuyama
- Department of Emergency Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kengo Yoshii
- Department of Mathematics and Statistics in Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masami Tanaka
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazumichi Matsumoto
- Faculty of Clinical Laboratory, University Hospital Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuki Sudo
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuichi Tokuda
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Natsue Omi
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masakazu Nakano
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takaaki Nakaya
- Department of Infectious Diseases, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naohisa Fujita
- Department of Infection Control and Laboratory Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Kyoto Prefectural Institute of Public Health and Environment, Kyoto, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Teiji Sawa
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- University Hospital Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Tashiro
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Bon Ohta
- Department of Emergency Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Kollet O, Das A, Karamanos N, Auf dem Keller U, Sagi I. Redefining metalloproteases specificity through network proteolysis. Trends Mol Med 2024; 30:147-163. [PMID: 38036391 PMCID: PMC11004056 DOI: 10.1016/j.molmed.2023.11.001] [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: 09/09/2023] [Revised: 10/18/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
Proteolytic processes on cell surfaces and extracellular matrix (ECM) sustain cell behavior and tissue integrity in health and disease. Matrix metalloproteases (MMPs) and a disintegrin and metalloproteases (ADAMs) remodel cell microenvironments through irreversible proteolysis of ECM proteins and cell surface bioactive molecules. Pan-MMP inhibitors in inflammation and cancer clinical trials have encountered challenges due to promiscuous activities of MMPs. Systems biology advances revealed that MMPs initiate multifactorial proteolytic cascades, creating new substrates, activating or suppressing other MMPs, and generating signaling molecules. This review highlights the intricate network that underscores the role of MMPs beyond individual substrate-enzyme activities. Gaining insight into MMP function and tissue specificity is crucial for developing effective drug discovery strategies and novel therapeutics. This requires considering the dynamic cellular processes and consequences of network proteolysis.
Collapse
Affiliation(s)
- Orit Kollet
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| | - Alakesh Das
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| | - Nikos Karamanos
- University of Patras, Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, Patras, Greece
| | - Ulrich Auf dem Keller
- Technical University of Denmark, Department of Biotechnology and Biomedicine, Lyngby, Denmark
| | - Irit Sagi
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel.
| |
Collapse
|
19
|
Rajamanickam A, Kumar NP, Venkataraman A, Varadarjan P, Selladurai E, Sankaralingam T, Thiruvengadam K, Selvam R, Thimmaiah A, Natarajan S, Ramaswamy G, Putlibai S, Sadasivam K, Sundaram B, Hissar S, Ranganathan UD, Babu S. Sex-specific differences in systemic immune responses in MIS-C children. Sci Rep 2024; 14:1720. [PMID: 38243064 PMCID: PMC10799056 DOI: 10.1038/s41598-024-52116-1] [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: 06/12/2023] [Accepted: 01/14/2024] [Indexed: 01/21/2024] Open
Abstract
Multisystem Inflammatory Syndrome in Children (MIS-C) is a rare manifestation of Severe Acute Respiratory Syndrome-CoronaVirus-2 (SARS-CoV-2) infection that can result in increased morbidity and mortality. Mounting evidence describes sex disparities in the clinical outcomes of coronavirus disease 2019 (COVID-19). However, there is a lack of information on sex-specific differences in immune responses in MIS-C. This study is an observational and cross-sectional study and we wanted to examine immune parameters such as cytokines, chemokines, acute phase proteins (APPs), growth factors, microbial translocation markers (MTMs), complement components and matrix metalloproteinases (MMPs) in MIS-C children, based on sex. Male children were associated with heightened levels of pro-inflammatory cytokines-IFNγ, IL-2, TNFα, IL-1α, IL-1β, IL-6, IL-12, G-CSF and GM-CSF, chemokines-CCL2, CCL11, CXCL1, CXCL8 and CXCL10, acute phase proteins-α-2M, CRP, growth factors VEGF and TGFα, microbial translocation markers- iFABP, LBP, EndoCAb, complement components-C1q, MBL and C3 and matrix metalloproteinases MMP-8 and MMP-9 compared to female children with MIS-C. These results indicate that the heightened immune response in males is a characteristic feature of MIS-C. These findings might explain the differential disease pathogenesis in males compared to females with MIS-C and facilitate a deeper understanding of this disease.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institutes of Health-National Institute for Research in Tuberculosis - International Center for Excellence in Research, Chennai, India.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Syed Hissar
- National Institute for Research in Tuberculosis, Chennai, India
| | | | - Subash Babu
- National Institutes of Health-National Institute for Research in Tuberculosis - International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
20
|
Sampaio Moura N, Schledwitz A, Alizadeh M, Patil SA, Raufman JP. Matrix metalloproteinases as biomarkers and therapeutic targets in colitis-associated cancer. Front Oncol 2024; 13:1325095. [PMID: 38288108 PMCID: PMC10824561 DOI: 10.3389/fonc.2023.1325095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Colorectal cancer (CRC) remains a major cause of morbidity and mortality. Therapeutic approaches for advanced CRC are limited and rarely provide long-term benefit. Enzymes comprising the 24-member matrix metalloproteinase (MMP) family of zinc- and calcium-dependent endopeptidases are key players in extracellular matrix degradation, a requirement for colon tumor expansion, invasion, and metastasis; hence, MMPs are an important research focus. Compared to sporadic CRC, less is known regarding the molecular mechanisms and the role of MMPs in the development and progression of colitis-associated cancer (CAC) - CRC on a background of chronic inflammatory bowel disease (IBD) - primarily ulcerative colitis and Crohn's disease. Hence, the potential of MMPs as biomarkers and therapeutic targets for CAC is uncertain. Our goal was to review data regarding the role of MMPs in the development and progression of CAC. We sought to identify promising prognostic and therapeutic opportunities and novel lines of investigation. A key observation is that since MMPs may be more active in early phases of CAC, using MMPs as biomarkers of advancing neoplasia and as potential therapeutic targets for adjuvant therapy in those with advanced stage primary CAC rather than overt metastases may yield more favorable outcomes.
Collapse
Affiliation(s)
- Natalia Sampaio Moura
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alyssa Schledwitz
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Madeline Alizadeh
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Seema A. Patil
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
- Medical Service, Veterans Affairs Maryland Healthcare System, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, Baltimore, MD, United States
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
21
|
Dhaliwal M, Muthu V, Sharma A, Raj K, Rudramurthy SM, Agarwal R, Kaur H, Rawat A, Singh S, Chakrabarti A. Immune and metabolic perturbations in COVID-19-associated pulmonary mucormycosis: A transcriptome analysis of innate immune cells. Mycoses 2024; 67:e13679. [PMID: 38214399 DOI: 10.1111/myc.13679] [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: 10/18/2023] [Accepted: 11/23/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND AND OBJECTIVES The mechanisms underlying COVID-19-associated pulmonary mucormycosis (CAPM) remain unclear. We use a transcriptomic analysis of the innate immune cells to investigate the host immune and metabolic response pathways in patients with CAPM. PATIENTS AND METHODS We enrolled subjects with CAPM (n = 5), pulmonary mucormycosis (PM) without COVID-19 (n = 5), COVID-19 (without mucormycosis, n = 5), healthy controls (n = 5) without comorbid illness and negative for SARS-CoV-2. Peripheral blood samples from cases were collected before initiating antifungal therapy, and neutrophils and monocytes were isolated. RNA sequencing was performed using Illumina HiSeqX from monocytes and neutrophils. Raw reads were aligned with HISAT-2 pipeline and DESeq2 was used for differential gene expression. Gene ontology (GO) and metabolic pathway analysis were performed using Shiny GO application and R packages (ggplot2, Pathview). RESULTS The derangement of core immune and metabolic responses in CAPM patients was noted. Pattern recognition receptors, dectin-2, MCL, FcRγ receptors and CLEC-2, were upregulated, but signalling pathways such as JAK-STAT, IL-17 and CARD-9 were downregulated; mTOR and MAP-kinase signalling were elevated in monocytes from CAPM patients. The complement receptors, NETosis, and pro-inflammatory responses, such as S100A8/A9, lipocalin and MMP9, were elevated. The major metabolic pathways of glucose metabolism-glycolysis/gluconeogenesis, pentose phosphate pathway, HIF signalling and iron metabolism-ferroptosis were also upregulated in CAPM. CONCLUSIONS We identified significant alterations in the metabolic pathways possibly leading to cellular iron overload and a hyperglycaemic state. Immune responses revealed altered recognition, signalling, effector functions and a pro-inflammatory state in monocytes and neutrophils from CAPM patients.
Collapse
Affiliation(s)
- Manpreet Dhaliwal
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Valliappan Muthu
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arunima Sharma
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Khem Raj
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Shivaprakash M Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritesh Agarwal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harsimran Kaur
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Surjit Singh
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arunaloke Chakrabarti
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
22
|
Al-Abdulla N, Bakhsh A, Mannocci F, Proctor G, Moyes D, Niazi SA. Successful endodontic treatment reduces serum levels of cardiovascular disease risk biomarkers-high-sensitivity C-reactive protein, asymmetric dimethylarginine, and matrix metalloprotease-2. Int Endod J 2023; 56:1499-1516. [PMID: 37787168 DOI: 10.1111/iej.13979] [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: 03/12/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023]
Abstract
AIM To investigate serum biomarkers of inflammation 2 years following non-surgical root canal re-treatment (Re-RCT) and peri-apical surgery (PS). The results were correlated with signs and symptoms, treatment outcome, metabolic syndrome factors, infection with severe acute respiratory syndrome coronavirus 2 SARS-CoV-2 (COVID-19) infection and COVID-19 vaccination. METHODOLOGY Subjects from our previous study were recalled for 2 years post-treatment follow-up. Changes to the patient's history (medical, dental, social) were noted. Periapical health of the treated teeth was examined both clinically and radiographically. Blood pressure, fasting HbA1C and low-density lipoprotein (LDL), high-density lipoprotein (HDL), triglycerides and total cholesterol (TC) levels were measured. Serum inflammatory marker levels were assayed using a Bio-Rad Bio-Plex 200 analyser and values at different time points within the same group were compared using a Wilcoxon signed-rank test and differences between groups with a Mann-Whitney test. Linear associations were tested using Pearson's correlations. RESULTS The recall percentage at 2 years was 56.9% (n = 37), with a 100% radiographic success rate using periapical radiographs. In total, 21 cases (56.8%) were completely healed, and 16 cases (43.2%) were healing. Higher matrix metalloprotease 2 (MMP2) levels were present in the healing group compared to the healed group. Serum levels of high-sensitivity C-reactive protein (hs-CRP), asymmetric dimethylarginine (ADMA) and MMP-2 were significantly reduced (p ≤ .001) whereas other biomarkers showed significant increases at 2 year compared to pre-operative levels, while FGF-23 and ICAM-1 were not significantly increased. HbA1C (p = .015), TC (p = .003), LDL (p = .003) and HDL (p = .003) reduced significantly at 2 years post-treatment compared to their preoperative levels. COVID infection showed a significant association with MMP-9 (p = .048). CONCLUSIONS hs-CRP, ADMA and MMP-2 can be regarded as prognostic biomarkers of successful Re-RCT and PS as they reduced at 2 year recall in cases which showed evidence of clinical and radiographic success. The successful treatment of chronic apical periodontitis is correlated with improvements in metabolic syndrome indicators, better glycemic control, and reduction at 2 year of some systemic inflammatory markers which are related to risks of cardiovascular disease events.
Collapse
Affiliation(s)
- Noor Al-Abdulla
- Department of Endodontics, Centre of Oral Clinical & Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, Guy's Dental Hospital, King's College London, London, UK
| | - Abdulaziz Bakhsh
- Department of Endodontics, Centre of Oral Clinical & Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, Guy's Dental Hospital, King's College London, London, UK
- Department of Restorative Dentistry, Division of Endodontics, Faculty of Dental Medicine, Umm Al-Qura University, Makkah, Kingdom of Saudi Arabia
| | - Francesco Mannocci
- Department of Endodontics, Centre of Oral Clinical & Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, Guy's Dental Hospital, King's College London, London, UK
| | - Gordon Proctor
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, Guy's Dental Hospital, King's College London, London, UK
| | - David Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, Guy's Dental Hospital, King's College London, London, UK
| | - Sadia Ambreen Niazi
- Department of Endodontics, Centre of Oral Clinical & Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, Guy's Dental Hospital, King's College London, London, UK
| |
Collapse
|
23
|
Ding L, Guo H, Zhang C, Jin H, Guo X, Li T. Elevated matrix metalloproteinase‑9 expression is associated with COVID‑19 severity: A meta‑analysis. Exp Ther Med 2023; 26:545. [PMID: 37928509 PMCID: PMC10623216 DOI: 10.3892/etm.2023.12244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/05/2023] [Indexed: 11/07/2023] Open
Abstract
The present meta-analysis investigated the clinical value of serum matrix metalloproteinase (MMP)-9 levels in Coronavirus Disease 2019 (COVID-19) patients. Studies assessing the outcomes of patients with COVID-19 in correlation with the MMP-9 levels were retrieved from PubMed, Web of Science, EMBASE, Cochrane, WANFANG, and CNKI. A meta-analysis was performed to compare the serum MMP-9 levels between different patient groups: Severe vs. non-severe; acute respiratory distress syndrome (ARDS) vs. non-ARDS; non-survivors vs. survivors; neurologic syndrome vs. non-neurologic syndrome; and obese diabetic vs. non-obese diabetic. A total of 2,062 COVID-19-confirmed patients from 12 studies were included in this meta-analysis. The serum MMP-9 levels were significantly higher in patients with severe COVID-19 than in those with non-severe COVID-19 [weighted mean difference (WMD) 246.61 (95% confidence interval (CI), 115.86-377.36), P<0.001]. Patients with ARDS exhibited significantly higher MMP-9 levels than those without ARDS [WMD 248.55 (95% CI, 63.84-433.25), P<0.001]. The MMP-9 levels in the non-survivors did not significantly differ from those in the survivors [WMD 37.79 (95% CI, -18.08-93.65), P=0.185]. Patients with comorbidities, including neurological syndromes, and obese diabetic patients had significantly higher MMP-9 levels than those without comorbidities [WMD 170.73 (95% CI, 95.61-245.85), P<0.001]. Serum MMP-9 levels were associated with COVID-19 severity and may serve as a therapeutic target for improving the prognosis of patients with COVID-19.
Collapse
Affiliation(s)
- Ling Ding
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161005, P.R. China
- Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang 161005, P.R. China
| | - Haipeng Guo
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161005, P.R. China
- Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang 161005, P.R. China
| | - Chao Zhang
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161005, P.R. China
- Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang 161005, P.R. China
| | - Huixin Jin
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161005, P.R. China
- Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang 161005, P.R. China
| | - Xuyang Guo
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161005, P.R. China
- Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang 161005, P.R. China
| | - Tong Li
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161005, P.R. China
- Department of Laboratory Medicine, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang 161005, P.R. China
| |
Collapse
|
24
|
Murphy SL, Halvorsen B, Holter JC, Huse C, Tveita A, Trøseid M, Hoel H, Kildal AB, Holten AR, Lerum TV, Skjønsberg OH, Michelsen AE, Aaløkken TM, Tonby K, Lind A, Dudman S, Granerud BK, Heggelund L, Bøe S, Dyrholt-Riise AM, Aukrust P, Barratt-Due A, Ueland T, Dahl TB. Circulating markers of extracellular matrix remodelling in severe COVID-19 patients. J Intern Med 2023; 294:784-797. [PMID: 37718572 DOI: 10.1111/joim.13725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
BACKGROUND Abnormal remodelling of the extracellular matrix (ECM) has generally been linked to pulmonary inflammation and fibrosis and may also play a role in the pathogenesis of severe COVID-19. To further elucidate the role of ECM remodelling and excessive fibrogenesis in severe COVID-19, we examined circulating levels of mediators involved in various aspects of these processes in COVID-19 patients. METHODS Serial blood samples were obtained from two cohorts of hospitalised COVID-19 patients (n = 414). Circulating levels of ECM remodelling mediators were quantified by enzyme immunoassays in samples collected during hospitalisation and at 3-month follow-up. Samples were related to disease severity (respiratory failure and/or treatment at the intensive care unit), 60-day total mortality and pulmonary pathology after 3-months. We also evaluated the direct effect of inactivated SARS-CoV-2 on the release of the different ECM mediators in relevant cell lines. RESULTS Several of the measured markers were associated with adverse outcomes, notably osteopontin (OPN), S100 calcium-binding protein A12 and YKL-40 were associated with disease severity and mortality. High levels of ECM mediators during hospitalisation were associated with computed tomography thorax pathology after 3-months. Some markers (i.e. growth differential factor 15, galectin 3 and matrix metalloproteinase 9) were released from various relevant cell lines (i.e. macrophages and lung cell lines) in vitro after exposure to inactivated SARS-CoV-2 suggesting a direct link between these mediators and the causal agent of COVID-19. CONCLUSION Our findings highlight changes to ECM remodelling and particularly a possible role of OPN, S100A12 and YKL-40 in the pathogenesis of severe COVID-19.
Collapse
Affiliation(s)
- Sarah Louise Murphy
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jan Cato Holter
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Camilla Huse
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anders Tveita
- Department of Internal Medicine, Baerum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Hedda Hoel
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Internal Medicine, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Anders Benjamin Kildal
- Department of Anesthesiology and Intensive Care, University Hospital of North Norway, Tromsø, Norway
- Department of Clinical Medicine, Faculty of Health Sciences, UIT - The Arctic University of Norway, Tromsø, Norway
| | - Aleksander Rygh Holten
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Acute Medicine, Oslo University Hospital, Oslo, Norway
| | - Tøri Vigeland Lerum
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Pulmonary Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Ole Henning Skjønsberg
- Department of Internal Medicine, Lovisenberg Diaconal Hospital, Oslo, Norway
- Department of Radiology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trond M Aaløkken
- Department of Internal Medicine, Lovisenberg Diaconal Hospital, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Kristian Tonby
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Andreas Lind
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Susanne Dudman
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Beathe Kiland Granerud
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Lars Heggelund
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Simen Bøe
- Department of Anesthesiology and Intensive Care, Hammerfest County Hospital, Hammerfest, Norway
| | - Anne Ma Dyrholt-Riise
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andreas Barratt-Due
- Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, Oslo, Norway
- Department of Anesthesia and Intensive Care Medicine, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Tuva Børresdatter Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
25
|
Scialò F, Cernera G, Esposito S, Pinchera B, Gentile I, Di Domenico M, Bianco A, Pastore L, Amato F, Castaldo G. The MBL2 genotype relates to COVID-19 severity and may help to select the optimal therapy. Clin Chem Lab Med 2023; 61:2143-2149. [PMID: 37313996 DOI: 10.1515/cclm-2023-0183] [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: 02/22/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Sars-CoV-2 acute infection is clinically heterogeneous, ranging from asymptomatic cases to patients with a severe, systemic clinical course. Among the involved factors age and preexisting morbidities play a major role; genetic host susceptibility contributes to modulating the clinical expression and outcome of the disease. Mannose-binding lectin is an acute-phase protein that activates the lectin-complement pathway, promotes opsonophagocytosis and modulates inflammation, and is involved in several bacterial and viral infections in humans. Understanding its role in Sars-CoV-2 infection could help select a better therapy. METHODS We studied MBL2 haplotypes in 419 patients with acute COVID-19 in comparison to the general population and related the haplotypes to clinical and laboratory markers of severity. RESULTS We recorded an enhanced frequency of MBL2 null alleles in patients with severe acute COVID-19. The homozygous null genotypes were significantly more frequent in patients with advanced WHO score 4-7 (OR of about 4) and related to more severe inflammation, neutrophilia, and lymphopenia. CONCLUSIONS Subjects with a defective MBL2 genotype (i.e., 0/0) are predisposed to a more severe acute Sars-CoV-2 infection; they may benefit from early replacement therapy with recombinant MBL. Furthermore, a subset of subjects with the A/A MBL genotype develop a relevant increase of serum MBL during the early phases of the disease and develop a more severe pulmonary disease; in these patients, the targeting of the complement may help. Therefore, COVID-19 patients should be tested at hospitalization with serum MBL analysis and MBL2 genotype, to define the optimal therapy.
Collapse
Affiliation(s)
- Filippo Scialò
- Dipartimento di Scienze Mediche Traslazionali, Università della Campania "L. Vanvitelli", Naples, Italy
- CEINGE-Biotecnologie Avanzate, scarl, Naples, Italy
| | - Gustavo Cernera
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, scarl, Naples, Italy
| | | | - Biagio Pinchera
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Ivan Gentile
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Marina Di Domenico
- Dipartimento di Medicina di Precisione, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Andrea Bianco
- Dipartimento di Scienze Mediche Traslazionali, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Lucio Pastore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, scarl, Naples, Italy
| | - Felice Amato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, scarl, Naples, Italy
| | - Giuseppe Castaldo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, scarl, Naples, Italy
| |
Collapse
|
26
|
Saleki K, Alijanizadeh P, Azadmehr A. Is neuropilin-1 the neuroimmune initiator of multi-system hyperinflammation in COVID-19? Biomed Pharmacother 2023; 167:115558. [PMID: 37748412 DOI: 10.1016/j.biopha.2023.115558] [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: 07/09/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023] Open
Abstract
A major immunopathological feature of Coronavirus disease-2019 (COVID-19) is excessive inflammation in the form of "cytokine storm". The storm is characterized by injurious levels of cytokines which form a complicated network damaging different organs, including the lungs and the brain. The main starter of "cytokine network" hyperactivation in COVID-19 has not been discovered yet. Neuropilins (NRPs) are transmembrane proteins that act as neuronal guidance and angiogenesis modulators. The crucial function of NRPs in forming the nervous and vascular systems has been well-studied. NRP1 and NRP2 are the two identified homologs of NRP. NRP1 has been shown as a viral entry pathway for SARS-CoV2, which facilitates neuroinvasion by the virus within the central or peripheral nervous systems. These molecules directly interact with various COVID-19-related molecules, such as specific regions of the spike protein (major immune element of SARS-CoV2), vascular endothelial growth factor (VEGF) receptors, VEGFR1/2, and ANGPTL4 (regulator of vessel permeability and integrity). NRPs mainly play a role in hyperinflammatory injury of the CNS and lungs, and also the liver, kidney, pancreas, and heart in COVID-19 patients. New findings have suggested NRPs good candidates for pharmacotherapy of COVID-19. However, therapeutic targeting of NRP1 in COVID-19 is still in the preclinical phase. This review presents the implications of NRP1 in multi-organ inflammation-induced injury by SARS-CoV2 and provides insights for NRP1-targeting treatments for COVID-19 patients.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences(SBMU), Tehran, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
27
|
Huang SM, Hsieh CY, Ting JU, De Castro-Cruz KA, Wang CC, Lee CJ, Tsai PW. Anti-COVID-19, Anti-Inflammatory, and Anti-Osteoarthritis Activities of Sesamin from Sesamum indicum L. Bioengineering (Basel) 2023; 10:1263. [PMID: 38002386 PMCID: PMC10669907 DOI: 10.3390/bioengineering10111263] [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: 09/07/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
During the COVID-19 (coronavirus disease 2019) outbreak, many people were infected, and the symptoms may persist for several weeks or months for recovering patients. This is also known as "long COVID" and includes symptoms such as fatigue, joint pain, muscle pain, et cetera. The COVID-19 virus may trigger hyper-inflammation associated with cytokine levels in the body. COVID-19 can trigger inflammation in the joints, which can lead to osteoarthritis (OA), while long-term COVID-19 symptoms may lead to joint damage and other inflammation problems. According to several studies, sesame has potent anti-inflammatory properties due to its major constituent, sesamin. This study examined sesamin's anti-inflammatory, anti-osteoarthritis, and anti-COVID-19 effects. Moreover, in vivo and in vitro assays were used to determine sesamin's anti-inflammatory activity against the RAW264.7 and SW1353 cell lines. Sesamin had a dose-dependent effect (20 mg/kg) in a monoiodoacetic acid (MIA)-induced osteoarthritis rat model. Sesamin reduced paw swelling and joint discomfort. In addition, the findings indicated that sesamin suppressed the expression of iNOS (inducible nitric oxide synthase) and COX-2 (cyclooxygenase-2) in the RAW264.7 cell line within the concentration range of 6.25-50 μM. Furthermore, sesamin also had a suppressive effect on MMP (matrix metalloproteinase) expression in chondrocytes and the SW1353 cell line within the same concentration range of 6.25-50 μM. To examine the anti-viral activity, an in silico analysis was performed to evaluate sesamin's binding affinity with SARS-CoV-2 RdRp (severe acute respiratory syndrome coronavirus 2 RNA-dependent RNA polymerase) and human ACE2 (angiotensin-converting enzyme 2). Compared to the controls, sesamin exhibited strong binding affinities towards SARS-CoV-2 RdRp and human ACE2. Furthermore, sesamin had a higher binding affinity for the ACE2 target protein. This study suggests that sesamin shows potential anti-SARS-CoV-2 activity for drug development.
Collapse
Affiliation(s)
- Shu-Ming Huang
- Department of Nutrition, College of Medical and Health Care, Hungkuang University, Taichung 433, Taiwan;
- Department of Nutrition, Nantou Hospital of Ministry of Health and Welfare, Nantou 540, Taiwan
| | - Cheng-Yang Hsieh
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-C.W.)
- Laboratory of Oncology, Pharmacy Practice and Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Jasmine U. Ting
- Department of Chemistry, College of Science, De La Salle University, Metro Manila 1004, Philippines;
| | - Kathlia A. De Castro-Cruz
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Metro Manila 1002, Philippines;
| | - Ching-Chiung Wang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-C.W.)
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Orthopedics Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Chia-Jung Lee
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-C.W.)
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Po-Wei Tsai
- Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan
| |
Collapse
|
28
|
Dzhalilova D, Kosyreva A, Lokhonina A, Tsvetkov I, Vishnyakova P, Makarova O, Fatkhudinov T. Molecular and phenotypic distinctions of macrophages in tolerant and susceptible to hypoxia rats. PeerJ 2023; 11:e16052. [PMID: 37842051 PMCID: PMC10573310 DOI: 10.7717/peerj.16052] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/16/2023] [Indexed: 10/17/2023] Open
Abstract
Individual hypoxia tolerance is a major influence on the course and outcome of infectious and inflammatory diseases. Macrophages, which play central roles in systemic inflammatory response and other immunity reactions, are subject to functional activation orchestrated by several transcription factors including hypoxia inducible factors (HIFs). HIF-1 expression levels and the lipopolysaccharide (LPS)-induced systemic inflammatory response severity have been shown to correlate with hypoxia tolerance. Molecular and functional features of macrophages, depending on the organisms resistance to hypoxia, can determine the severity of the course of infectious and inflammatory diseases, including the systemic inflammatory response. The purpose is the comparative molecular and functional characterization of non-activated and LPS-activated bone marrow-derived macrophages under normoxia in rats with different tolerance to oxygen deprivation. Hypoxia resistance was assessed by gasping time measurement in an 11,500 m altitude-equivalent hypobaric decompression chamber. Based on the outcome, the animals were assigned to three groups termed 'tolerant to hypoxia' (n = 12), 'normal', and 'susceptible to hypoxia' (n = 13). The 'normal' group was excluded from subsequent experiments. One month after hypoxia resistance test, the blood was collected from the tail vein to isolate monocytes. Non-activated and LPS-activated macrophage cultures were investigated by PCR, flow cytometry and Western blot methods. Gene expression patterns of non-activated cultured macrophages from tolerant and susceptible to hypoxia animals differed. We observed higher expression of VEGF and CD11b and lower expression of Tnfa, Il1b and Epas1 in non-activated cultures obtained from tolerant to hypoxia animals, whereas HIF-1α mRNA and protein expression levels were similar. LPS-activated macrophage cultures derived from susceptible to hypoxia animals expressed higher levels of Hif1a and CCR7 than the tolerant group; in addition, the activation was associated with increased content of HIF-1α in cell culture medium. The observed differences indicate a specific propensity toward pro-inflammatory macrophage polarization in susceptible to hypoxia rats.
Collapse
Affiliation(s)
- Dzhuliia Dzhalilova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russian Federation
| | - Anna Kosyreva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russian Federation
| | - Anastasiya Lokhonina
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russian Federation
| | - Ivan Tsvetkov
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russian Federation
| | - Polina Vishnyakova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russian Federation
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
| | - Olga Makarova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russian Federation
| | - Timur Fatkhudinov
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russian Federation
| |
Collapse
|
29
|
Wu W, Cheng C, Yuan D, Peng L, Li L. Explore intersection genes of oxymatrine and COVID-19 with lung cancer as potential therapeutic targets based on network pharmacology. J Med Microbiol 2023; 72. [PMID: 37855710 DOI: 10.1099/jmm.0.001766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
Introduction. Oxymatrine is a natural quinazine alkaloid extracted from Sophora flavescens and has many medicinal values. Oxymatrine showed protective effects, viral inhibition and effects against lung cancer.Hypothesis/Gap Statement. Individuals with lung cancer exhibit heightened vulnerability to COVID-19 infection due to compromised immune function. In conjunction with COVID-19, it is hypothesized that oxymatrine may exert potent pharmacological effects on lung cancer patients.Aim. The objective of this study was to assess the pharmacological mechanisms and targets of oxymatrine in relation to COVID-19 lung cancer.Methodology. Utilizing network pharmacology analysis, a selection of 2628 genes were identified as co-targets for both COVID-19 and lung cancer. Subsequently, a clinicopathological analysis was conducted by integrating RNA-Seq and clinical data obtained from the TCGA-LUAD lung cancer dataset, which was acquired from the official TCGA website. The identification of pharmacological targets for oxymatrine was accomplished through the utilization of various databases including Pharm mapper, SWISS Target prediction, and STITCH. These identified targets were further investigated for protein-protein interaction (PPI) using STRING, as well as for gene ontology (GO) and KEGG pathways.Results. The effects of oxymatrine on COVID-19-induced lung cancer were mediated by immune regulation, cytoprotection, antiviral, and anti-inflammatory activities, immune regulation, and control of related signalling pathways, including the formation of the neutrophil extracellular trap, phagosome, Toll-like receptor signalling pathway, apoptosis, proteoglycans in cancer, extracellular matrix disassembly, and proteolysis involved in cellular protein catabolism. Furthermore, important substances and genes like ALB, MMP3, MMP1, and TLR4 may affect how oxymatrine suppresses lung cancer/COVID-19 development.Conclusion. To treat COVID-19 or lung cancer paired with COVID-19, oxymatrine may improve the therapeutic efficacy of current clinical antiviral medicines and immunotherapy.
Collapse
Affiliation(s)
- Wei Wu
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Chuan Cheng
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Dongdong Yuan
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Li Peng
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Le Li
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| |
Collapse
|
30
|
Martucci LF, Eichler RA, Silva RN, Costa TJ, Tostes RC, Busatto GF, Seelaender MC, Duarte AJ, Souza HP, Ferro ES. Intracellular peptides in SARS-CoV-2-infected patients. iScience 2023; 26:107542. [PMID: 37636076 PMCID: PMC10448160 DOI: 10.1016/j.isci.2023.107542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/29/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Intracellular peptides (InPeps) generated by the orchestrated action of the proteasome and intracellular peptidases have biological and pharmacological significance. Here, human plasma relative concentration of specific InPeps was compared between 175 patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and 45 SARS-CoV-2 non-infected patients; 2,466 unique peptides were identified, of which 67% were InPeps. The results revealed differences of a specific group of peptides in human plasma comparing non-infected individuals to patients infected by SARS-CoV-2, following the results of the semi-quantitative analyses by isotope-labeled electrospray mass spectrometry. The protein-protein interactions networks enriched pathways, drawn by genes encoding the proteins from which the peptides originated, revealed the presence of the coronavirus disease/COVID-19 network solely in the group of patients fatally infected by SARS-CoV-2. Thus, modulation of the relative plasma levels of specific InPeps could be employed as a predictive tool for disease outcome.
Collapse
Affiliation(s)
- Luiz Felipe Martucci
- Department of Pharmacology, Biomedical Sciences Institute, São Paulo 05508-000, Brazil
| | | | - Renée N.O. Silva
- Department of Pharmacology, Biomedical Sciences Institute, São Paulo 05508-000, Brazil
| | - Tiago J. Costa
- Department of Pharmacology, Ribeirao Preto Medical School, Ribeirão Preto 14049-900, Brazil
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, Ribeirão Preto 14049-900, Brazil
| | - Geraldo F. Busatto
- Department of Psichiatry, Medical School and Hospital das Clínicas, University of São Paulo, 01246-903 SP, Brazil
| | - Marilia C.L. Seelaender
- Department of Surgery, Medical School and Hospital das Clínicas, University of São Paulo, 01246-903 SP, Brazil
| | - Alberto J.S. Duarte
- Department of Patology, Medical School and Hospital das Clínicas, University of São Paulo, 01246-903 SP, Brazil
| | - Heraldo P. Souza
- Department of Internal Medicine, Medical School and Hospital das Clínicas, University of São Paulo, 01246-903 SP, Brazil
| | - Emer S. Ferro
- Department of Pharmacology, Biomedical Sciences Institute, São Paulo 05508-000, Brazil
- Department of Patology, Medical School and Hospital das Clínicas, University of São Paulo, 01246-903 SP, Brazil
- Department of Internal Medicine, Medical School and Hospital das Clínicas, University of São Paulo, 01246-903 SP, Brazil
| |
Collapse
|
31
|
Ceasovschih A, Sorodoc V, Shor A, Haliga RE, Roth L, Lionte C, Onofrei Aursulesei V, Sirbu O, Culis N, Shapieva A, Tahir Khokhar MAR, Statescu C, Sascau RA, Coman AE, Stoica A, Grigorescu ED, Banach M, Thomopoulos C, Sorodoc L. Distinct Features of Vascular Diseases in COVID-19. J Inflamm Res 2023; 16:2783-2800. [PMID: 37435114 PMCID: PMC10332421 DOI: 10.2147/jir.s417691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic was declared in early 2020 after several unexplained pneumonia cases were first reported in Wuhan, China, and subsequently in other parts of the world. Commonly, the disease comprises several clinical features, including high temperature, dry cough, shortness of breath, and hypoxia, associated with findings of interstitial pneumonia on chest X-ray and computer tomography. Nevertheless, severe forms of acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) are not limited to the respiratory tract but also may be extended to other systems, including the cardiovascular system. The bi-directional relationship between atherosclerosis and COVID-19 is accompanied by poor prognosis. The immune response hyperactivation due to SARS-CoV-2 infection causes an increased secretion of cytokines, endothelial dysfunction, and arterial stiffness, which promotes the development of atherosclerosis. Also, due to the COVID-19 pandemic, access to healthcare amenities was reduced, resulting in increased morbidity and mortality in patients at risk. Furthermore, as lockdown measures were largely adopted worldwide, the sedentary lifestyle and the increased consumption of processed nutrients or unhealthy food increased, and in the consequence, we might observe even 70% of overweight and obese population. Altogether, with the relatively low ratio of vaccinated people in many countries, and important health debt appeared, which is now and will be for next decade a large healthcare challenge. However, the experience gained in the COVID-19 pandemic and the new methods of patients' approaching have helped the medical system to overcome this crisis and will hopefully help in the case of new possible epidemics.
Collapse
Affiliation(s)
- Alexandr Ceasovschih
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, Iasi, 700111, Romania
| | - Victorita Sorodoc
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, Iasi, 700111, Romania
| | - Annabelle Shor
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
| | - Raluca Ecaterina Haliga
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, Iasi, 700111, Romania
| | - Lynn Roth
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, 2610, Belgium
| | - Catalina Lionte
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, Iasi, 700111, Romania
| | | | - Oana Sirbu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, Iasi, 700111, Romania
| | - Nicolae Culis
- Nottingham University Hospitals NHS Trust, Queen’s Medical Center, Nottingham, NG72UH, UK
| | - Albina Shapieva
- Cardiac Electrophysiology Department, Petrovsky National Research Center of Surgery, Moscow, 119991, Russia
| | | | - Cristian Statescu
- Department of Cardiology, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Iasi, 700503, Romania
| | - Radu A Sascau
- Department of Cardiology, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Iasi, 700503, Romania
| | - Adorata Elena Coman
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
| | - Alexandra Stoica
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, Iasi, 700111, Romania
| | - Elena-Daniela Grigorescu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, 93338, Poland
| | - Costas Thomopoulos
- Department of Cardiology, Elena Venizelou General Hospital, Athens, GR-11522, Greece
| | - Laurentiu Sorodoc
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, 700115, Romania
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, Iasi, 700111, Romania
| |
Collapse
|
32
|
Jang H, Choudhury S, Yu Y, Sievers BL, Gelbart T, Singh H, Rawlings SA, Proal A, Tan GS, Qian Y, Smith D, Freire M. Persistent immune and clotting dysfunction detected in saliva and blood plasma after COVID-19. Heliyon 2023; 9:e17958. [PMID: 37483779 PMCID: PMC10362241 DOI: 10.1016/j.heliyon.2023.e17958] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
A growing number of studies indicate that coronavirus disease 2019 (COVID-19) is associated with inflammatory sequelae, but molecular signatures governing the normal versus pathologic convalescence process have not been well-delineated. Here, we characterized global immune and proteome responses in matched plasma and saliva samples obtained from COVID-19 patients collected between 20 and 90 days after initial clinical symptoms resolved. Convalescent subjects showed robust total IgA and IgG responses and positive antibody correlations in saliva and plasma samples. Shotgun proteomics revealed persistent inflammatory patterns in convalescent samples including dysfunction of salivary innate immune cells, such as neutrophil markers (e.g., myeloperoxidase), and clotting factors in plasma (e.g., fibrinogen), with positive correlations to acute COVID-19 disease severity. Saliva samples were characterized by higher concentrations of IgA, and proteomics showed altered myeloid-derived pathways that correlated positively with SARS-CoV-2 IgA levels. Beyond plasma, our study positions saliva as a viable fluid to monitor normal and aberrant immune responses including vascular, inflammatory, and coagulation-related sequelae.
Collapse
Affiliation(s)
- Hyesun Jang
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | | | - Yanbao Yu
- Department of Chemistry & Biochemistry, University of Delaware, Newark, DE, USA, 19716
| | - Benjamin L Sievers
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | - Terri Gelbart
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | - Harinder Singh
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | - Stephen A Rawlings
- MMP Adult Infectious Disease, Maine Medical Center, South Portland, ME, 04106, USA
| | - Amy Proal
- PolyBio Research Foundation. Mercer Island, WA, USA
| | - Gene S Tan
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yu Qian
- Informatics, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | - Davey Smith
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marcelo Freire
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
33
|
Min JS, Jin YH, Kwon S. Auraptene Has Antiviral Activity against Human Coronavirus OC43 in MRC-5 Cells. Nutrients 2023; 15:2960. [PMID: 37447286 DOI: 10.3390/nu15132960] [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: 05/31/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Auraptene (7-geranyloxycoumarin) is the abundant prenyloxycoumarin found in the fruits of Citrus spp. Auraptene has a variety of pharmacological and therapeutic functions, such as anticancer, antioxidant, immunomodulatory, and anti-inflammation activities, with excellent safety profiles. In this study, we evaluated the anticoronaviral activity of auraptene in HCoV-OC43-infected human lung fibroblast MRC-5 cells. We found that auraptene effectively inhibited HCoV-OC43-induced cytopathic effects with 4.3 μM IC50 and 6.1 μM IC90, resulting in a selectivity index (CC50/IC50) of >3.5. Auraptene treatment also decreased viral RNA levels in HCoV-OC43-infected cells, as detected through quantitative real-time PCR, and decreased the expression level of spike proteins and nucleocapsid proteins in virus-infected cells, as detected through the Western blot analysis and immunofluorescence staining. Time-of-addition analysis showed auraptene's inhibitory effects at the post-entry stage of the virus life cycle; however, auraptene did not induce the antiviral interferon families, IFN-α1, IFN-β1, and IFN-λ1. Additionally, auraptene-treated MRC-5 cells during HCoV-OC43 infection decreased the MMP-9 mRNA levels which are usually increased due to the infection, as auraptene is a previously reported MMP-9 inhibitor. Therefore, auraptene showed antiviral activity against HCoV-OC43 infection, and we suggest that auraptene has the potential to serve as a therapeutic agent against human coronavirus.
Collapse
Affiliation(s)
- Jung Sun Min
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Young-Hee Jin
- KM Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea
| | - Sunoh Kwon
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| |
Collapse
|
34
|
Zingaropoli MA, Latronico T, Pasculli P, Masci GM, Merz R, Ciccone F, Dominelli F, Del Borgo C, Lichtner M, Iafrate F, Galardo G, Pugliese F, Panebianco V, Ricci P, Catalano C, Ciardi MR, Liuzzi GM, Mastroianni CM. Tissue Inhibitor of Matrix Metalloproteinases-1 (TIMP-1) and Pulmonary Involvement in COVID-19 Pneumonia. Biomolecules 2023; 13:1040. [PMID: 37509076 PMCID: PMC10377146 DOI: 10.3390/biom13071040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Background: The aim of the study was to longitudinally evaluate the association between MMP-2, MMP-9, TIMP-1 and chest radiological findings in COVID-19 patients. Methods: COVID-19 patients were evaluated based on their hospital admission (baseline) and three months after hospital discharge (T post) and were stratified into ARDS and non-ARDS groups. As a control group, healthy donors (HD) were enrolled. Results: At the baseline, compared to HD (n = 53), COVID-19 patients (n = 129) showed higher plasma levels of MMP-9 (p < 0.0001) and TIMP-1 (p < 0.0001) and the higher plasma activity of MMP-2 (p < 0.0001) and MMP-9 (p < 0.0001). In the ARDS group, higher plasma levels of MMP-9 (p = 0.0339) and TIMP-1 (p = 0.0044) and the plasma activity of MMP-2 (p = 0.0258) and MMP-9 (p = 0.0021) compared to non-ARDS was observed. A positive correlation between the plasma levels of TIMP-1 and chest computed tomography (CT) score (ρ = 0.2302, p = 0.0160) was observed. At the T post, a reduction in plasma levels of TIMP-1 (p < 0.0001), whereas an increase in the plasma levels of MMP-9 was observed (p = 0.0088). Conclusions: The positive correlation between TIMP-1 with chest CT scores highlights its potential use as a marker of fibrotic burden. At T post, the increase in plasma levels of MMP-9 and the reduction in plasma levels of TIMP-1 suggested that inflammation and fibrosis resolution were still ongoing.
Collapse
Affiliation(s)
- Maria Antonella Zingaropoli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Tiziana Latronico
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Patrizia Pasculli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Giorgio Maria Masci
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Roberta Merz
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Federica Ciccone
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Federica Dominelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Cosmo Del Borgo
- Infectious Diseases Unit, Santa Maria Goretti Hospital, Sapienza, University of Rome, 04100 Latina, Italy
| | - Miriam Lichtner
- Infectious Diseases Unit, Santa Maria Goretti Hospital, Sapienza, University of Rome, 04100 Latina, Italy
- Department of Neurosciences Mental Health and Sensory Organs, Sapienza University of Rome, 00161 Rome, Italy
| | - Franco Iafrate
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Gioacchino Galardo
- Medical Emergency Unit, Sapienza University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Francesco Pugliese
- Department of Specialist Surgery and Organ Transplantation "Paride Stefanini", Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy
| | - Valeria Panebianco
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Paolo Ricci
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Unit of Emergency Radiology, Policlinico Umberto I, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Carlo Catalano
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Grazia Maria Liuzzi
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
35
|
Sai Ramesh A, Adarshan S, Lohedan H, Naveen Kumar T, Thasleema Nasrin MR, Aarthi Shree G, Dinakarkumar Y, Ramalingam RJ, Karnan M. Computational analysis of the phytocompounds of Mimusops elengi against spike protein of SARS CoV2 - An Insilico model. Int J Biol Macromol 2023:125553. [PMID: 37356683 PMCID: PMC10289265 DOI: 10.1016/j.ijbiomac.2023.125553] [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: 02/24/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
The COVID-19 pandemic has been a global health crisis for over three years now, with the virus causing widespread illness and death. The urgent need for safe and effective therapeutic drugs has prompted the exploration of alternative medicine systems such as Ayurveda and Siddha. This study focuses on the potential therapeutic properties of the Ayurvedic plant, Mimusops elengi. In silico techniques were employed to analyze the bioactivity of the plant, including target prediction, gene ontology analysis, OMIM analysis, and molecular docking analysis. The results revealed 36 phytocompounds that interacted with 1431 receptors in the human body, and two compounds - hederagenin and quercetin - showed exceptionally high binding affinities toward their corresponding receptors, IL6 and MMP9. These results provide important insight into the potential therapeutic activity of M. elengi and its compounds in combating COVID-19. However, further research and clinical trials are necessary to validate these findings and develop safe and effective drugs. The study highlights the importance of combining traditional medicine with modern scientific methods to find effective treatments for global health challenges.
Collapse
Affiliation(s)
- A Sai Ramesh
- Department of Biotechnology, Vel Tech High Tech Dr. Rangarajan Dr. Sakunthala Engineering College, Avadi, Chennai 600062, Tamil Nadu, India
| | - S Adarshan
- Department of Biotechnology, Alagappa University, Karaikudi 630003, Tamil Nadu, India
| | - Hamad Lohedan
- Department of Chemistry, College of Science, Kind Saud University, Riyadh 11451, Saudi Arabia
| | - T Naveen Kumar
- Department of Biotechnology, Vel Tech High Tech Dr. Rangarajan Dr. Sakunthala Engineering College, Avadi, Chennai 600062, Tamil Nadu, India
| | - M R Thasleema Nasrin
- Department of Biotechnology, Vel Tech High Tech Dr. Rangarajan Dr. Sakunthala Engineering College, Avadi, Chennai 600062, Tamil Nadu, India
| | - G Aarthi Shree
- Department of Biotechnology, Vel Tech High Tech Dr. Rangarajan Dr. Sakunthala Engineering College, Avadi, Chennai 600062, Tamil Nadu, India
| | - Yuvaraj Dinakarkumar
- Department of Biotechnology, Vel Tech High Tech Dr. Rangarajan Dr. Sakunthala Engineering College, Avadi, Chennai 600062, Tamil Nadu, India.
| | - R Jothi Ramalingam
- Department of Chemistry, College of Science, Kind Saud University, Riyadh 11451, Saudi Arabia
| | - Muthusamy Karnan
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, South Korea
| |
Collapse
|
36
|
Jones TW, Almuntashiri S, Chase A, Alhumaid A, Somanath PR, Sikora A, Zhang D. Plasma matrix metalloproteinase-3 predicts mortality in acute respiratory distress syndrome: a biomarker analysis of a randomized controlled trial. Respir Res 2023; 24:166. [PMID: 37349704 PMCID: PMC10286483 DOI: 10.1186/s12931-023-02476-5] [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: 03/13/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Matrix metalloproteinase-3 (MMP-3) is a proteolytic enzyme involved in acute respiratory distress syndrome (ARDS) pathophysiology that may serve as a lung-specific biomarker in ARDS. METHODS This study was a secondary biomarker analysis of a subset of Albuterol for the Treatment of Acute Lung Injury (ALTA) trial patients to determine the prognostic value of MMP-3. Plasma sample MMP-3 was measured by enzyme-linked immunosorbent assay. The primary outcome was the area under the receiver operating characteristic (AUROC) of MMP-3 at day 3 for the prediction of 90-day mortality. RESULTS A total of 100 unique patient samples were evaluated and the AUROC analysis of day three MMP-3 showed an AUROC of 0.77 for the prediction of 90-day mortality (95% confidence interval: 0.67-0.87), corresponding to a sensitivity of 92% and specificity of 63% and an optimal cutoff value of 18.4 ng/mL. Patients in the high MMP-3 group (≥ 18.4 ng/mL) showed higher mortality compared to the non-elevated MMP-3 group (< 18.4 ng/mL) (47% vs. 4%, p < 0.001). A positive difference in day zero and day three MMP-3 concentration was predictive of mortality with an AUROC of 0.74 correlating to 73% sensitivity, 81% specificity, and an optimal cutoff value of + 9.5 ng/mL. CONCLUSIONS Day three MMP-3 concentration and difference in day zero and three MMP-3 concentrations demonstrated acceptable AUROCs for predicting 90-day mortality with a cut-point of 18.4 ng/mL and + 9.5 ng/mL, respectively. These results suggest a prognostic role of MMP-3 in ARDS.
Collapse
Affiliation(s)
- Timothy W. Jones
- Department of Pharmacy, Augusta University Medical Center, 1120 15th St., Augusta, GA 30912 USA
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Sultan Almuntashiri
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Aaron Chase
- Department of Pharmacy, Augusta University Medical Center, 1120 15th St., Augusta, GA 30912 USA
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Abdullah Alhumaid
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Payaningal R. Somanath
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Andrea Sikora
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Duo Zhang
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| |
Collapse
|
37
|
Russo E, Corrao S, Di Gaudio F, Alberti G, Caprnda M, Kubatka P, Kruzliak P, Miceli V, Conaldi PG, Borlongan CV, La Rocca G. Facing the Challenges in the COVID-19 Pandemic Era: From Standard Treatments to the Umbilical Cord-Derived Mesenchymal Stromal Cells as a New Therapeutic Strategy. Cells 2023; 12:1664. [PMID: 37371134 PMCID: PMC10297457 DOI: 10.3390/cells12121664] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which counts more than 650 million cases and more than 6.6 million of deaths worldwide, affects the respiratory system with typical symptoms such as fever, cough, sore throat, acute respiratory distress syndrome (ARDS), and fatigue. Other nonpulmonary manifestations are related with abnormal inflammatory response, the "cytokine storm", that could lead to a multiorgan disease and to death. Evolution of effective vaccines against SARS-CoV-2 provided multiple options to prevent the infection, but the treatment of the severe forms remains difficult to manage. The cytokine storm is usually counteracted with standard medical care and anti-inflammatory drugs, but researchers moved forward their studies on new strategies based on cell therapy approaches. The perinatal tissues, such as placental membranes, amniotic fluid, and umbilical cord derivatives, are enriched in mesenchymal stromal cells (MSCs) that exert a well-known anti-inflammatory role, immune response modulation, and tissue repair. In this review, we focused on umbilical-cord-derived MSCs (UC-MSCs) used in in vitro and in vivo studies in order to evaluate the weakening of the severe symptoms, and on recent clinical trials from different databases, supporting the favorable potential of UC-MSCs as therapeutic strategy.
Collapse
Affiliation(s)
- Eleonora Russo
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Simona Corrao
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | | | - Giusi Alberti
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, University Hospital Bratislava, 81499 Bratislava, Slovakia;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03649 Martin, Slovakia;
| | - Peter Kruzliak
- Research and Development Services, Pradlacka 18, 61300 Brno, Czech Republic;
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Cesario Venturina Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Giampiero La Rocca
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| |
Collapse
|
38
|
Salomão R, Assis V, de Sousa Neto IV, Petriz B, Babault N, Durigan JLQ, de Cássia Marqueti R. Involvement of Matrix Metalloproteinases in COVID-19: Molecular Targets, Mechanisms, and Insights for Therapeutic Interventions. BIOLOGY 2023; 12:843. [PMID: 37372128 PMCID: PMC10295079 DOI: 10.3390/biology12060843] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
MMPs are enzymes involved in SARS-CoV-2 pathogenesis. Notably, the proteolytic activation of MMPs can occur through angiotensin II, immune cells, cytokines, and pro-oxidant agents. However, comprehensive information regarding the impact of MMPs in the different physiological systems with disease progression is not fully understood. In the current study, we review the recent biological advances in understanding the function of MMPs and examine time-course changes in MMPs during COVID-19. In addition, we explore the interplay between pre-existing comorbidities, disease severity, and MMPs. The reviewed studies showed increases in different MMP classes in the cerebrospinal fluid, lung, myocardium, peripheral blood cells, serum, and plasma in patients with COVID-19 compared to non-infected individuals. Individuals with arthritis, obesity, diabetes, hypertension, autoimmune diseases, and cancer had higher MMP levels when infected. Furthermore, this up-regulation may be associated with disease severity and the hospitalization period. Clarifying the molecular pathways and specific mechanisms that mediate MMP activity is important in developing optimized interventions to improve health and clinical outcomes during COVID-19. Furthermore, better knowledge of MMPs will likely provide possible pharmacological and non-pharmacological interventions. This relevant topic might add new concepts and implications for public health in the near future.
Collapse
Affiliation(s)
- Rebecca Salomão
- Laboratory of Molecular Analysis, Postgraduate Program in Health and Sciences and Technology, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil
| | - Victoria Assis
- Laboratory of Molecular Analysis, Postgraduate Program in Rehabilitation Sciences, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil; (V.A.); (J.L.Q.D.)
| | - Ivo Vieira de Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-907, SP, Brazil;
| | - Bernardo Petriz
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasilia, Brasilia 71966-700, DF, Brazil;
- Laboratory of Exercise Molecular Physiology, University Center UDF, Brasília 71966-900, DF, Brazil
| | - Nicolas Babault
- INSERM UMR1093-CAPS, UFR des Sciences du Sport, Université de Bourgogne, F-21000 Dijon, France;
- Centre d’Expertise de la Performance, UFR des Sciences du Sport, Université de Bourgogne, F-21000 Dijon, France
| | - João Luiz Quaglioti Durigan
- Laboratory of Molecular Analysis, Postgraduate Program in Rehabilitation Sciences, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil; (V.A.); (J.L.Q.D.)
| | - Rita de Cássia Marqueti
- Laboratory of Molecular Analysis, Postgraduate Program in Health and Sciences and Technology, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil
- Laboratory of Molecular Analysis, Postgraduate Program in Rehabilitation Sciences, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil; (V.A.); (J.L.Q.D.)
| |
Collapse
|
39
|
Rombauts A, Bódalo Torruella M, Abelenda-Alonso G, Perera-Bel J, Ferrer-Salvador A, Acedo-Terrades A, Gabarrós-Subirà M, Oriol I, Gudiol C, Nonell L, Carratalà J. Dynamics of Gene Expression Profiling and Identification of High-Risk Patients for Severe COVID-19. Biomedicines 2023; 11:biomedicines11051348. [PMID: 37239019 DOI: 10.3390/biomedicines11051348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023] Open
Abstract
The clinical manifestations of SARS-CoV-2 infection vary widely, from asymptomatic infection to the development of acute respiratory distress syndrome (ARDS) and death. The host response elicited by SARS-CoV-2 plays a key role in determining the clinical outcome. We hypothesized that determining the dynamic whole blood transcriptomic profile of hospitalized adult COVID-19 patients and characterizing the subgroup that develops severe disease and ARDS would broaden our understanding of the heterogeneity in clinical outcomes. We recruited 60 hospitalized patients with RT-PCR-confirmed SARS-CoV-2 infection, among whom 19 developed ARDS. Peripheral blood was collected using PAXGene RNA tubes within 24 h of admission and on day 7. There were 2572 differently expressed genes in patients with ARDS at baseline and 1149 at day 7. We found a dysregulated inflammatory response in COVID-19 ARDS patients, with an increased expression of genes related to pro-inflammatory molecules and neutrophil and macrophage activation at admission, in addition to an immune regulation loss. This led, in turn, to a higher expression of genes related to reactive oxygen species, protein polyubiquitination, and metalloproteinases in the latter stages. Some of the most significant differences in gene expression found between patients with and without ARDS corresponded to long non-coding RNA involved in epigenetic control.
Collapse
Affiliation(s)
- Alexander Rombauts
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain
| | | | - Gabriela Abelenda-Alonso
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain
| | - Júlia Perera-Bel
- MARGenomics, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Anna Ferrer-Salvador
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | | | - Maria Gabarrós-Subirà
- MARGenomics, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Isabel Oriol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain
| | - Carlota Gudiol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, 08007 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Lara Nonell
- MARGenomics, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, 08007 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
40
|
Volchkova EV, Titova ON, Kuzubova NA, Lebedeva ES. Potential predictors of severe course and outcome of community-acquired pneumonia. PULMONOLOGIYA 2023; 33:225-232. [DOI: 10.18093/0869-0189-2023-33-2-225-232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Severe pneumonia is a condition with a high risk of death and mandatory hospitalization in the intensive care unit. The incidence of severe pneumonia has increased dramatically during the pandemic of new coronavirus infection. Timely diagnosis and early initiation of adequate treatment of severe pneumonia are crucial for improving survival of critically ill patients.The aim of this review was to analyze published scientific research on molecular markers that allow to objectively assess the severity of pneumonia and to determine treatment tactics based on the predicted outcome upon admission to the hospital. A systematic search was conducted in the electronic databases PubMed, Medline, Web of Science for the period 2019 - 2022.Conclusion. The review focuses on the prognostic role of a number of markers of immune response, vascular transformation, as well as angiotensin II and angiotensin converting enzyme-2. Further prospective studies of potential predictors of severe pneumonia will enable using marker molecules in a comprehensive clinical and laboratory diagnosis for early prediction of the hospitalized patient’s condition and expected outcome.
Collapse
Affiliation(s)
- E. V. Volchkova
- Pediatrics and Child Health Research Institute of the “Central Clinical Hospital of the Russian Academy of Sciences”, Ministry of Education and Science of Russia
| | - O. N. Titova
- Federal State Budgetary Educational Institution of Higher Education “Academician I.P.Pavlov First St. Petersburg State Medical University”, Ministry of Healthcare of Russian Federation
| | - N. A. Kuzubova
- Federal State Budgetary Educational Institution of Higher Education “Academician I.P.Pavlov First St. Petersburg State Medical University”, Ministry of Healthcare of Russian Federation
| | - E. S. Lebedeva
- Federal State Budgetary Educational Institution of Higher Education “Academician I.P.Pavlov First St. Petersburg State Medical University”, Ministry of Healthcare of Russian Federation
| |
Collapse
|
41
|
Abstract
Vascular age is determined by functional and structural changes in the arterial wall. When measured by its proxy, pulse wave velocity, it has been shown to predict cardiovascular and total mortality. Disconcordance between chronological and vascular age might represent better or worse vascular health. Cell senescence is caused by oxidative stress and sustained cell replication. Senescent cells acquire senescence-associated secretory phenotype. Oxidative stress, endothelial dysfunction, dysregulation of coagulation and leucocyte infiltration are observed in the aging endothelium. All of these mechanisms lead to increased vascular calcification and stiffness. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can involve the vascular endothelium. It enters cells using angiotensin-converting enzyme 2 (ACE-2) receptors, which are abundant in endothelial cells. The damage this virus does to the endothelium can be direct or indirect. Indirect damage is caused by hyperinflammation. Direct damage results from effects on ACE-2 receptors. The reduction of ACE-2 levels seen during coronavirus disease 2019 (COVID-19) infection might cause vasoconstriction and oxidative stress. COVID-19 and vascular aging share some pathways. Due to the novelty of the virus, there is an urgent need for studies that investigate its long-term effects on vascular health.
Collapse
Affiliation(s)
- Ignas Badaras
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania,Ignas Badaras, Faculty of Medicine, Vilnius
University, M. K. Ciurlionio g. 21/27, LT-03101, Vilnius 01513, Lithuania.
| | - Agnė Laučytė-Cibulskienė
- Department of Nephrology, Skåne University
Hospital, Malmö, Sweden,Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
42
|
Roy A, Sarkar A, Nayak D, Das S. Ultradiluted SARS-CoV-2 Spike Protein mitigates hyperinflammation in lung via ferritin and MMP-9 regulation in BALB/c mice. Virus Res 2023; 329:199091. [PMID: 36918101 PMCID: PMC10015450 DOI: 10.1016/j.virusres.2023.199091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023]
Abstract
AIM This study investigated the prophylactic and therapeutic role of ultradiluted preparation of the Delta variant of SARS-CoV-2 recombinant spike (S) protein during S antigen-induced inflammatory process of disease progression along with the probable mechanism of action. MAIN METHODS Ultradiluted S protein (UDSP) was prepared and administered orally to adult BALB/c mice before and after administration of S antigen intranasally. After an observation period of 72 h, animals were sacrificed and expression level of ferritin was assayed through ELISA. The genetic expressions of cytokines, IL-6, IL-10, IL-1β, TNFα, IL-17, MMP-9, TIMP-1, ferritin light and heavy chains, and mitochondrial ferritin from lung tissues were investigated through RT-PCR. Formalin-fixed lung tissue sections were stained with hematoxylin and eosin to observe the degree of pathological changes. The activity of MMP-9 in lung tissues was investigated through gelatin zymography and immunofluorescence of MMP-9 in lung tissue sections was performed to revalidate the finding from gelatin zymography. Systems biology approach was used to elucidate a probable pathway where UDSP attenuated the inflammation through the regulation of pro- and anti-inflammatory cytokines. KEY FINDINGS UDSP attenuated the S antigen-induced hyperinflammation in the lung by regulating pro- and anti-inflammatory cytokines, calming cytokine storm, reducing ferritin level both in transcriptional and translational levels, and restoring critical ratio of MMP-9: TIMP-1. SIGNIFICANCE Our findings suggest a probable pathway by which UDSP might have attenuated inflammation through the regulation of cytokines, receptors, and other molecules. This proclaims UDSP as a promising antiviral agent in the treatment of COVID-19-induced immunopathogenesis.
Collapse
Affiliation(s)
- Anirban Roy
- Virology Laboratory, DAC Regional Research Institute, Kolkata, West Bengal 700035, India
| | - Avipsha Sarkar
- Virology Laboratory, DAC Regional Research Institute, Kolkata, West Bengal 700035, India
| | - Debadatta Nayak
- CCRH, Institutional Area, Janakpuri, New Delhi, Delhi 110058, India
| | - Satadal Das
- Virology Laboratory, DAC Regional Research Institute, Kolkata, West Bengal 700035, India.
| |
Collapse
|
43
|
Khalid U, Dimov D, Vlaykova T. Matrix metalloproteinases in COVID-19: underlying significance. BIOTECHNOL BIOTEC EQ 2023. [DOI: 10.1080/13102818.2023.2186137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Affiliation(s)
- Usman Khalid
- Faculty of Medicine, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Dimo Dimov
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine, Trakia University, Stara Zagora, Bulgaria
| | - Tatyana Vlaykova
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine, Trakia University, Stara Zagora, Bulgaria
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
44
|
Plasma N-Cleaved Galectin-9 Is a Surrogate Marker for Determining the Severity of COVID-19 and Monitoring the Therapeutic Effects of Tocilizumab. Int J Mol Sci 2023; 24:ijms24043591. [PMID: 36835000 PMCID: PMC9964849 DOI: 10.3390/ijms24043591] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Galectin-9 (Gal-9) is known to contribute to antiviral responses in coronavirus disease 2019 (COVID-19). Increased circulating Gal-9 in COVID-19 is associated with COVID-19 severity. In a while, the linker-peptide of Gal-9 is susceptible to proteolysis that can cause the change or loss of Gal-9 activity. Here, we measured plasma levels of N-cleaved-Gal9, which is Gal9 carbohydrate-recognition domain at the N-terminus (NCRD) with attached truncated linker peptide that differs in length depending on the type of proteases, in COVID-19. We also investigated the time course of plasma N-cleaved-Gal9 levels in severe COVID-19 treated with tocilizumab (TCZ). As a result, we observed an increase in plasma N-cleaved-Gal9 levels in COVID-19 and its higher levels in COVID-19 with pneumonia compared to the mild cases (healthy: 326.1 pg/mL, mild: 698.0 pg/mL, and with pneumonia: 1570 pg/mL). N-cleaved-Gal9 levels were associated with lymphocyte counts, C-reactive protein (CRP), soluble interleukin-2 receptor (sIL-2R), D-dimer, and ferritin levels, and ratio of percutaneous oxygen saturation to fraction of inspiratory oxygen (S/F ratio) in COVID-19 with pneumonia and discriminated different severity groups with high accuracy (area under the curve (AUC): 0.9076). Both N-cleaved-Gal9 and sIL-2R levels were associated with plasma matrix metalloprotease (MMP)-9 levels in COVID-19 with pneumonia. Furthermore, a decrease in N-cleaved-Gal9 levels was associated with a decrease of sIL-2R levels during TCZ treatment. N-cleaved-Gal9 levels showed a moderate accuracy (AUC: 0.8438) for discriminating the period before TCZ from the recovery phase. These data illustrate that plasma N-cleaved-Gal9 is a potential surrogate marker for assessing COVID-19 severity and the therapeutic effects of TCZ.
Collapse
|
45
|
Donniacuo M, De Angelis A, Rafaniello C, Cianflone E, Paolisso P, Torella D, Sibilio G, Paolisso G, Castaldo G, Urbanek K, Rossi F, Berrino L, Cappetta D. COVID-19 and atrial fibrillation: Intercepting lines. Front Cardiovasc Med 2023; 10:1093053. [PMID: 36755799 PMCID: PMC9899905 DOI: 10.3389/fcvm.2023.1093053] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Almost 20% of COVID-19 patients have a history of atrial fibrillation (AF), but also a new-onset AF represents a frequent complication in COVID-19. Clinical evidence demonstrates that COVID-19, by promoting the evolution of a prothrombotic state, increases the susceptibility to arrhythmic events during the infective stages and presumably during post-recovery. AF itself is the most frequent form of arrhythmia and is associated with substantial morbidity and mortality. One of the molecular factors involved in COVID-19-related AF episodes is the angiotensin-converting enzyme (ACE) 2 availability. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses ACE2 to enter and infect multiple cells. Atrial ACE2 internalization after binding to SARS-CoV-2 results in a raise of angiotensin (Ang) II, and in a suppression of cardioprotective Ang(1-7) formation, and thereby promoting cardiac hypertrophy, fibrosis and oxidative stress. Furthermore, several pharmacological agents used in COVID-19 patients may have a higher risk of inducing electrophysiological changes and cardiac dysfunction. Azithromycin, lopinavir/ritonavir, ibrutinib, and remdesivir, used in the treatment of COVID-19, may predispose to an increased risk of cardiac arrhythmia. In this review, putative mechanisms involved in COVID-19-related AF episodes and the cardiovascular safety profile of drugs used for the treatment of COVID-19 are summarized.
Collapse
Affiliation(s)
- Maria Donniacuo
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Concetta Rafaniello
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Pasquale Paolisso
- Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | | | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giuseppe Castaldo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| |
Collapse
|
46
|
Huang JJ, Wang CW, Liu Y, Zhang YY, Yang NB, Yu YC, Jiang Q, Song QF, Qian GQ. Role of the extracellular matrix in COVID-19. World J Clin Cases 2023; 11:73-83. [PMID: 36687194 PMCID: PMC9846981 DOI: 10.12998/wjcc.v11.i1.73] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/14/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
An outbreak of coronavirus disease 2019 (COVID-19) has spread globally, with over 500 million cases and 6 million deaths to date. COVID-19 is associated with a systemic inflammatory response and abnormalities of the extracellular matrix (ECM), which is also involved in inflammatory storms. Upon viral infection, ECM proteins are involved in the recruitment of inflammatory cells and interference with target organ metabolism, including in the lungs. Additionally, serum biomarkers of ECM turnover are associated with the severity of COVID-19 and may serve as potential targets. Consequently, understanding the expression and function of ECM, particularly of the lung, during severe acute respiratory syndrome of the coronavirus 2 infection would provide valuable insights into the mechanisms of COVID-19 progression. In this review, we summarize the current findings on ECM, such as hyaluronic acid, matrix metalloproteinases, and collagen, which are linked to the severity and inflammation of COVID-19. Some drugs targeting the extracellular surface have been effective. In the future, these ECM findings could provide novel perspectives on the pathogenesis and treatment of COVID-19.
Collapse
Affiliation(s)
- Jia-Jia Huang
- School of Medicine, Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Chu-Wen Wang
- School of Medicine, Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Ying Liu
- School of Medicine, Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Ying-Ying Zhang
- School of Medicine, Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Nai-Bin Yang
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo 315000, Zhejiang Province, China
| | - Yu-Chun Yu
- Department of Endocrinology, Ningbo Ninth Hospital, Ningbo 315000, Zhejiang Province, China
| | - Qi Jiang
- Department of Digestive, Ningbo First Hospital, Ningbo 315000, Zhejiang Province, China
| | - Qi-Fa Song
- Medical Data Center, Ningbo First Hospital, Ningbo 315000, Zhejiang Province, China
| | - Guo-Qing Qian
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo 315000, Zhejiang Province, China
| |
Collapse
|
47
|
Almuntashiri S, Zhang D, Somanath PR, Sikora A. MMP3 in Severe COVID-19: A Biomarker or Therapeutic Target? Infect Disord Drug Targets 2023; 23:e190622206159. [PMID: 35726419 PMCID: PMC11042506 DOI: 10.2174/1871526522666220619121539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 02/08/2023]
Abstract
Identifying novel therapies is a critical need in the treatment of coronavirus disease-19 (COVID-19) and acute respiratory distress syndrome (ARDS). Stromelysin-1, also known as matrixmetalloproteinase 3 (MMP3), has been investigated as a diagnostic biomarker and a potential pharmacological target. Here, we discuss the recent findings of Gelzo et al. in the context of additional MMP3 investigations to delineate its exact role in diagnosis, prognostication, and phenotyping, in addition to its promising role as a therapeutic target in COVID-19-associated respiratory failure.
Collapse
Affiliation(s)
- Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of
Pharmacy, University of Georgia, Augusta, GA, 30912, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of
Pharmacy, University of Georgia, Augusta, GA, 30912, USA
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, College of
Pharmacy, University of Georgia, Augusta, GA, 30912, USA
| | - Andrea Sikora
- Clinical and Experimental Therapeutics, College of
Pharmacy, University of Georgia, Augusta, GA, 30912, USA
| |
Collapse
|
48
|
Di Donato M. Editorial: March 2022: Ovarian and prostate cancer awareness month. Front Endocrinol (Lausanne) 2023; 14:1133963. [PMID: 36860370 PMCID: PMC9969119 DOI: 10.3389/fendo.2023.1133963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/12/2023] [Indexed: 02/15/2023] Open
|
49
|
Mimicking Gene-Environment Interaction of Higher Altitude Dwellers by Intermittent Hypoxia Training: COVID-19 Preventive Strategies. BIOLOGY 2022; 12:biology12010006. [PMID: 36671699 PMCID: PMC9855005 DOI: 10.3390/biology12010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
Cyclooxygenase 2 (COX2) inhibitors have been demonstrated to protect against hypoxia pathogenesis in several investigations. It has also been utilized as an adjuvant therapy in the treatment of COVID-19. COX inhibitors, which have previously been shown to be effective in treating previous viral and malarial infections are strong candidates for improving the COVID-19 therapeutic doctrine. However, another COX inhibitor, ibuprofen, is linked to an increase in the angiotensin-converting enzyme 2 (ACE2), which could increase virus susceptibility. Hence, inhibiting COX2 via therapeutics might not always be protective and we need to investigate the downstream molecules that may be involved in hypoxia environment adaptation. Research has discovered that people who are accustomed to reduced oxygen levels at altitude may be protected against the harmful effects of COVID-19. It is important to highlight that the study's conclusions only applied to those who regularly lived at high altitudes; they did not apply to those who occasionally moved to higher altitudes but still lived at lower altitudes. COVID-19 appears to be more dangerous to individuals residing at lower altitudes. The downstream molecules in the (COX2) pathway have been shown to adapt in high-altitude dwellers, which may partially explain why these individuals have a lower prevalence of COVID-19 infection. More research is needed, however, to directly address COX2 expression in people living at higher altitudes. It is possible to mimic the gene-environment interaction of higher altitude people by intermittent hypoxia training. COX-2 adaptation resulting from hypoxic exposure at altitude or intermittent hypoxia exercise training (IHT) seems to have an important therapeutic function. Swimming, a type of IHT, was found to lower COX-2 protein production, a pro-inflammatory milieu transcription factor, while increasing the anti-inflammatory microenvironment. Furthermore, Intermittent Hypoxia Preconditioning (IHP) has been demonstrated in numerous clinical investigations to enhance patients' cardiopulmonary function, raise cardiorespiratory fitness, and increase tissues' and organs' tolerance to ischemia. Biochemical activities of IHP have also been reported as a feasible application strategy for IHP for the rehabilitation of COVID-19 patients. In this paper, we aim to highlight some of the most relevant shared genes implicated with COVID-19 pathogenesis and hypoxia. We hypothesize that COVID-19 pathogenesis and hypoxia share a similar mechanism that affects apoptosis, proliferation, the immune system, and metabolism. We also highlight the necessity of studying individuals who live at higher altitudes to emulate their gene-environment interactions and compare the findings with IHT. Finally, we propose COX2 as an upstream target for testing the effectiveness of IHT in preventing or minimizing the effects of COVID-19 and other oxygen-related pathological conditions in the future.
Collapse
|
50
|
Almuntashiri S, Jones TW, Wang X, Sikora A, Zhang D. Plasma TIMP-1 as a sex-specific biomarker for acute lung injury. Biol Sex Differ 2022; 13:70. [PMID: 36482481 PMCID: PMC9733313 DOI: 10.1186/s13293-022-00481-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) confers high morbidity and mortality, with a death rate reaching 40%. Pre-clinical and clinical studies have cited sex-specific sex hormones as a critical contributor to divergent immunologic responses. Therefore, exploration of sex and sex hormone roles following lung injury and ARDS development is needed. Tissue inhibitor of metalloproteinase-1 (TIMP-1) was the first-discovered natural collagenase inhibitor and is located exclusively on the X chromosome. This study aimed to evaluate the prognostic role of circulating TIMP-1, and if concentration differences between males and females correlate with the mortality of ARDS patients. METHODS Human plasma samples from 100 ARDS patients enrolled in Albuterol to Treat Acute Lung Injury (ALTA) trial on the day of randomization were evaluated. The amount of TIMP-1 was measured using an enzyme-linked immunoassay (ELISA). Area under the receiver operating characteristic (AUROC) was computed to assess the predictive power of TIMP-1 for 30 and 90-day mortality. Chi-squared tests and Kaplan-Meier curves were computed to assess different variables and survival. RESULTS AUROC analysis of TIMP-1 and 30-day mortality among females showed that TIMP-1 exhibited an AUC of 0.87 (95% confidence interval [CI] 0.78 to 0.97; P = 0.0014) with an optimal cut-off value of 159.7 ng/mL producing a 100% sensitivity and 74% specificity. For 90-day mortality, AUROC analysis showed an AUC of 0.82 (95% confidence interval [CI] 0.67 to 0.97; P = 0.0016) with a similar cut-off value producing a 90% sensitivity and 76.47% specificity. Stratifying subjects by TIMP-1 concentration as high (≥ 159.7 ng/mL) or low (< 159.7 ng/mL) indicated that high TIMP-1 was associated with increased 30 and 90-day mortality rates (all P < 0.0001). Lastly, high TIMP-1 group was associated with worse other outcomes including ventilator-free days (VFDs) and ICU-free days (all P < 0.05). CONCLUSION Circulating TIMP-1 appeared to be a promising biomarker for mortality among females with ARDS. The high TIMP-1 group showed worse VFDs and ICU-free days. Circulating TIMP-1 may be a sex-specific biomarker in the setting of ARDS and could improve ARDS phenotyping as well as provide a novel therapeutic target in females.
Collapse
Affiliation(s)
- Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.,Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, 55473, Saudi Arabia
| | - Timothy W Jones
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Andrea Sikora
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA, 30901, USA.,Department of Pharmacy, Augusta University Medical Center, Augusta, GA, 30912, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA. .,Department of Medicine, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|