1
|
Li Y, Chen J, Xiao L, Guo Z, Huang J, Gao S, Li J, Li B, Liu Z. High-Lethality Precision-Guided Nanomissile for Broad-Spectrum Virucidal and Anti-Inflammatory Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27974-27987. [PMID: 40314777 DOI: 10.1021/acsami.5c03831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Viral infection, especially the past SARS-CoV-2 pandemic, has posed severe threat toward globalized healthcare, whereas vaccine and drug development can hardly keep up with the rate of virus mutation and resistance. In severe COVID-19 patients, the virus triggers a cytokine storm marked by excessive pro-inflammatory cytokine release, resulting in acute respiratory distress syndrome (ARDS). Therefore, a comprehensive strategy for viral neutralization and inflammation suppression is highly demanded. Herein, we designed a high-lethality precision-guided nanomissile for broad-spectrum virucidal and anti-inflammatory therapy. The nanomissile was a nanoscale molecularly imprinted polymer (nanoMIP) harboring hypervalent mannose-binding cavities and loaded with a magnetocaloric core and photothermal dye ICG. It demonstrated an ultrafast heating rate, increasing from 25.2 to 55.9 °C within 60 s under alternating magnetic field (AMF) and near-infrared (NIR) laser irradiation. In addition, the nanomissile exhibited a unique double-punch mechanism, being capable of targeting not only the conserved high-mannose glycans of SARS-CoV-2, HIV-1, LASV, and PDCoV with Kd values reaching 10-10 M but also heat-inactivating the virions right away. Beyond this, it also exhibited significant anti-inflammatory and immunomodulatory properties. In the mouse model, the nanomissile exerted outstanding therapeutic and prophylactic effects while inhibiting virus replication and protecting lung injury. Thus, this potently broad-spectrum virucidal strategy opens a new access to eradicating viral infectivity and inflammatory storm suspension.
Collapse
Affiliation(s)
- Ying Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jingran Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Li Xiao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Zhanchen Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jin Huang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Song Gao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Zhen Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
2
|
Xiong Y, Wang J, Shang X, Chen T, Fraser DD, Fonseca GJ, Rousseau S, Ding J. Efficient and scalable construction of clinical variable networks for complex diseases with RAMEN. CELL REPORTS METHODS 2025; 5:101022. [PMID: 40215965 DOI: 10.1016/j.crmeth.2025.101022] [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: 02/25/2024] [Revised: 12/09/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Understanding the interplay among clinical variables-such as demographics, symptoms, and laboratory results-and their relationships with disease outcomes is critical for advancing diagnostics and understanding mechanisms in complex diseases. Existing methods fail to capture indirect or directional relationships, while existing Bayesian network learning methods are computationally expensive and only infer general associations without focusing on disease outcomes. Here we introduce random walk- and genetic algorithm-based network inference (RAMEN), a method for Bayesian network inference that uses absorbing random walks to prioritize outcome-relevant variables and a genetic algorithm for efficient network refinement. Applied to COVID-19 (Biobanque québécoise de la COVID-19), intensive care unit (ICU) septicemia (MIMIC-III), and COPD (CanCOLD) datasets, RAMEN reconstructs networks linking clinical markers to disease outcomes, such as elevated lactate levels in ICU patients. RAMEN demonstrates advantages in computational efficiency and scalability compared to existing methods. By modeling outcome-specific relationships, RAMEN provides a robust tool for uncovering critical disease mechanisms, advancing diagnostics, and enabling personalized treatment strategies.
Collapse
Affiliation(s)
- Yiwei Xiong
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Jingtao Wang
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Xiaoxiao Shang
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Mathematics and Statistics, McGill University, 805 Sherbrooke Street West, Montreal, QC H3A 0B9, Canada
| | - Tingting Chen
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Hematology Department, Beijing Luhe Hospital, Capital Medical University, Xinhua South Road No. 82, Tongzhou District, Beijing 101149, China
| | - Douglas D Fraser
- Children's Health Research Institute, Victoria Research Laboratories, 800 Commissioners Road East, London, ON N6C 2V5, Canada; Lawson Health Research Institute, London, ON N6C 2R5, Canada; Department of Pediatrics, Western University, London, ON N6A 5C1, Canada; Department of Physiology & Pharmacology, Western University, London, ON N6A 5C1, Canada; Department of Clinical Neurological Sciences, Western University, London, ON N6A 5C1, Canada
| | - Gregory J Fonseca
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada.
| | - Simon Rousseau
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada.
| | - Jun Ding
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; School of Computer Science, McGill University, 3480 Rue University, Montreal, QC H3A 2A7, Canada; Mila-Quebec AI Institute, 6666 Rue Saint-Urbain, Montreal, QC H2S 3H1, Canada.
| |
Collapse
|
3
|
Obeagu EI, Obeagu GU. Exploring the intricate relationship between peptic ulcers and immunohematological responses: A narrative review. Medicine (Baltimore) 2025; 104:e42187. [PMID: 40228282 PMCID: PMC11999392 DOI: 10.1097/md.0000000000042187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Peptic ulcers have long been a focus of medical research due to their significant impact on public health worldwide. Traditionally attributed to factors such as Helicobacter pylori infection and excessive gastric acid secretion, recent scientific endeavors have increasingly unveiled the pivotal role of immunohematological responses in the pathogenesis and clinical course of peptic ulcers. This review aims to synthesize and analyze the intricate relationship between peptic ulcers and immunohematological responses, shedding light on the complex interplay between the immune system and ulcer development, progression, and healing. Immunological factors, encompassing inflammatory mediators, immune cells, and the host response to H pylori, play a substantial role in the multifaceted landscape of peptic ulcers. Inflammation orchestrated by cytokines and chemokines derived from immune cells intricately contributes to mucosal damage and repair processes. Moreover, the chronic nature of H pylori infection triggers a cascade of immune responses, involving both innate and adaptive immunity, which significantly influences the course of ulceration. This paper consolidates current knowledge while highlighting the need for further research elucidating the intricate immunological pathways involved in peptic ulcer pathogenesis. The integration of immunology into the broader context of peptic ulcer disease presents opportunities for innovative therapeutic interventions aimed at modulating immune responses for improved clinical outcomes and enhanced patient care. Ultimately, unraveling the intricate relationship between peptic ulcers and immunohematological responses holds significant promise in advancing the understanding and management of this prevalent gastrointestinal disorder.
Collapse
|
4
|
Pai SK, Chakraborty K, Pai AA, Dhara S, James MT. Seaweeds: Nature's super therapeutics? Immunomodulatory and anti-viral properties of sulfated rhamno xyloglucuronan isolated from Ulva fasciata Delile. Int J Biol Macromol 2025; 300:139978. [PMID: 39826735 DOI: 10.1016/j.ijbiomac.2025.139978] [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: 10/11/2024] [Revised: 12/19/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Green seaweeds, which constitute a significant portion of the global seaweed population, exhibit a wide range of therapeutic properties. The study aimed to isolate a (1 → 4) linked sulfated rhamno xyloglucuronan, designated as UFP-2, from the edible green seaweed Ulva fasciata Delile, and to evaluate its efficacy in modulating immune responses and inhibiting infection by the SARS-CoV-2 Delta variant. Anti-inflammatory potential of UFP-2 was demonstrated through the regulation of key cytokines involved in inflammatory responses triggered by viral infections, including interferons (IFN-α/γ), interleukin (IL-1β/12/33), and tumor necrosis factor (TNF-α). Confocal microscopy and flow cytometry analyses indicated downregulation of IFN-α and IL-1β, while TNF-α expression reduced from 29.28 % in lipopolysaccharide (LPS)-induced CALU-1 cells to 1.6-5.4 %, upon UFP-2 treatment. Treatment with UFP-2 at 125 μg/mL significantly downregulated the overexpression of IL-1β level in SARS-CoV-2-infected CALU-1 cells. Administering UFP-2 to SARS-CoV-2 (delta variant) induced cells led to a higher cycle threshold (Ct) values (20.34), indicating reduced viral load, with viral copy numbers decreasing from over 6.5 × 107 to <2.5 × 107 per cell. Structure-activity relationship analysis indicates that the sulfate groups and overall hydrophilicity of UFP-2 may enhance its binding affinity to target receptors, potentially disrupting viral entry and replication processes.
Collapse
Affiliation(s)
- Shilpa Kamalakar Pai
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin 682018, Kerala State, India; Department of Applied Chemistry, Cochin University of Science and Technology, South Kalamassery, Cochin, Kerala State, India
| | - Kajal Chakraborty
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin 682018, Kerala State, India.
| | - Ashwin Ashok Pai
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin 682018, Kerala State, India; Department of Chemistry, Mangalore University, Mangalagangothri 574199, Karnataka State, India
| | - Shubhajit Dhara
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin 682018, Kerala State, India; Department of Chemistry, Mangalore University, Mangalagangothri 574199, Karnataka State, India
| | - Mereeta Thundathil James
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin 682018, Kerala State, India
| |
Collapse
|
5
|
Yuan G, Mao J, Li Z. Systematically investigate the mechanism underlying the therapeutic effect of emodin in treatment of prostate cancer. Discov Oncol 2025; 16:413. [PMID: 40148580 PMCID: PMC11950527 DOI: 10.1007/s12672-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVE To systematically investigate the mechanism underlying the therapeutic effect of emodin in treatment of prostate cancer. METHODS Combine network pharmacology, molecular docking, molecular dynamics and experimental verification to explored the mechanism. Using the network pharmacology method, through the TCMSP, DisGeNET and Genecards database, the corresponding targets and related signaling pathways of emodin were screened, and emodin and core targets were studied by molecular docking and molecular dynamics uasing Cytoscape 3.7.2 and other software. The biological processes, cellular components and molecular functions of the key targets were determined by GO enrichment analysis. KEGG enrichment analysis identified signaling pathways associated with key targets. GEPIA and Kaplan-Meier database were used to determine the relationship between the expression of core genes in normal people and prostate cancer patients and the prognosis of patients. Cell proliferation inhibition experiment was carried out by MTT method. The mRNA and protein levels of CASP3, TNF, IL1B, TP53, PPARG, and MYC in PC-3 cells were evaluated by RT-PCR and WB method respectively. RESULTS There were 31 common targets which closely related to emodin in the treatment of prostate cancer. PPI network analysis showed that MYC, PPARG, TP53, TNF, CASP3, IL1B were the core targets. Go and KEGG enrichment analysis showed that pathways in cancer and IL-17 signaling pathway were the key pathways. Molecular docking and molecular dynamics results indicated that emodin had good binding to prostate cancer and 6 core proteins, and the binding force with TP53 protein was the strongest and most stable. The expression of CASP3 protein in normal people was stronger than that in patients with prostate cancer, and the expression of TP53 protein was closely related to the survival rate of patients with prostate cancer. Experimental verification result revealed that EM significantly increased mRNA expressions of CASP3, PPARG and decreased protein expressions of TNF, TP53, MYC at concentrations ranging from 0.1 to 1.6 μmol/L. Emodin significantly increased protein expressions of CASP3, PPARG and decreased protein expressions of TNF, TP53, MYC, IL1B at concentrations ranging from 10 to 160 µmol/L. CONCLUSION Emodin and TP53 have the best binding and stable conformation among core genes. Emodin exhibits a significant inhibitory effect on PC-3 cells at concentration 0.4 ~ 1.6 μmol/L. It showed that anti-prostate cancer properties by regulating cancer and 1L-17 signaling pathway through up-regulating the expressions of CASP3, PPARG genes/proteins, down-regulating IL1B, TP53, TNF, MYC genes/proteins.
Collapse
Affiliation(s)
- Gang Yuan
- Pharmacy Department, Jiulongpo Hospital of Chongqing University of Chinese Medicine, Chongqing Jiulongpo Traditional Chinese Medicine Hospital, Chongqing, 400050, China
| | - Jingxin Mao
- Chongqing Medical and Pharmaceutical College, No. 82, Middle Road of University Town, Shapingba District, Chongqing, 400030, China.
| | - Zheng Li
- Pharmacy Department, Chongqing University Affiliated Renji Hospital, No. 121 Wangxi Road, Nan'an District, Chongqing, 401336, China.
| |
Collapse
|
6
|
Satyanarayanan SK, Yip TF, Han Z, Zhu H, Qin D, Lee SMY. Role of toll-like receptors in post-COVID-19 associated neurodegenerative disorders? Front Med (Lausanne) 2025; 12:1458281. [PMID: 40206484 PMCID: PMC11979212 DOI: 10.3389/fmed.2025.1458281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
In the intricate realm of interactions between hosts and pathogens, Toll-like receptors (TLRs), which play a crucial role in the innate immune response, possess the ability to identify specific molecular signatures. This includes components originating from pathogens such as SARS-CoV-2, as well as the resulting damage-associated molecular patterns (DAMPs), the endogenous molecules released after cellular damage. A developing perspective suggests that TLRs play a central role in neuroinflammation, a fundamental factor in neurodegenerative conditions like Alzheimer's and Parkinson's disease (PD). This comprehensive review consolidates current research investigating the potential interplay between TLRs, their signaling mechanisms, and the processes of neurodegeneration following SARS-CoV-2 infection with an aim to elucidate the involvement of TLRs in the long-term neurological complications of COVID-19 and explore the potential of targeting TLRs as a means of implementing intervention strategies for the prevention or treatment of COVID-19-associated long-term brain outcomes.
Collapse
Affiliation(s)
- Senthil Kumaran Satyanarayanan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
| | - Tsz Fung Yip
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zixu Han
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
| | - Huachen Zhu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Suki Man Yan Lee
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Šutković J. Neutrophils and COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:347-384. [PMID: 40246349 DOI: 10.1016/bs.pmbts.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Neutrophils are the first line of defense against pathogens, most effectively by forming Neutrophil Extracellular Traps (NETs). Neutrophiles are further classified into several subpopulations during their development, eliminating pathogens through various mechanisms. However, due to the chaotic and uncontrolled immune response, NETs are often severely resulting in tissue damage and lung infections. The uncontrolled and poorly acknowledged host response regarding the cytokine storm is one of the major causes of severe COVID-19 conditions. Specifically, the increased formation of low-density neutrophils (LDNs), together with neutrophil extracellular traps (NETs) is closely linked with the severity and poor prognosis in patients with COVID-19. In this review, we discuss in detail the ontogeny of neutrophils at different stages and their recruitment and activation after infections, focusing on SARS-CoV-2. In addition, this chapter summarized the research progress on potential targeted drugs (NETs and Cytokine inhibitors) for neutrophil medical therapy and hoped to provide reference for the development of related therapeutic drugs for critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Jasmin Šutković
- Department Genetics and Bioegnineering, International University of Sarajevo, Hrasnička cesta, Bosnia & Herzegovina.
| |
Collapse
|
8
|
Liang KH, Chen YC, Hsu CY, Kao ZK, Tsai PH, Huang HY, Chu YC, Ho HL, Liao YC, Lee YC, Huang CC, Wei TC, Liao YJ, Lu YH, Kuo CT, Chiou SH. Predictive biosignatures for hospitalization in patients with virologically confirmed COVID-19. J Chin Med Assoc 2025; 88:246-252. [PMID: 39730204 DOI: 10.1097/jcma.0000000000001203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, presents with varying severity among individuals. Both viral and host factors can influence the severity of acute and chronic COVID-19, with chronic COVID-19 commonly referred to as long COVID. SARS-CoV-2 infection can be properly diagnosed by performing real-time reverse transcription polymerase chain reaction analysis of nasal swab samples. Pulse oximetry, chest X-ray, and complete blood count (CBC) analysis can be used to assess the condition of the patient to ensure that the appropriate medical care is delivered. This study aimed to develop biosignatures that can be used to distinguish between patients who are likely to develop severe disease and require hospitalization from patients who can be safely monitored in less intensive settings. METHODS A retrospective investigation was conducted on 7897 adult patients with virologically confirmed SARS-CoV-2 infection between January 26, 2020, and November 30, 2023; all patients underwent comprehensive CBC testing at Taipei Veterans General Hospital. Among them, 1867 patients were independently recruited for a population study involving genome-wide genotyping of approximately 424 000 genomic variants. Therefore, the participants were divided into two patient cohorts, one with genomic data (n = 1867) and one without (n = 6030) for model validation and training, respectively. RESULTS We constructed and validated a biosignature model by using a combination of CBC measurements to predict subsequent hospitalization events (hazard ratio = 3.38, 95% confidence interval: 3.07-3.73 for the training cohort and 3.03 [2.46-3.73] for the validation cohort; both p < 10 -8 ). The obtained scores were used to identify the top quartile of patients, who formed the "very high risk" group with a significantly higher cumulative incidence of hospitalization (log-rank p < 10 -8 in both the training and validation cohorts). The "very high risk" group exhibited a cumulative hospitalization rate of >60%, whereas the rate for the other patients was approximately 30% over a 1.5-year period, providing a binary classification of patients with distinct hospitalization risks. To investigate the genetic factors mediating this risk, we conducted a genome-wide association study. Specific regions in chromosomes 7 and 10 and the mitochondrial chromosome (M), harboring IKAROS family zinc finger 1 ( IKZF1 ), actin binding LIM protein 1 ( ABLIM1 ), and mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 3 ( MT-ND3 ), exhibited prominent associations with binary risk classification. The identified exonic variants of IKZF1 are linked to several autoimmune diseases. Notably, people with different genotypes of the leading variants (rs4132601, rs141492519, and Affx-120744614) exhibited varying cumulative hospitalization rates after infection. CONCLUSION We successfully developed and validated a biosignature model of COVID-19 severe disease in virologically confirmed patients. The identified genomic variants provide new insights for infectious disease research and medical care.
Collapse
Affiliation(s)
- Kung-Hao Liang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Biosafety Level 3 Laboratory, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- College of Medicine, Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- School of Pharmaceutical Sciences, Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Chun Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Yuli Branch, Hualien, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chun-Yi Hsu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Zih-Kai Kao
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hsin-Yi Huang
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yuan-Chia Chu
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, ROC
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Chu Liao
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
- Department of Neurology, School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Brain Research Center, National Yang Ming Chao Tung University School of Medicine, Taipei, Taiwan, ROC
| | - Yi-Chung Lee
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
- Department of Neurology, School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Brain Research Center, National Yang Ming Chao Tung University School of Medicine, Taipei, Taiwan, ROC
| | - Chi-Cheng Huang
- Department of Surgery, Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- College of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Tzu-Chun Wei
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Urology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Shu-Tien Urological Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Jia Liao
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
| | - Yung-Hsiu Lu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chen-Tsung Kuo
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
9
|
Jahantigh HR, Elsharkawy A, Guglani A, Arora K, Patterson LD, Kumar M. Neurobiological Alterations Induced by SARS-CoV-2: Insights from Variant-Specific Host Gene Expression Patterns in hACE2-Expressing Mice. Viruses 2025; 17:329. [PMID: 40143258 PMCID: PMC11946589 DOI: 10.3390/v17030329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Since the onset of the COVID-19 pandemic, various severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants have emerged. Although the primary site of SARS-CoV-2 infection is the lungs, it can also affect the brain and induce neurological symptoms. However, the specific effects of different variants on the brain remain unclear. In this study, a whole-transcriptome analysis was conducted using the brain tissues of K18-hACE2 mice infected with the ancestral B.1 (Wuhan) variant and with major SARS-CoV-2 variants of concern, including B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 (Delta) and B.1.529 (Omicron). After sequencing, differential gene expression, gene ontology (GO) and genome pathway enrichment analyses were performed. An Immune Cell Abundance Identifier (ImmuCellAI) was used to identify the abundance of different cell populations. Additionally, RT-qPCR was used to validate the RNA-seq data. The viral load and hierarchical clustering analyses divided the samples into two different clusters with notable differences in gene expression at day 6 post-infection for all variants compared to the control group. GO and the Kyoto Encyclopedia of genes and genomes enrichment analyses revealed similar patterns of pathway enrichment for different variants. ImmuCellAI revealed the changes in immune cell populations, including the decrease in CD4+ T and B cell proportions and the increase in CD8+ T and dendritic cell proportions. A co-expression network analysis revealed that some genes, such as STAT1, interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α), were dysregulated in all variants. A RT-qPCR analysis for IL-6, CXCL10 and IRF7 further validated the RNA-seq analysis. In conclusion, this study provides, for the first time, an extensive transcriptome analysis of a K18-hACE2 mouse brain after infection with major SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Hamid Reza Jahantigh
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
| | - Amany Elsharkawy
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
- Center of Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Anchala Guglani
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
| | - Komal Arora
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
| | - Lila D. Patterson
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
- Center of Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
10
|
Warner BM, Vendramelli R, Boese AS, Audet J, Tailor N, Meilleur C, Glowach N, Willman M, Truong T, Moffat E, Tierney K, Kosak B, Dhanidina I, Engstrom J, Korczak B, McGowan I, Embury-Hyatt C, Kobasa D. Treatment with the CCR5 antagonist OB-002 reduces lung pathology, but does not prevent disease in a Syrian hamster model of SARS-CoV-2 infection. PLoS One 2025; 20:e0316952. [PMID: 39908288 PMCID: PMC11798459 DOI: 10.1371/journal.pone.0316952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/18/2024] [Indexed: 02/07/2025] Open
Abstract
Since the emergence of SARS-CoV-2 and the COVID-19 pandemic, a wide range of treatment options have been evaluated in preclinical studies and clinical trials, with several being approved for use in humans. Immunomodulatory drugs have shown success in dampening the deleterious inflammatory response seen in severe COVID-19 patients, but there remains an urgent need for development of additional therapeutic options for COVID-19 treatment. A potential drug target is the CCR5-CCL5 axis, and blocking this pathway may protect against severe disease. Here we evaluated whether OB-002, an analog of human CCL5 and a potent antagonist of CCR5, provides therapeutic benefit in SARS-CoV-2 infected Syrian hamsters. Daily treatment with OB-002 altered immune gene transcription in the lungs, and reduced pathology following infection, but did not prevent weight loss or viral replication in the lungs of infected animals, even in combination with the antiviral drug remdesivir. Our data suggest that targeting the CCR5-CCL5 pathway in SARS-CoV-2 infection in hamsters is insufficient to significantly impact disease development in this model.
Collapse
Affiliation(s)
- Bryce M. Warner
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Robert Vendramelli
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Amrit S. Boese
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Jonathan Audet
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Nikesh Tailor
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Courtney Meilleur
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Nathan Glowach
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Marnie Willman
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Thang Truong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Estella Moffat
- National Centre for Foreign Animal Diseases, Canadian Food Inspection Agency, Winnipeg, Manitoba, Canada
| | - Kevin Tierney
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | - Carissa Embury-Hyatt
- National Centre for Foreign Animal Diseases, Canadian Food Inspection Agency, Winnipeg, Manitoba, Canada
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
11
|
Obeagu EI. Influence of cytokines on the recovery trajectory of HIV patients on antiretroviral therapy: A review. Medicine (Baltimore) 2025; 104:e41222. [PMID: 40184131 PMCID: PMC11709159 DOI: 10.1097/md.0000000000041222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/26/2024] [Accepted: 12/18/2024] [Indexed: 04/05/2025] Open
Abstract
Cytokines, critical signaling molecules in the immune system, significantly influence the pathophysiology of Human Immunodeficiency Virus (HIV) infection and the effectiveness of antiretroviral therapy (ART). Dysregulated cytokine production, characterized by elevated pro-inflammatory and anti-inflammatory cytokines, plays a pivotal role in chronic inflammation and immune activation in untreated HIV patients. ART initiation leads to changes in cytokine levels, typically resulting in decreased systemic inflammation, though the extent and persistence of these changes vary among individuals. Despite successful viral suppression with ART, many HIV patients experience persistent immune activation and inflammation, driven by ongoing cytokine dysregulation. This persistent inflammatory state is associated with adverse clinical outcomes, including cardiovascular disease, neurocognitive impairment, and non-AIDS-related cancers. Understanding the specific cytokine profiles that contribute to these outcomes is crucial for developing targeted therapeutic interventions to improve long-term health. Cytokine modulation presents a promising avenue for enhancing immune recovery and reducing chronic inflammation in HIV patients on ART. Identifying cytokine patterns that serve as biomarkers for disease progression and treatment response can help tailor individualized treatment strategies. Future research should focus on adjunctive therapies that target cytokine activity to mitigate residual inflammation, thereby improving the overall health and quality of life for HIV patients.
Collapse
|
12
|
Vasquez R, Song JH, Mendoza RM, Hwang I, Bagon BB, Engstrand L, Valeriano VD, Kang D. Oral Immunisation With Non-GMO Surface Displayed SARS-CoV-2 Spike Epitopes on Bacteria-Like Particles Provokes Robust Humoral and Cellular Immune Responses, and Modulated the Gut Microbiome in Mice. Microb Biotechnol 2025; 18:e70073. [PMID: 39797809 PMCID: PMC11724470 DOI: 10.1111/1751-7915.70073] [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: 09/10/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) is a fatal disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). To date, several vaccines have been developed to combat the spread of this virus. Mucosal vaccines using food-grade bacteria, such as Lactobacillus spp., are promising strategies for developing safe and effective vaccines against SARS-CoV-2. In this study, we designed a non-GMO surface-displayed SARS-CoV-2 spike S1 epitope on Limosilactobacillus fermentum-derived bacteria-like particles (BLPs). After that, we evaluated its efficacy to induce immune responses in immunocompetent mice. Moreover, we examined the influence of oral immunisation on the gut microbiome and microbiota metabolites. Twenty-eight 6-week-old male C57BL/6 mice were orally immunised with the following: PBS (control), Lm. fermentum-derived BLPs only, BLPs displaying SARS-CoV-2 spike S1-2, or BLPs displaying SARS-CoV-2 spike S1-3 epitopes. Our results showed that mucosal immunisation of mice with surface-displayed SARS-CoV-2 spike epitopes provoked high-level secretory IgA and systemic IgG production. Moreover, the immunisation exhibited a Th1-like immune response, characterised by an elevated IgG2a-to-IgG1 ratio and high antiviral IFN-γ production. In addition, we observed gut microbiome modulation and increased butyrate production in immunised mice. Overall, the use of Lm. fermentum-derived BLPs and the anchor CshA to display SARS-CoV-2 spike S1epitopes is a promising novel strategy in developing a cost-effective, non-GMO mucosal vaccine alternative against SARS-CoV-2.
Collapse
Affiliation(s)
- Robie Vasquez
- Department of Animal BiotechnologyDankook UniversityCheonanKorea
| | - Ji Hoon Song
- Department of Animal BiotechnologyDankook UniversityCheonanKorea
| | | | - In‐Chan Hwang
- Department of Animal BiotechnologyDankook UniversityCheonanKorea
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR)Karolinska InstitutetStockholmSweden
| | | | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR)Karolinska InstitutetStockholmSweden
| | - Valerie Diane Valeriano
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR)Karolinska InstitutetStockholmSweden
| | - Dae‐Kyung Kang
- Department of Animal BiotechnologyDankook UniversityCheonanKorea
| |
Collapse
|
13
|
Kombe Kombe AJ, Fotoohabadi L, Gerasimova Y, Nanduri R, Lama Tamang P, Kandala M, Kelesidis T. The Role of Inflammation in the Pathogenesis of Viral Respiratory Infections. Microorganisms 2024; 12:2526. [PMID: 39770727 PMCID: PMC11678694 DOI: 10.3390/microorganisms12122526] [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: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Viral respiratory infections (VRIs) are a leading cause of morbidity and mortality worldwide, making them a significant public health concern. During infection, respiratory viruses, including Influenza virus, SARS-CoV-2, and respiratory syncytial virus (RSV), trigger an antiviral immune response, specifically boosting the inflammatory response that plays a critical role in their pathogenesis. The inflammatory response induced by respiratory viruses can be a double-edged sword since it can be initially induced to be antiviral and protective/reparative from virus-induced injuries. Still, it can also be detrimental to host cells and tissues. However, the mechanisms that differentiate the complex crosstalk between favorable host inflammatory responses and harmful inflammatory responses are poorly understood. This review explores the complex interplay between viral pathogens and the host immune response, mainly focusing on the role of inflammation in the pathogenesis of VRIs. We discuss how inflammation can both contain and exacerbate the progression of viral infections, highlighting potential therapeutic targets and emerging drugs for modulating the aberrant inflammatory responses during VRIs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Theodoros Kelesidis
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine and Infectious Diseases, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
14
|
Zhu R, Zhao Y, Yin H, Shu L, Ma Y, Tao Y. Identification of immune-related hub genes and potential molecular mechanisms involved in COVID-19 via integrated bioinformatics analysis. Sci Rep 2024; 14:29964. [PMID: 39622956 PMCID: PMC11612211 DOI: 10.1038/s41598-024-81803-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024] Open
Abstract
COVID-19, caused by the SARS-CoV-2 virus, poses significant health challenges worldwide, particularly due to severe immune-related complications. Understanding the molecular mechanisms and identifying key immune-related genes (IRGs) involved in COVID-19 pathogenesis is critical for developing effective prevention and treatment strategies. This study employed computational tools to analyze biological data (bioinformatics) and a method for inferring causal relationships based on genetic variations, known as Mendelian randomization (MR), to explore the roles of IRGs in COVID-19. We identified differentially expressed genes (DEGs) from datasets available in the Gene Expression Omnibus (GEO), comparing COVID-19 patients with healthy controls. IRGs were sourced from the ImmPort database. We conducted functional enrichment analysis, pathway analysis, and immune infiltration assessments to determine the biological significance of the identified IRGs. A total of 360 common differential IRGs were identified. Among these genes, CD1C, IL1B, and SLP1 have emerged as key IRGs with potential protective effects against COVID-19. Pathway enrichment analysis revealed that CD1C is involved in terpenoid backbone biosynthesis and Th17 cell differentiation, while IL1B is linked to B-cell receptor signaling and the NF-kappa B signaling pathway. Significant correlations were observed between key genes and various immune cells, suggesting that they influence immune cell modulation in COVID-19. This study provides new insights into the immune mechanisms underlying COVID-19, highlighting the crucial role of IRGs in disease progression. These findings suggest that CD1C and IL1B could be potential therapeutic targets. The integrated bioinformatics and MR analysis approach offers a robust framework for further exploring immune responses in COVID-19 patients, as well as for targeted therapy and vaccine development.
Collapse
Affiliation(s)
- Rui Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yaping Zhao
- Department of Pharmacy, Shaoxing Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Shaoxing, 312000, China
| | - Hui Yin
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Linfeng Shu
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yuhang Ma
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yingli Tao
- Department of Reproductive Immunology, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China.
| |
Collapse
|
15
|
Seethamraju H, Yang OO, Loftus R, Ogbuagu O, Sammartino D, Mansour A, Sacha JB, Ojha S, Hansen SG, Arman AC, Lalezari JP. A Randomized Placebo-Controlled Trial of Leronlimab in Mild-To-Moderate COVID-19. Clin Ther 2024; 46:891-899. [PMID: 39353749 DOI: 10.1016/j.clinthera.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE Early in the course of the SARS-CoV-2 pandemic it was hypothesised that host genetics played a role in the pathophysiology of COVID-19 including a suggestion that the CCR5-Δ32 mutation may be protective in SARS-CoV-2 infection. Leronlimab is an investigational CCR5-specific humanized IgG4 monoclonal antibody currently in development for HIV-1 infection. We aimed to explore the impact of leronlimab on the severity of disease symptoms among participants with mild-to-moderate COVID-19. METHODS The TEMPEST trial was a randomized, double-blind, placebo-controlled study in participants with mild-to-moderate COVID-19. Participants were randomly assigned in a 2:1 ratio to receive subcutaneous leronlimab (700 mg) or placebo on days 0 and 7. The primary efficacy endpoint was assessed by change in total symptom score based on fever, myalgia, dyspnea, and cough, at end of treatment (day 14). FINDINGS Overall, 84 participants were randomized and treated with leronlimab (n = 56) or placebo (n = 28). No difference was observed in change in total symptom score (P = 0.8184) or other pre-specified secondary endpoints between treatments. However, in a post hoc analysis, 50.0% of participants treated with leronlimab demonstrated improvements from baseline in National Early Warning Score 2 (NEWS2) at day 14, compared with 20·8% of participants in the placebo group (post hoc; p = 0.0223). Among participants in this trial with mild-to-moderate COVID-19 adverse events rates were numerically but not statistically significantly lower in leronlimab participants (33.9%) compared with placebo participants (50.0%). IMPLICATIONS At the time the TEMPEST trial was designed although CCR5 was known to be implicated in COVID-19 disease severity the exact pathophysiology of SARS-CoV-2 infection was poorly understood. Today it is well accepted that SARS-CoV-2 infection in asymptomatic-to-mild cases is primarily characterized by viral replication, with a heightened immune response, accompanied by diminished viral replication in moderate-to-severe disease and a peak in inflammatory responses with excessive production of pro-inflammatory cytokines in critical disease. It is therefore perhaps not surprising that no differences between treatments were observed in the primary endpoint or in pre-specified secondary endpoints among participants with mild-to-moderate COVID-19. However, the results of the exploratory post hoc analysis showing that participants in the leronlimab group had greater improvement in NEWS2 assessment compared to placebo provided a suggestion that leronlimab may be associated with a lower likelihood of people with mild-to-moderate COVID-19 progressing to more severe disease and needs to be confirmed in other appropriately designed clinical trials. CLINICALTRIALS gov number, NCT04343651 https://classic. CLINICALTRIALS gov/ct2/show/NCT04343651.
Collapse
Affiliation(s)
| | - Otto O Yang
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | | | | | | | - Jonah B Sacha
- Oregon Health & Science University, Portland, Oregon
| | - Sohita Ojha
- Oregon Health & Science University, Portland, Oregon
| | | | | | | |
Collapse
|
16
|
Al-Sammarraie MR, Al-Sammarraie MR, Azaiez F, Al-Rubae ZMM, Litaiem H, Taay YM. mRNA vaccination reduces the thrombotic possibility in COVID-19: Inflammation risk estimates. Int Immunopharmacol 2024; 140:112776. [PMID: 39079343 DOI: 10.1016/j.intimp.2024.112776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/08/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024]
Abstract
Thrombosis is a common clinical feature associated with morbidity and mortality in coronavirus disease-2019 (COVID-19) patients. Cytokine storm in COVID-19 increases patients' systemic inflammation, which can cause multiple health consequences. In this work, we aimed to indicate the effect of Pfizer-BioNTech vaccination on the modulation of monocyte chemoattractant protein-3 (MCP-3), matrix metalloproteinase 1 (MMP-1), and tumor necrosis factor-alpha (TNF-α) levels, and other systemic inflammatory biomarkers that associates with COVID-19 severity in patients who suffers from thrombosis consequences. For this purpose, ninety people were collected from Ibn Al-Nafees Hospital and divided into three groups each of which contained 30 people, 15 of them were venous thromboembolism (VTE) positive and the other were VTE negative. The three groups were non-vaccinated COVID-19, vaccinated COVID-19, and control. The levels of MCP-3 and TNF-α were significantly (p < 0.05) increased in vaccinated and non-vaccinated COVID-19 patients regardless of their thrombosis condition, while MMP-1 level was non-significantly (p > 0.05) higher in vaccinated patients compared to control. MCP-3 and TNF-α were correlated positively with D-dimer (r = 0.544 and r = 0.513, respectively) in non-vaccinated patients, while MMP-1 and TNF-α were correlated positively with D-dimer (r = 0.624 and r = 0.575, respectively) in vaccinated patients. The odds ratio of MCP-3 (2.252), MMP-1 (1.062), and TNF-α (1.360) were reduced in vaccinated patients (2.093, 1.022, and 1.301 for MCP-3, MMP-1, and TNF-α respectively). Thus, MCP-3 plays a vital role in COVID-19 pathophysiology, and vaccination can reduce the risk of developing VTE in COVID-19 patients, and improve the inflammatory condition of patients.
Collapse
Affiliation(s)
- Marwah Raad Al-Sammarraie
- Department of Chemistry, College of Science for Women, University of Baghdad, Baghdad, Iraq; Laboratory of Inorganic Chemistry, Faculty of Sciences, University of Sfax, Tunisia.
| | | | - Fatma Azaiez
- Laboratory Clinical Virology Pasteur Institute and Department of Toxicology, Faculty of Pharmacy Monastir, Tunisia
| | - Zeinab M M Al-Rubae
- Department of Chemistry, College of Education for Pure Science, University of Baghdad, Baghdad, Iraq
| | - Hejer Litaiem
- Laboratory of Inorganic Chemistry, Faculty of Sciences, University of Sfax, Tunisia
| | - Yasser M Taay
- Department of Chemistry, College of Science, University of Baghdad, Baghdad, Iraq.
| |
Collapse
|
17
|
Zhu Z, Ling X, Wang G, Xie J. G-CSFR-induced leukocyte transendothelial migration during the inflammatory response is regulated by the ICAM1-PKCa axis: based on multiomics integration analysis. Cell Biol Toxicol 2024; 40:90. [PMID: 39433604 PMCID: PMC11493794 DOI: 10.1007/s10565-024-09934-w] [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: 06/24/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024]
Abstract
As an indispensable inflammatory mediator during sepsis, granulocyte colony-stimulating factor (G-CSF) facilitates neutrophil production by activating G-CSFR. However, little is known about the role of intracellular downstream signalling pathways in the induction of inflammation. To explore the functions of molecules in regulating G-CSFR signalling, RNA sequencing and integrated proteomic and phosphoproteomic analyses were conducted to predict the differentially expressed molecules in modulating the inflammatory response after G-CSFR expression was either up- or downregulated, in addition to the confirmation of their biological function by diverse experimental methods. In the integrated bioinformatic analysis, 3190 differentially expressed genes (DEGs) and 1559 differentially expressed proteins (DEPs) were identified in multiple-group comparisons (p < 0.05, FC > ± 1.5) using enrichment analyses, as well as those classic pathways such as the TNF, NFkappaB, IL-17, and TLR signalling pathways. Among them, 201 proteins, especillay intercellular cell adhesion molecule-1 (ICAM1) and PKCa, were identified as potential molecules involved in inflammation according to the protein-protein interaction (PPI) analysis, and the leukocyte transendothelial migration (TEM) pathway was attributed to the intervention of G-CSFR. Compared with the control and TNF-a treatment, the G-CSFR (G-CSFROE)-overexpressing led to an obvious increase in the number of leukocytes with the TEM phenotype. Mechanically, the expression of ICAM1 and PKCa was significantly up- and downregulated by G-CSFROE, which directly led to increased TEM; moreover, PKCa expression was negatively regulated by ICAM1 expression, leading to aberrant leukocyte TEM. Altogether, the ICAM1‒PKCa axis was found a meaningful target in the leukocyte TEM induced by G-CSFR upregulation.
Collapse
Affiliation(s)
- Zhipeng Zhu
- Department of Anesthesiology, Run Run Shaw Hospital, Zhejiang University School of Medicine, Shangcheng District, Qingchun East Road 3, Hangzhou, 310016, China.
- The Second Affiliated Hospital of Jiaxing University, Zhejiang, 314000, China.
| | - Xiaoyan Ling
- Department of Outpatient Nursing, the Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Gaojian Wang
- Department of Anesthesiology, Run Run Shaw Hospital, Zhejiang University School of Medicine, Shangcheng District, Qingchun East Road 3, Hangzhou, 310016, China
| | - Junran Xie
- Department of Anesthesiology, Run Run Shaw Hospital, Zhejiang University School of Medicine, Shangcheng District, Qingchun East Road 3, Hangzhou, 310016, China.
| |
Collapse
|
18
|
Vuitika L, Côrtes N, Malaquias VB, Silva JDQ, Lira A, Prates-Syed WA, Schimke LF, Luz D, Durães-Carvalho R, Balan A, Câmara NOS, Cabral-Marques O, Krieger JE, Hirata MH, Cabral-Miranda G. A self-adjuvanted VLPs-based Covid-19 vaccine proven versatile, safe, and highly protective. Sci Rep 2024; 14:24228. [PMID: 39414952 PMCID: PMC11484777 DOI: 10.1038/s41598-024-76163-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024] Open
Abstract
Vaccination has played a critical role in mitigating COVID-19. Despite the availability of licensed vaccines, there remains a pressing need for improved vaccine platforms that provide high protection, safety, and versatility, while also reducing vaccine costs. In response to these challenges, our aim is to create a self-adjuvanted vaccine against SARS-CoV-2, utilizing Virus-Like Particles (VLPs) as the foundation. To achieve this, we produced bacteriophage (Qβ) VLPs in a prokaryotic system and purified them using a rapid and cost-effective strategy involving organic solvents. This method aims to solubilize lipids and components of the cell membrane to eliminate endotoxins present in bacterial samples. For vaccine formulation, Receptor Binding Domain (RBD) antigens were conjugated using chemical crosslinkers, a process compatible with Good Manufacturing Practice (GMP) standards. Transmission Electron Microscopy (TEM) confirmed the expected folding and spatial configuration of the QβVLPs vaccine. Additionally, vaccine formulation assessment involved SDS-PAGE stained with Coomassie Brilliant Blue, Western blotting, and stereomicroscopic experiments. In vitro and in vivo evaluations of the vaccine formulation were conducted to assess its capacity to induce a protective immune response without causing side effects. Vaccine doses of 20 µg and 50 µg stimulated the production of neutralizing antibodies. In in vivo testing, the group of animals vaccinated with 50 µg of vaccine formulation provided complete protection against virus infection, maintaining stable body weight without showing signs of disease. In conclusion, the QβVLPs-RBD vaccine has proven to be effective and safe, eliminating the necessity for supplementary adjuvants and offering a financially feasible approach. Moreover, this vaccine platform demonstrates flexibility in targeting Variants of Concern (VOCs) via established conjugation protocols with VLPs.
Collapse
Affiliation(s)
- Larissa Vuitika
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nelson Côrtes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vanessa B Malaquias
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jaqueline D Q Silva
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Aline Lira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Wasim A Prates-Syed
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29., University of São Paulo School of Medicine, São Paulo, Brazil
| | - Daniela Luz
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
| | - Ricardo Durães-Carvalho
- São Paulo School of Medicine, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
- Interunit Bioinformatics Graduate Program, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Andrea Balan
- Applied Structural Biology Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Niels O S Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29., University of São Paulo School of Medicine, São Paulo, Brazil
- DO'R Institute for research, São Paulo, Brazil, IDOR, São Paulo, Brazil
| | - José E Krieger
- Heart Institute, Clinical Hospital, Faculty of Medicine, Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mario H Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil.
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
19
|
Obeagu EI, Tukur M, Akaba K. Impacts of COVID-19 on hemostasis: coagulation abnormalities and management perspectives. Ann Med Surg (Lond) 2024; 86:5844-5850. [PMID: 39359765 PMCID: PMC11444586 DOI: 10.1097/ms9.0000000000002237] [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: 01/04/2024] [Accepted: 05/22/2024] [Indexed: 10/04/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has transcended its initial characterization as a respiratory illness, revealing substantial implications for hemostasis and coagulation pathways. COVID-19-associated coagulopathies have emerged as critical determinants of disease severity and prognosis, presenting a multifaceted challenge in clinical management. This paper aims to elucidate the intricate interplay between COVID-19 and hemostasis, delving into the underlying mechanisms of coagulation abnormalities, exploring the spectrum of thrombotic complications, and discussing evolving management strategies. Therapeutic interventions and anticoagulation strategies tailored for managing COVID-19-related coagulopathies form a significant focus, encompassing prophylactic and therapeutic approaches, heparin-based therapies, and individualized treatment paradigms. This paper underscores the imperative for ongoing research endeavors to refine diagnostic modalities, identify novel therapeutic targets, and ascertain long-term sequelae of COVID-19-induced coagulation abnormalities. Ultimately, a comprehensive understanding of the intricate relationship between COVID-19 and hemostasis is pivotal in devising effective management strategies to mitigate thrombotic risks, improve clinical outcomes, and pave the way for tailored interventions in affected individuals.
Collapse
Affiliation(s)
| | - Muhammad Tukur
- Department of Science Education, Faculty of Education, Kampala International University, Kampala, Uganda
| | - Kingsley Akaba
- Department of Haematology, University of Calabar, Calabar, Cross-River State, Nigeria
| |
Collapse
|
20
|
Aljuaid A. Increased Activation Markers of Adaptive Immunity in Patients with Severe COVID-19. J Clin Med 2024; 13:5664. [PMID: 39407725 PMCID: PMC11477269 DOI: 10.3390/jcm13195664] [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: 06/16/2024] [Revised: 09/04/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Introduction: COVID-19 is a pandemic disease and is widespread over the world. This disease shows a 5.1% mortality. The understanding of the disease has expanded rapidly in many areas, including virological, epidemiological, clinical, and management dimensions. To better understand the inflammatory and immune profiles that impact the pathogenesis and development of severe COVID-19 symptoms, further studies are essential. This research aims to explore the inflammatory and adaptive immune responses associated with COVID-19, considering factors such as genetic diversity and environmental exposure among Saudi patients. The goal is to determine if patients with severe COVID-19 exhibit different disease phenotypes. Materials and Methods: This case-control study includes 115 participants (healthy and with COVID-19 infection), 55 of which had confirmed cases of COVID-19 in intensive care units (ICUs) at different hospitals in Makkah City, Saudi Arabia. Whole blood samples were collected from June to September 2021 for cellular analyses, and inflammation marker data were collected from hospital records. The expression of activation markers on B (CD27 and CD38) and T cells (CD27 and HLA-DR) was obtained using the flow cytometry technique. Also, serum was collected for cytokine measurements, including IL-6, INF-γ, and TNF- α. Results: The results indicated that lymphopenia and excessive T cell activation were more prevalent in severe cases than in healthy individuals. Furthermore, the results revealed that severe COVID-19 patients had an increased frequency of CD19+ B cells, with changes in B cell subsets. The current study implies impairment and changes in the phenotype of adaptive cells (including T and B cells), with an increase in HLA-DR molecules and inflammation markers with pro-inflammatory cytokines in severe COVID-19 cases. Conclusions: The current study implies impairment and changes in the phenotype of adaptive cells (including T and B cells), with an increase in HLA-DR molecules and inflammation markers in severe COVID-19 cases, which could be targeted for therapeutic interventions. This might be a valuable approach for the diagnosis and treatment of severe COVID-19 cases.
Collapse
Affiliation(s)
- Abdulelah Aljuaid
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
21
|
Chikopela T, Mwesigwa N, Masenga SK, Kirabo A, Shibao CA. The Interplay of HIV and Long COVID in Sub-Saharan Africa: Mechanisms of Endothelial Dysfunction. Curr Cardiol Rep 2024; 26:859-871. [PMID: 38958890 DOI: 10.1007/s11886-024-02087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Long COVID affects approximately 5 million people in Africa. This disease is characterized by persistent symptoms or new onset of symptoms after an acute SARS-CoV-2 infection. Specifically, the most common symptoms include a range of cardiovascular problems such as chest pain, orthostatic intolerance, tachycardia, syncope, and uncontrolled hypertension. Importantly, these conditions appear to have endothelial dysfunction as the common denominator, which is often due to impaired nitric oxide (NO) mechanisms. This review discusses the role of mechanisms contributing to endothelial dysfunction in Long COVID, particularly in people living with HIV. RECENT FINDINGS Recent studies have reported that increased inflammation and oxidative stress, frequently observed in Long COVID, may contribute to NO dysfunction, ultimately leading to decreased vascular reactivity. These mechanisms have also been reported in people living with HIV. In regions like Africa, where HIV infection is still a major public health challenge with a prevalence of approximately 26 million people in 2022. Specifically, endothelial dysfunction has been reported as a major mechanism that appears to contribute to cardiovascular diseases and the intersection with Long COVID mechanisms is of particular concern. Further, it is well established that this population is more likely to develop Long COVID following infection with SARS-CoV-2. Therefore, concomitant infection with SARS-CoV-2 may lead to accelerated cardiovascular disease. We outline the details of the worsening health problems caused by Long COVID, which exacerbate pre-existing conditions such as endothelial dysfunction. The overlapping mechanisms of HIV and SARS-CoV-2, particularly the prolonged inflammatory response and chronic hypoxia, may increase susceptibility to Long COVID. Addressing these overlapping health issues is critical as it provides clinical entry points for interventions that could improve and enhance outcomes and quality of life for those affected by both HIV and Long COVID in the region.
Collapse
Affiliation(s)
- Theresa Chikopela
- Department of Human Physiology, Faculty of Medicine, Lusaka Apex Medical University, Lusaka, Zambia
| | - Naome Mwesigwa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Sepiso K Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone, Zambia
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Cyndya A Shibao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA.
| |
Collapse
|
22
|
Yu Q, Zhou X, Kapini R, Arsecularatne A, Song W, Li C, Liu Y, Ren J, Münch G, Liu J, Chang D. Cytokine Storm in COVID-19: Insight into Pathological Mechanisms and Therapeutic Benefits of Chinese Herbal Medicines. MEDICINES (BASEL, SWITZERLAND) 2024; 11:14. [PMID: 39051370 PMCID: PMC11270433 DOI: 10.3390/medicines11070014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/20/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Cytokine storm (CS) is the main driver of SARS-CoV-2-induced acute respiratory distress syndrome (ARDS) in severe coronavirus disease-19 (COVID-19). The pathological mechanisms of CS are quite complex and involve multiple critical molecular targets that turn self-limited and mild COVID-19 into a severe and life-threatening concern. At present, vaccines are strongly recommended as safe and effective treatments for preventing serious illness or death from COVID-19. However, effective treatment options are still lacking for people who are at the most risk or hospitalized with severe disease. Chinese herbal medicines have been shown to improve the clinical outcomes of mild to severe COVID-19 as an adjunct therapy, particular preventing the development of mild to severe ARDS. This review illustrates in detail the pathogenesis of CS-involved ARDS and its associated key molecular targets, cytokines and signalling pathways. The therapeutic targets were identified particularly in relation to the turning points of the development of COVID-19, from mild symptoms to severe ARDS. Preclinical and clinical studies were reviewed for the effects of Chinese herbal medicines together with conventional therapies in reducing ARDS symptoms and addressing critical therapeutic targets associated with CS. Multiple herbal formulations, herbal extracts and single bioactive phytochemicals with or without conventional therapies demonstrated strong anti-CS effects through multiple mechanisms. However, evidence from larger, well-designed clinical trials is lacking and their detailed mechanisms of action are yet to be well elucidated. More research is warranted to further evaluate the therapeutic value of Chinese herbal medicine for CS in COVID-19-induced ARDS.
Collapse
Affiliation(s)
- Qingyuan Yu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
- Xiyuan Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Rotina Kapini
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Anthony Arsecularatne
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Wenting Song
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Chunguang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Yang Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Junguo Ren
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Gerald Münch
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| |
Collapse
|
23
|
Jian Z, Jiang C, Zhu L, Li F, Deng L, Ai Y, Lai S, Xu Z. Infectivity and pathogenesis characterization of getah virus (GETV) strain via different inoculation routes in mice. Heliyon 2024; 10:e33432. [PMID: 39040396 PMCID: PMC11260979 DOI: 10.1016/j.heliyon.2024.e33432] [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: 10/30/2023] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
In recent years, the epidemiological profile of Getah virus (GETV) has become increasingly serious, posing a huge threat to animal and public health in China. GETV can cause multi-species infection, including horses, pigs, rats, cattle, kangaroos, reptiles and birds. However, there were few reports on the efficiency of the virus entering the host via routes of different systems. In the present study, a GETV strain (SC201807) was obtained from a piglet's blood in 2018 in Sichuan, China. First, we established a quantitative real-time polymerase chain reaction (qRT-PCR) SYBR assay specific to GETV. Then, we evaluated the infection efficiency of different routes using mouse animal model. 108 male mice were randomly divided into four groups as follows: intramuscular, intraoral and intranasal infection routes, and negative control. All mice in the experimental group were inoculated with 4 × 102.85 TCID50 GETV virus. Tissue tropism experiments show that GETV has a wide range of tissue distribution, and intramuscular infection is the first to infect all tissues of the body, and suggest that oral infection may be a new GETV transmission route. Histopathological examination results showed that intramuscular injection of GETV mainly caused different degrees of pathological damage to the tissues, and could rapidly induce a large amount of inflammatory regulatory factors such as IL-6 and TNF-α. Our data may help us to evaluate the risk of transmission of Porcine Getah virus and provide an experimental basis for the prevention and control of Porcine Getah virus.
Collapse
Affiliation(s)
- Zhijie Jian
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Chaoyuan Jiang
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
- Chengdu Zhongji Agriculture and Animal Husbandry Co., Ltd, No. 37, Middle Section, Heshan Street, Pujiang County, Chengdu, Sichuan Province, China
| | - Ling Zhu
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Fengqin Li
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
- College of Animal Science, Xichang University, Xichang, 615000, Sichuan, China
| | - Lishuang Deng
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Yanru Ai
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Siyuan Lai
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Zhiwen Xu
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| |
Collapse
|
24
|
de Antonellis P, Ferrucci V, Miceli M, Bibbo F, Asadzadeh F, Gorini F, Mattivi A, Boccia A, Russo R, Andolfo I, Lasorsa VA, Cantalupo S, Fusco G, Viscardi M, Brandi S, Cerino P, Monaco V, Choi DR, Cheong JH, Iolascon A, Amente S, Monti M, Fava LL, Capasso M, Kim HY, Zollo M. Targeting ATP2B1 impairs PI3K/Akt/FOXO signaling and reduces SARS-COV-2 infection and replication. EMBO Rep 2024; 25:2974-3007. [PMID: 38816514 PMCID: PMC11239940 DOI: 10.1038/s44319-024-00164-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: 09/01/2022] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024] Open
Abstract
ATP2B1 is a known regulator of calcium (Ca2+) cellular export and homeostasis. Diminished levels of intracellular Ca2+ content have been suggested to impair SARS-CoV-2 replication. Here, we demonstrate that a nontoxic caloxin-derivative compound (PI-7) reduces intracellular Ca2+ levels and impairs SARS-CoV-2 infection. Furthermore, a rare homozygous intronic variant of ATP2B1 is shown to be associated with the severity of COVID-19. The mechanism of action during SARS-CoV-2 infection involves the PI3K/Akt signaling pathway activation, inactivation of FOXO3 transcription factor function, and subsequent transcriptional inhibition of the membrane and reticulum Ca2+ pumps ATP2B1 and ATP2A1, respectively. The pharmacological action of compound PI-7 on sustaining both ATP2B1 and ATP2A1 expression reduces the intracellular cytoplasmic Ca2+ pool and thus negatively influences SARS-CoV-2 replication and propagation. As compound PI-7 lacks toxicity in vitro, its prophylactic use as a therapeutic agent against COVID-19 is envisioned here.
Collapse
Affiliation(s)
- Pasqualino de Antonellis
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
- Elysium Cell Bio Ita SRL, Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Veronica Ferrucci
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
- Elysium Cell Bio Ita SRL, Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Marco Miceli
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
| | - Francesca Bibbo
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Fatemeh Asadzadeh
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
- European School of Molecular Medicine, SEMM, Naples, Italy
| | - Francesca Gorini
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Alessia Mattivi
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | | | - Roberta Russo
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Immacolata Andolfo
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | | | | | - Giovanna Fusco
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Naples, 80055, Italy
| | - Maurizio Viscardi
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Naples, 80055, Italy
| | - Sergio Brandi
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Naples, 80055, Italy
| | - Pellegrino Cerino
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Naples, 80055, Italy
| | - Vittoria Monaco
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Department of Chemical Sciences, University 'Federico II' University of Naples, Naples, 80125, Italy
| | - Dong-Rac Choi
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Elysiumbio Inc., #2007, Samsung Cheil B/D, 309, Teheran-ro, Gangnam-gu, Seoul, 06151, Korea
| | - Jae-Ho Cheong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Achille Iolascon
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Stefano Amente
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Maria Monti
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Department of Chemical Sciences, University 'Federico II' University of Naples, Naples, 80125, Italy
| | - Luca L Fava
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Mario Capasso
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy
| | - Hong-Yeoul Kim
- Elysiumbio Inc., #2007, Samsung Cheil B/D, 309, Teheran-ro, Gangnam-gu, Seoul, 06151, Korea
| | - Massimo Zollo
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples, 80131, Italy.
- Elysium Cell Bio Ita SRL, Via Gaetano Salvatore 486, 80145, Naples, Italy.
- European School of Molecular Medicine, SEMM, Naples, Italy.
- DAI Medicina di Laboratorio e Trasfusionale, 'Federico II' University of Naples, 80131, Naples, Italy.
| |
Collapse
|
25
|
Cervantes KC, Danguilan RA, Arakama MHI, Chua E, Abad CLR, Rosete-Liquete RMO. Outcomes of Kidney Transplantation Among COVID-19 Recovered Patients in a Single Transplant Center. Transplant Proc 2024; 56:540-545. [PMID: 38413307 DOI: 10.1016/j.transproceed.2023.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 11/26/2023] [Indexed: 02/29/2024]
Abstract
BACKGROUND The COVID-19 pandemic significantly lowered kidney transplantation (KT) rates worldwide, and studies regarding outcomes of patients who developed COVID-19 infection before KT are limited, especially in low to middle-income countries. BACKGROUND To determine the 1-year graft and patient survival of kidney transplant recipients who recovered from COVID-19 infection before KT. METHODS We retrospectively reviewed all adult end-stage renal disease patients who underwent KT at the National Kidney and Transplant Institute from June 2020 through October 2021. Transplant parameters, graft and patient survival, pretransplant COVID-19 infection, and post-KT infectious complications were recorded. Data was analyzed using two-tailed descriptive statistical tests. RESULTS Of the 219 recipients, 23 (11%) had COVID-19 infection within 1 to 16 months before KT. The mean age of KT recipients was 36 years (range, 25-57), and 61.9% had chronic glomerulonephritis as primary renal disease. The mean duration from COVID-19 recovery to KT was 79 days (range, 21-207). There was no significant difference in the 1-year biopsy-proven acute rejection in the 2 groups, at 4.5% vs 12.5% for the COVID-19 and non-COVID-19 group, respectively. Both the 1-year graft and patient survival were similar in the COVID-19 and non-COVID-19 groups at 98.4% vs 100% and 100% vs 98.44%, respectively. CONCLUSION There was no significant difference in biopsy-proven acute rejection, 1-year graft, and patient survival among patients who had a prior COVID-19 infection vs those who did not. Kidney transplantation appears safe when performed at least 1 month from COVID-19 infection.
Collapse
Affiliation(s)
- Krystal C Cervantes
- Department of Adult Nephrology, National Kidney and Transplant Institute, Quezon City, Philippines.
| | - Romina A Danguilan
- Department of Adult Nephrology, National Kidney and Transplant Institute, Quezon City, Philippines
| | - Mel-Hatra I Arakama
- Department of Adult Nephrology, National Kidney and Transplant Institute, Quezon City, Philippines
| | - Eric Chua
- Department of Adult Nephrology, National Kidney and Transplant Institute, Quezon City, Philippines
| | - Cybele Lara R Abad
- Department of Internal Medicine, National Kidney and Transplant Institute, Quezon City, Philippines
| | - Rose Marie O Rosete-Liquete
- Department of Organ Transplant and Vascular Surgery, National Kidney and Transplant Institute, Quezon City, Philippines
| |
Collapse
|
26
|
Vajdi M, Karimi A, Hassanizadeh S, Farhangi MA, Bagherniya M, Askari G, Roufogalis BD, Davies NM, Sahebkar A. Effect of polyphenols against complications of COVID-19: current evidence and potential efficacy. Pharmacol Rep 2024; 76:307-327. [PMID: 38498260 DOI: 10.1007/s43440-024-00585-6] [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: 09/23/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024]
Abstract
The COVID-19 pandemic that started in 2019 and resulted in significant morbidity and mortality continues to be a significant global health challenge, characterized by inflammation, oxidative stress, and immune system dysfunction.. Developing therapies for preventing or treating COVID-19 remains an important goal for pharmacology and drug development research. Polyphenols are effective against various viral infections and can be extracted and isolated from plants without losing their therapeutic potential. Researchers have developed methods for separating and isolating polyphenols from complex matrices. Polyphenols are effective in treating common viral infections, including COVID-19, and can also boost immunity. Polyphenolic-based antiviral medications can mitigate SARS-CoV-2 enzymes vital to virus replication and infection. Individual polyphenolic triterpenoids, flavonoids, anthraquinonoids, and tannins may also inhibit the SARS-CoV-2 protease. Polyphenol pharmacophore structures identified to date can explain their action and lead to the design of novel anti-COVID-19 compounds. Polyphenol-containing mixtures offer the advantages of a well-recognized safety profile with few known severe side effects. However, studies to date are limited, and further animal studies and randomized controlled trials are needed in future studies. The purpose of this study was to review and present the latest findings on the therapeutic impact of plant-derived polyphenols on COVID-19 infection and its complications. Exploring alternative approaches to traditional therapies could aid in developing novel drugs and remedies against coronavirus infection.
Collapse
Affiliation(s)
- Mahdi Vajdi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arash Karimi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shirin Hassanizadeh
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Abbasalizad Farhangi
- Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Bagherniya
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Basil D Roufogalis
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Stangis M, Adesse D, Sharma B, Castro E, Kumar K, Kumar N, Minevich M, Toborek M. The S1 subunits of SARS-CoV-2 variants differentially trigger the IL-6 signaling pathway in human brain endothelial cells and downstream impact on microglia activation. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:7-15. [PMID: 38532784 PMCID: PMC10961483 DOI: 10.1515/nipt-2023-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/27/2023] [Indexed: 03/28/2024]
Abstract
Objectives Cerebrovascular complications are prevalent in COVID-19 infection and post-COVID conditions; therefore, interactions of SARS-CoV-2 with cerebral microvascular cells became an emerging concern. Methods We examined the inflammatory responses of human brain microvascular endothelial cells (HBMEC), the main structural element of the blood-brain barrier (BBB), following exposure to the S1 subunit of the spike protein of different SARS-CoV-2 variants. Specifically, we used the S1 subunit derived from the D614 variant of SARS-CoV-2, which started widely circulating in March of 2020, and from the Delta variant, which started widely circulating in early 2021. We then further examined the impact of the HBMEC secretome, produced in response to the S1 exposure, on microglial proinflammatory responses. Results Treatment with S1 derived from the D614 variant and from the Delta variant resulted in differential alterations of the IL-6 signaling pathway. Moreover, the HBMEC secretome obtained after exposure to the S1 subunit of the D614 variant activated STAT3 in microglial cells, indicating that proinflammatory signals from endothelial cells can propagate to other cells of the neurovascular unit. Overall, these results indicate the potential for different SARS-CoV-2 variants to induce unique cellular signatures and warrant individualized treatment strategies. The findings from this study also bring further awareness to proinflammatory responses involving brain microvasculature in COVID-19 and demonstrate how the surrounding microglia react to each unique variant derived response.
Collapse
Affiliation(s)
- Michael Stangis
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL33136, USA
| | - Daniel Adesse
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL33136, USA
- Laboratory of Structural Biology, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ21040-360, Brazil
| | - Bhavya Sharma
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL33136, USA
| | - Eduardo Castro
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL33136, USA
| | - Kush Kumar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL33136, USA
| | - Neil Kumar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL33136, USA
| | - Masha Minevich
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL33136, USA
| |
Collapse
|
28
|
Alipourfard I, Darvishi M, Khalighfard A, Ghazi F, Mobed A. Nanomaterial-based methods for sepsis management. Enzyme Microb Technol 2024; 174:110380. [PMID: 38147783 DOI: 10.1016/j.enzmictec.2023.110380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/26/2023] [Accepted: 12/12/2023] [Indexed: 12/28/2023]
Abstract
Sepsis is a serious disease caused by an impaired host immune response to infection, resulting in organ dysfunction, tissue damage and is responsible for high in-hospital mortality (approximately 20%). Recently, WHO documented sepsis as a global health priority. Nevertheless, there is still no effective and specific therapy for clinically detecting sepsis. Nanomaterial-based approaches have appeared as promising tools for identifying bacterial infections. In this review, recent biosensors are introduced and summarized as nanomaterial-based platforms for sepsis management and severe complications. Biosensors can be used as tools for the diagnosis and treatment of sepsis and as nanocarriers for drug delivery. In general, diagnostic methods for sepsis-associated bacteria, biosensors developed for this purpose are presented in detail, and their strengths and weaknesses are discussed. In other words, readers of this article will gain a comprehensive understanding of biosensors and their applications in sepsis management.
Collapse
Affiliation(s)
- Iraj Alipourfard
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia, Katowice, Poland
| | - Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), Department of Aerospace and Subaquatic Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Arghavan Khalighfard
- Department of Nursing and Midwifery٫ Faculty of Midwifery٬ Zanjan University of Medical Sciences, Zanjan, Iran
| | - Farhood Ghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5154853431, Iran
| | - Ahmad Mobed
- Infectious and Tropical Diseases Research Center, Clinical Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Zhang W, Gorelik AJ, Wang Q, Norton SA, Hershey T, Agrawal A, Bijsterbosch JD, Bogdan R. Associations between COVID-19 and putative markers of neuroinflammation: A diffusion basis spectrum imaging study. Brain Behav Immun Health 2024; 36:100722. [PMID: 38298902 PMCID: PMC10825665 DOI: 10.1016/j.bbih.2023.100722] [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: 07/24/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
COVID-19 remains a significant international public health concern. Yet, the mechanisms through which symptomatology emerges remain poorly understood. While SARS-CoV-2 infection may induce prolonged inflammation within the central nervous system, the evidence primarily stems from limited small-scale case investigations. To address this gap, our study capitalized on longitudinal UK Biobank neuroimaging data acquired prior to and following COVID-19 testing (N = 416 including n = 224 COVID-19 cases; Mage = 58.6). Putative neuroinflammation was assessed in gray matter structures and white matter tracts using non-invasive Diffusion Basis Spectrum Imaging (DBSI), which estimates inflammation-related cellularity (DBSI-restricted fraction; DBSI-RF) and vasogenic edema (DBSI-hindered fraction; DBSI-HF). We hypothesized that COVID-19 case status would be associated with increases in DBSI markers after accounting for potential confound (age, sex, race, body mass index, smoking frequency, and data acquisition interval) and multiple testing. COVID-19 case status was not significantly associated with DBSI-RF (|β|'s < 0.28, pFDR >0.05), but with greater DBSI-HF in left pre- and post-central gyri and right middle frontal gyrus (β's > 0.3, all pFDR = 0.03). Intriguingly, the brain areas exhibiting increased putative vasogenic edema had previously been linked to COVID-19-related functional and structural alterations, whereas brain regions displaying subtle differences in cellularity between COVID-19 cases and controls included regions within or functionally connected to the olfactory network, which has been implicated in COVID-19 psychopathology. Nevertheless, our study might not have captured acute and transitory neuroinflammatory effects linked to SARS-CoV-2 infection, possibly due to symptom resolution before the imaging scan. Future research is warranted to explore the potential time- and symptom-dependent neuroinflammatory relationship with COVID-19.
Collapse
Affiliation(s)
- Wei Zhang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Aaron J. Gorelik
- Department of Psychological & Brain Sciences, Washington University, St. Louis, MO, United States
| | - Qing Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Sara A. Norton
- Department of Psychological & Brain Sciences, Washington University, St. Louis, MO, United States
| | - Tamara Hershey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Psychological & Brain Sciences, Washington University, St. Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Janine D. Bijsterbosch
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences, Washington University, St. Louis, MO, United States
| |
Collapse
|
30
|
Ferrigno I, Verzellesi L, Ottone M, Bonacini M, Rossi A, Besutti G, Bonelli E, Colla R, Facciolongo N, Teopompi E, Massari M, Mancuso P, Ferrari AM, Pattacini P, Trojani V, Bertolini M, Botti A, Zerbini A, Giorgi Rossi P, Iori M, Salvarani C, Croci S. CCL18, CHI3L1, ANG2, IL-6 systemic levels are associated with the extent of lung damage and radiomic features in SARS-CoV-2 infection. Inflamm Res 2024:10.1007/s00011-024-01852-1. [PMID: 38308760 DOI: 10.1007/s00011-024-01852-1] [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: 09/22/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/05/2024] Open
Abstract
OBJECTIVE AND DESIGN We aimed to identify cytokines whose concentrations are related to lung damage, radiomic features, and clinical outcomes in COVID-19 patients. MATERIAL OR SUBJECTS Two hundred twenty-six patients with SARS-CoV-2 infection and chest computed tomography (CT) images were enrolled. METHODS CCL18, CHI3L1/YKL-40, GAL3, ANG2, IP-10, IL-10, TNFα, IL-6, soluble gp130, soluble IL-6R were quantified in plasma samples using Luminex assays. The Mann-Whitney U test, the Kruskal-Wallis test, correlation and regression analyses were performed. Mediation analyses were used to investigate the possible causal relationships between cytokines, lung damage, and outcomes. AVIEW lung cancer screening software, pyradiomics, and XGBoost classifier were used for radiomic feature analyses. RESULTS CCL18, CHI3L1, and ANG2 systemic levels mainly reflected the extent of lung injury. Increased levels of every cytokine, but particularly of IL-6, were associated with the three outcomes: hospitalization, mechanical ventilation, and death. Soluble IL-6R showed a slight protective effect on death. The effect of age on COVID-19 outcomes was partially mediated by cytokine levels, while CT scores considerably mediated the effect of cytokine levels on outcomes. Radiomic-feature-based models confirmed the association between lung imaging characteristics and CCL18 and CHI3L1. CONCLUSION Data suggest a causal link between cytokines (risk factor), lung damage (mediator), and COVID-19 outcomes.
Collapse
Affiliation(s)
- Ilaria Ferrigno
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Verzellesi
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marta Ottone
- Unit of Epidemiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Martina Bonacini
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandro Rossi
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Besutti
- Unit of Radiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences With Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Efrem Bonelli
- Unit of Radiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Clinical Chemistry and Endocrinology Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Rossana Colla
- Clinical Chemistry and Endocrinology Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Nicola Facciolongo
- Unit of Respiratory Diseases, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elisabetta Teopompi
- Multidisciplinary Internal Medicine Unit, Guastalla Hospital, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marco Massari
- Unit of Infectious Diseases, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Pamela Mancuso
- Unit of Epidemiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Anna Maria Ferrari
- Department of Emergency, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Pierpaolo Pattacini
- Unit of Radiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Valeria Trojani
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marco Bertolini
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Andrea Botti
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandro Zerbini
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Paolo Giorgi Rossi
- Unit of Epidemiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Mauro Iori
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlo Salvarani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences With Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Rheumatology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Stefania Croci
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
31
|
Yang R, Guan X, Niu Z, Zhang R, Lv S, Xu X, Zhao Y, Wu J. Establishment of sex-specific predictive models for critical illness in Chinese people with the Omicron variant. Front Microbiol 2024; 14:1224132. [PMID: 38322760 PMCID: PMC10844546 DOI: 10.3389/fmicb.2023.1224132] [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: 05/17/2023] [Accepted: 12/27/2023] [Indexed: 02/08/2024] Open
Abstract
Introduction The Omicron variant has rapidly spread throughout the world compared to the Delta variant and poses a great threat to global healthcare systems due to its immune evasion and rapid spread. Sex has been identified as a factor significantly associated with COVID-19 mortality, but it remains unclear which clinical indicators could be identified as risk factors in each sex group and which sex-specific risk factors might shape the worse clinical outcome, especially for Omicrons. This study aimed to confirm the relationship between sex and the progression of the Omicron variant and to explore its sex-biased risk factors. Methods We conducted a retrospective study including 1,132 hospitalized patients with the COVID-19 Omicron variant from 5 December 2022 to 25 January 2023 at Shanghai General Hospital, and the medical history data and clinical index data of the inpatients for possible sex differences were compared and analyzed. Then, a sex-specific Lasso regression was performed to select the variables significantly associated with critical illness, including intensive care unit admission, invasive mechanical ventilation, or death. A logistic regression was used to construct a sex-specific predictive model distinctively for the critical illness outcome using selected covariates. Results Among the collected 115 clinical indicators, up to 72 showed significant sex differences, including the difference in merit and the proportion of people with abnormalities. More importantly, males had greater critical illness (28.4% vs. 19.9%) and a significantly higher intensive care unit occupancy (20.96% vs. 14.49%) and mortality (13.2% vs. 4.9%), and males over 80 showed worse outcomes than females. Predictive models (AUC: 0.861 for males and 0.898 for females) showed 12 risk factors for males and 10 for females. Through a comprehensive sex-stratified analysis of a large cohort of hospitalized Omicron-infected patients, we identified the specific risk factors for critical illness by developing prediction models. Discussion Sex disparities and the identified risk factors should be considered, especially in the personalized prevention and treatment of the COVID-19 Omicron variant.
Collapse
Affiliation(s)
- Rui Yang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Guan
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziguang Niu
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rulin Zhang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siang Lv
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xiang Xu
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Zhao
- Department of Medical Affairs, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wu
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
32
|
Elnosary ME, Shreadah MA, Ashour ML, Nabil-Adam A. Predictions based on inflammatory cytokine profiling of Egyptian COVID-19 with 2 potential therapeutic effects of certain marine-derived compounds. Int Immunopharmacol 2024; 126:111072. [PMID: 38006751 DOI: 10.1016/j.intimp.2023.111072] [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: 11/12/2022] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUNDS A worldwide coronavirus pandemic has affected many healthcare systems in 2019 (COVID-19). Following viral activation, cytokines and chemokines are released, causing inflammation and tissue death, particularly in the lungs, resulting in severe COVID-19 symptoms such as pneumonia and ARDS. COVID-19 induces the release of several chemokines and cytokines in different organs, such as the cardiovascular system and lungs. RESEARCH IDEA COVID-19 and its more severe effects, such as an elevated risk of death, are more common in patients with metabolic syndrome and the elderly. Cytokine storm and COVID-19 severity may be mitigated by immunomodulation targeting NF-κB activation in conjunction with TNF- α -inhibition. In severe cases of COVID-19, inhibiting the NF-κB/TNF- α, the pathway may be employed as a therapeutic option. MATERIAL AND METHODS The study will elaborate on the Egyptian pattern for COVID-19 patients in the first part of our study. An Egyptian patient with COVID-19 inflammatory profiling will be discussed in the second part of this article using approved marine drugs selected to inhabit the significant inflammatory signals. A biomarker profiling study is currently being performed on Egyptian patients with SARS-COV-2. According to the severity of the infection, participants were divided into four groups. The First Group was non-infected with SARS-CoV-2 (Control, n = 16), the Second Group was non-intensive care patients (non-ICU, n = 16), the Third Group was intensive care patients (ICU, n = 16), and the Fourth Group was ICU with endotracheal intubation (ICU + EI, n = 16). To investigate COVID-19 inflammatory biomarkers for Egyptian patients, several inflammatory, oxidative, antioxidant, and anti-inflammatory biomarkers were measured. The following are examples of blood tests: CRP, Ferritin, D-dimer, TNF-α, IL-8, IL-6., IL-Ib, CD8, NF-κB, MDA, and total antioxidants. RESULTS AND DISCUSSION The results of the current study revealed many logical findings, such as the elevation of CRP, Ferritin, D-dimer, TNF- α, CD8, IL-6, IL-, NF-κB, and MDA. Where a significant increase showed in ICU group results (23.05 ± 0.30, 2.35 ± 0.86, 433.4 ± 159.3, 26.67 ± 3.51, 7.52 ± 1.48, 7.49 ± 1.04, 5.76 ± 1.31, 7.41 ± 0.73) respectively, and also ICU group results (54.75 ± 3.44, 0.65 ± 0.13, 460.2 ± 121.42, 27.43 ± 2.52, 8.63 ± 2.68, 10.65 ± 2.75, 5.93 ± 1.4, 10.64 ± 0.86) respectively, as well as ICU + EI group results (117.63 ± 11.89, 1.22 ± 0.65, 918.8 ± 159.27, 26.68 ± 2.00, 6.68 ± 1.08, 11.68 ± 6.16, 6.23 ± 0.07, 22.41 ± 1.39),respectively.The elevation in laboratory biomarkers of cytokines storm in three infected groups with remarkable increases in the ICU + EI group was due to the elevation of oxidative stress and inflammatory storm molecules, which lead to highly inflammatory responses, specifically in severe patients of COVID-19. Another approach to be used in the current study is investigating new computational drug compounds for SARS-COV-2 protective agents from the marine environment. The results revealed that (Imatinib and Indinavir) had the highest affinity toward Inflammatory molecules and COVID-19 proteins (PDB ID: -7CZ4 and 7KJR), which may be used in the future as possible COVID-19 drug candidates. CONCLUSION The investigated inflammatory biomarkers in Egyptian COVID-19 patients showed a strong correlation between IL6, TNF-α, NF-κB, CRB, DHL, and ferritin as COVID-19 biomarkers and determined the severity of the infection. Also, the oxidative /antioxidant showed good biomarkers for infection recovery and progression of the patients.
Collapse
Affiliation(s)
- Mohamed E Elnosary
- Al-Azhar University, Faculty of Science, Botany and Microbiology Department, 11884 Nasr City, Cairo, Egypt.
| | - Mohamed Attia Shreadah
- Marine Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Egypt
| | - Mohamed L Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abbasia, Cairo 11566, Egypt; Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia.
| | - Asmaa Nabil-Adam
- Marine Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Egypt.
| |
Collapse
|
33
|
Papic I, Bistrovic P, Cikara T, Busic N, Keres T, Ortner Hadziabdic M, Lucijanic M. Corticosteroid Dosing Level, Incidence and Profile of Bacterial Blood Stream Infections in Hospitalized COVID-19 Patients. Viruses 2024; 16:86. [PMID: 38257786 PMCID: PMC10820464 DOI: 10.3390/v16010086] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
COVID-19 patients with severe or critical symptoms are often treated with corticosteroids, per contemporary guidelines. Due to their immunosuppressive and immunomodulatory properties, corticosteroids are associated with the development of superinfections. We aimed to retrospectively assess patterns of corticosteroid use and the profiles of bacterial blood stream infections associated with exposure to different dosing levels, in a cohort of 1558 real-life adult COVID-19 patients. A total of 1391 (89.3%) patients were treated with corticosteroids, with 710 (45.6%) patients receiving low, 539 (34.6%) high and 142 (9.1%) very high corticosteroid doses. Bacteremia developed in a total of 178 (11.4%) patients. The risk of bacteremia was of similar magnitude between the no and low-dose corticosteroid treatments (p = 0.352), whereas it progressively increased with high (OR 6.18, 95% CI (2.66-14.38), p < 0.001) and very high corticosteroid doses (OR 8.12, 95% CI (3.29-20.05), p < 0.001), compared to no corticosteroid treatment. These associations persisted after multivariate adjustments and were present independently of sex, comorbidity burden, and mechanical ventilation. The profiles of individual bacterial pathogens differed depending on the used corticosteroid doses. High and very high corticosteroid doses are frequently used for real-life COVID-19 patients with severe and critical clinical presentations and are associated with a higher risk of bacteremia independently of sex, comorbidity burden, and mechanical ventilation use.
Collapse
Affiliation(s)
- Ivan Papic
- Pharmacy Department, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Petra Bistrovic
- Cardiology Department, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Tomislav Cikara
- Cardiology Department, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Nikolina Busic
- Department of Internal Medicine, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Tatjana Keres
- Department of Internal Medicine, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Maja Ortner Hadziabdic
- Centre for Applied Pharmacy, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia
| | - Marko Lucijanic
- Hematology Department, University Hospital Dubrava, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
34
|
Oley MH, Oley MC, Langi FLFG, Kepel BJ, Pajan J, Kepel REM, Wagiu AMJ, Kalitouw F, Kalesaran L, Faruk M. Factors related to serum levels of intercellular adhesion molecule-1 in probable COVID-19 patients in surgical treatment: an observational study. Pan Afr Med J 2023; 46:117. [PMID: 38465016 PMCID: PMC10924607 DOI: 10.11604/pamj.2023.46.117.41690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/18/2023] [Indexed: 03/12/2024] Open
Abstract
Introduction COVID-19 causes a systemic inflammatory response, involving dysregulation and misexpression of many inflammatory cytokines. The recruitment and activation of inflammatory cells depend on the expression of many classes of inflammatory mediators, with increased expression of endothelial cell adhesion molecules being related to COVID-19 disease severity. With the World Health Organization having recently updated case definitions to suspect, probable, and confirmed, this study aimed to measure the mean value of intercellular adhesion molecule 1 (ICAM-1) and its relation to suspected COVID-19. Methods all suspected patients (n=20) were hospitalized and treated following the Indonesian National Guidelines for COVID-19 management. ICAM-1 levels were measured on days 1 and 7, demographic data were recorded, and routine blood count values were measured and additionally considered. Results the results showed that the levels of ICAM-1 in the 1st-day group (mean 271.3 ng/ml) were higher than those in the 7th-day group (mean 253.9 ng/ml). This difference was statistically significant (p = 0.00, p ≤ 0.05). All of the patients with suspected COVID-19 were included in this study and tested for COVID-19 by reverse transcription polymerase chain reaction (RT-PCR) testing. A total of 10 patients were confirmed positive with a COVID-19 infection, with elevated ICAM-1 levels compared to the confirmed negative patients (with a mean 1st day 296.8 versus a mean 7th day 279.0 ng/ml). ICAM-1 levels of all patients decreased by the seventh day. Conclusion the mean value of ICAM-1 levels for patients with confirmed positive COVID-19 cases was higher than those with suspected COVID-19 cases.
Collapse
Affiliation(s)
- Mendy Hatibie Oley
- Division of Plastic Reconstructive and Aesthetic Surgery, Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Division of Plastic Reconstructive and Aesthetic Surgery, Department of Surgery, R. D. Kandou Hospital, Manado, Indonesia
| | - Maximillian Christian Oley
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Division of Neurosurgery, Department of Surgery, R. D. Kandou Hospital, Manado, Indonesia
| | | | - Billy Johnson Kepel
- Department of Chemistry, Faculty of Medicine, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Jacob Pajan
- Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
| | | | | | - Ferry Kalitouw
- Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
| | - Laurens Kalesaran
- Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
| | - Muhammad Faruk
- Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
35
|
Dey R, Bishayi B. Microglial Inflammatory Responses to SARS-CoV-2 Infection: A Comprehensive Review. Cell Mol Neurobiol 2023; 44:2. [PMID: 38099973 PMCID: PMC11407175 DOI: 10.1007/s10571-023-01444-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/08/2023] [Indexed: 12/18/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is primarily a respiratory disease causing a worldwide pandemic in the year of 2019. SARS-CoV-2 is an enveloped, positive-stranded RNA virus that could invade the host through spike protein and exhibits multi-organ effects. The Brain was considered to be a potential target for SARS-CoV-2 infection. Although neuropsychiatric symptoms and cognitive impairments were observed in COVID-19 patients even after recovery the mechanism of action is not well documented. In this review, the contribution of microglia in response to SARS-CoV-2 infection was discussed aiming to design a therapeutic regimen for the management of neuroinflammation and psycho-behavioral alterations. Priming of microglia facilitates the hyper-activation state when it interacts with SARS-CoV-2 known as the 'second hit'. Moreover, the microgliosis produces reactive free radicals and pro-inflammatory cytokines like IL-1β, IFN-γ, and IL-6 which ultimately contribute to a 'cytokine storm', thereby increasing the occurrence of cognitive and neurological dysfunction. It was reported that elevated CCL11 may be responsible for psychiatric disorders and ROS/RNS-induced oxidative stress could promote major depressive disorder (MDD) and phenotypic switching. Additionally, during SARS-CoV-2 infection microglia-CD8+ T cell interaction may have a significant role in neuronal cell death. This cytokine-mediated cellular cross-talking plays a crucial role in pro-inflammatory and anti-inflammatory balance within the COVID-19 patient's brain. Therefore, all these aspects will be taken into consideration for developing novel therapeutic strategies to combat SARS-CoV-2-induced neuroinflammation.
Collapse
Affiliation(s)
- Rajen Dey
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Swami Vivekananda University, Telinipara, Barasat-Barrackpore Rd, Bara Kanthalia, West Bengal, 700121, India.
| | - Biswadev Bishayi
- Immunology Laboratory, Department of Physiology, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, West Bengal, 700009, India
| |
Collapse
|
36
|
Cao X, Xie YL, Zhou CL, Mu H. The value of age IgG and IL6 in estimating time of viral clearance in asymptomatic or mild patients with COVID-19. Front Microbiol 2023; 14:1256759. [PMID: 38125571 PMCID: PMC10731291 DOI: 10.3389/fmicb.2023.1256759] [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: 07/11/2023] [Accepted: 11/07/2023] [Indexed: 12/23/2023] Open
Abstract
Background The aim of this study was to investigate the relationship between Age, immunoglobin G (IgG), immunoglobin M (IgM), procalcitonin (PCT), and interleukin-6 (IL6), and the time to clear viral nucleic acids in asymptomatic and mild coronavirus disease 2019 (COVID-19) patients, as well as evaluated the predictive value of these biochemical indicators. Methods We performed a retrospective analysis on 1,570 individuals who were admitted to Tianjin First Central Hospital and diagnosed with asymptomatic or mild cases. Laboratory data were collected, including age, gender, levels of IgG, IgM, PCT and IL6, as well as results of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) nucleic acid test. These data were statistically analyzed using SPSS software, version 24.0. Results The results indicated that among mild patients, Age, IgG, and the time to clear viral nucleic acids were higher than asymptomatic patients (p < 0.05). And the time to clear viral nucleic acids was significantly correlated with Age, IgG, IgM, PCT, and IL6 (p < 0.05), IgG (r = -0.445, p < 0.001) showed moderate correlations. Using logistic regression analysis, we identified older age, high IL6 levels, and low IgG levels were risk factors for nucleic acid clearance exceeding 14 days (p < 0.05). When combining these three indicators to predict the probability of nucleic acid clearance exceeding 14 days in the 1,570 patients, the AUROC was found to be 0.727. Conclusion Age, IgG, and IL6 could potentially serve as useful predictors for nucleic acid clearance exceeding 14 days in asymptomatic and mild COVID-19 patients.
Collapse
Affiliation(s)
- Xi Cao
- Department of Clinical Laboratory, Tianjin First Central Hospital, Tianjin, China
| | - Yong-Li Xie
- Department of Clinical Laboratory, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China
| | - Chun-lei Zhou
- Department of Clinical Laboratory, Tianjin First Central Hospital, Tianjin, China
| | - Hong Mu
- Department of Clinical Laboratory, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
37
|
Ghaleh HEG, Izadi M, Javanbakht M, Ghanei M, Einollahi B, Jafari NJ, Alishiri G, Aslani J, Abolghasemi H, Simonetti V, Khafaei M, Zhao S, Saadat SH, Ahmadi M, Parvin S, Vazifedoust S, Alvanagh AG. Cytokine profile and antioxidants status in the moderate and severe COVID-19 patients: a trial of ozone therapy impact as a medicinal supplement. Inflammopharmacology 2023; 31:3029-3036. [PMID: 37436523 DOI: 10.1007/s10787-023-01288-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/27/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND Complementary ozone therapy has been identified as a revolutionary medical technique for a number of goals and ailments. At the present, it has been shown that ozone has medicinal qualities, such as antibacterial, antifungal, and antiparasitic properties. Coronavirus (SARS-CoV-2) is quickly spread over the globe. Cytokine storms and oxidative stress seem to play a substantial role in the most of acute attacks of the disease. The aim of this research was to assess the therapeutic advantages of complementary ozone therapy on the cytokine profile and antioxidant status in COVID-19 patients. METHODS The statistical sample of this study included two hundred patients with COVID-19. One hundred COVID-19 patients (treatment group) received 240 ml of the patient's blood and an equal volume of O2/O3 gas at a concentration of 35-50 μg/ml daily, which gradually increased in concentration, and were kept for 5-10 days and one hundred patients (control group) received standard treatment. The secretion levels of IL-6, TNF-α, IL-1β, IL-10 cytokines, SOD, CAT and GPx were compared between control patients (standard treatment) and standard treatment plus intervention (ozone) before and after treatment. RESULTS The findings indicated a significant decrease in the level of IL-6, TNF-α, IL-1β in group receiving complementary ozone therapy in compared with control group. Furthermore, a significant increase was found in the level of IL-10 cytokine. Moreover, SOD, CAT and GPx levels revealed a significant increase in complementary ozone therapy group compared to control group. CONCLUSIONS Our results revealed that complementary ozone therapy can be used as a medicinal complementary therapy to reduce and control inflammatory cytokines and oxidative stress status in patients with COVID-19 as revealed its antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
| | - Morteza Izadi
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nematollah Jonaidi Jafari
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Gholamhossein Alishiri
- Chemical Injuries Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Aslani
- Chemical Injuries Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hassan Abolghasemi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Vincenzo Simonetti
- Oxygen-Ozone Therapy Scientific Society, Gorle (BG), Italy
- Kaos Non-Profit Association, Caselle Torinese (TO), Italy
| | - Mostafa Khafaei
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shi Zhao
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China
| | - Seyed Hassan Saadat
- Behavioral Sciences Research Center, Lifestyle Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Parvin
- Chemical Injuries Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Soheil Vazifedoust
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
38
|
Khalid A, Aqeel RF, Nawaz A, Ahmad J, Fatima ST, Shahid S, Rao AA, Aktas G, Ijaz S, Shehryar M. 'Immune-inflammatory markers & clinical characteristics for outcomes in hospitalized SARS-CoV-2 infected patients of Pakistan: a retrospective analysis'. Hematology 2023; 28:2199629. [PMID: 37408482 DOI: 10.1080/16078454.2023.2199629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 04/02/2023] [Indexed: 07/07/2023] Open
Abstract
OBJECTIVE Accumulating evidence suggests the role of immune-inflammatory markers in early risk stratification and prognostication of COVID-19 patients. We aimed to evaluate their association with severity and the development of diagnostic scores with optimal thresholds in critical patients. SETTING AND PARTICIPANTS This retrospective case study includes hospitalized COVID-19 patients from March 2019 to March, 2022, in the developing area teaching hospital in Pakistan. Polymerase chain reaction (PCR) positive patients, n = 467 were investigated for clinical outcomes, comorbidities and disease prognosis. The plasma levels of Interleukin-6 (IL-6), Lactate dehydrogenase (LDH), C-reactive protein (CRP), Procalcitonin (PCT), ferritin and Complete blood count markers were measured. RESULTS Majority were males (58.8%) and patients with comorbidities had more severe disease. Hypertension and diabetes mellitus were the commonest comorbidities. Shortness of breath, myalgia and cough were the main symptoms. The hematological markers NLR, as well as the plasma levels of immune-inflammatory variables, IL-6, LDH, Procalcitonin, Erythrocyte sedimentation rate, Ferritin were markedly raised in severe and critical patients (p < 0.0001 for these markers). ROC analysis supports IL-6 as the most accurate marker with high prognostic relevance with proposed cut-off threshold (43 pg/ml), determining >90% of patients in terms of COVID-19 severity (AUC = 0.93, 91.7%, se; 90.3%sp). Furthermore, positive correlation with all other markers including NLR with cut-off = 2.99 (AUC = 0.87, se = 89.8%, sp = 88.4%), CRP with cut-offs at 42.9 mg/l, (AUC = 0.883, se = 89.3% and sp = 78.6%), LDH cut-off at 267μg/L, evidenced in >80% patients (AUC = 0.834 se = 84% and sp = 80%). Additionally, ESR and ferritin have the corresponding AUC 0.81 and 0.813 with cut-off at 55 mm/hr and 370, respectively. CONCLUSION Investigating the immune-inflammatory markers can assist physicians in providing prompt treatment and ICU admission in terms of COVID-19 severity. As a result, which may reduce the overall mortality of COVID-19 patients.
Collapse
Affiliation(s)
- Atiqa Khalid
- MBBS, Sahiwal Medical College, Hospital, Sahiwal
| | - Rao Faheem Aqeel
- Postgraduate Resident Pediatrician, Sahiwal Teaching Hospital, Sahiwal
| | - Amber Nawaz
- Pathology Department, Sahiwal Medical College, Sahiwal
| | - Jehangir Ahmad
- Department of Medicine, Divisional Headquarters Teaching Hospital Mirpur AJK
| | | | | | - Aqsa Aqeel Rao
- Assistant Professor, Wateen Medical & Dental College, Rawat
| | | | - Shaista Ijaz
- MBBS, YANGTZE University Medical School, Wuhan, China
| | - Muhammad Shehryar
- Post resident Gynaecology and obstetrics, Sir Ganga Raam Hospital, Lahore
| |
Collapse
|
39
|
Izquierdo MB, Romo AMN, Zafra AGS, Garrido JDDLG. Predictors of mortality in patients with COVID-19 by flow cytometry. CLINICAL IMMUNOLOGY COMMUNICATIONS 2023; 3:14-20. [PMID: 38014400 PMCID: PMC9918322 DOI: 10.1016/j.clicom.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 11/29/2023]
Abstract
Despite the great impact of severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2), we still lack techniques that allow us to anticipate the natural history of the disease in order to avoid or shorten the clinical period of the disease. The values of nine cytokines were measured in COVID-19+ patients admitted to the Hospital Universitario Reina Sofía (HURS) using flow cytometry. The cytokines measured are IL-1ß, IL-6, MCP-1, IP-10, IL-10, IL-8, IL-12, IFN-γ and TNF-α. Given the absence of previous studies on cytokine values in healthy patients using the flow cytometry technique, and the low availability of resources in the first waves of COVID-19, a control group was lacking, all resources were employed for monitoring sick patients. However, this study has revealed a greater increase in two specific cytokines, which are also found to be higher than the rest in healthy patients: MCP-1 and IP-10, which are mainly responsible for cytokine storm and post-disease thrombosis.
Collapse
Affiliation(s)
| | - Ana María Navas Romo
- Departamento de Inmunología, Hospital Universitario Reina Sofía, Córdoba, España
| | | | | |
Collapse
|
40
|
Shen YL, Hsieh YA, Hu PW, Lo PC, Hsiao YH, Ko HK, Lin FC, Huang CW, Su KC, Perng DW. Angiotensin-(1-7) attenuates SARS-CoV2 spike protein-induced interleukin-6 and interleukin-8 production in alveolar epithelial cells through activation of Mas receptor. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023; 56:1147-1157. [PMID: 37802686 DOI: 10.1016/j.jmii.2023.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 08/30/2023] [Accepted: 09/17/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND SARS-CoV-2 spike proteins (SP) can bind to the human angiotensin-converting enzyme 2 (ACE2) in human pulmonary alveolar epithelial cells (HPAEpiC) and trigger an inflammatory process. Angiotensin-(1-7) may have an anti-inflammatory effect through activation of Mas receptor. This study aims to investigate whether SARS-CoV-2 SP can induce inflammation through ACE2 in the alveolar epithelial cells which can be modulated through angiotensin-(1-7)/Mas receptor axis. METHODS HPAEpiC were treated with SARS-CoV-2 SP in the presence or absence of ACE2 antagonist-dalbavancin and Mas receptor agonist-angiotensin-(1-7). Proinflammatory cytokine production (IL-6 and IL-8) were measured at mRNA and protein levels. MAP kinase phosphorylation and transcription factor activation was determined by Western Blot. Mas receptor was blocked by either antagonist (A779) or knockdown (specific SiRNA). Experiments were replicated using A549 cells. FINDINGS SARS-CoV-2 SP (5 μg/mL) significantly induced MAP kinase (ERK1/2) phosphorylation, downstream transcription factor (activator protein-1, AP-1) activation and cytokine production (IL-6 and IL-8) at both mRNA and protein levels. Pretreatment with dalbavancin (10 μg/mL), or angiotensin-(1-7) (10 μM) significantly reduced ERK1/2 phosphorylation, AP-1 activation, and cytokine production. However, these angiotensin-(1-7)-related protective effects were significantly abolished by blocking Mas receptor with either antagonist (A799,10 μM) or SiRNA knockdown. INTERPRETATION SARS-CoV-2 SP can induce proinflammatory cytokine production, which can be inhibited by either ACE2 antagonist or Mas receptor agonist-angiotensin-(1-7). Angiotensin-(1-7)-related protective effect on cytokine reduction can be abolished by blocking Mas receptor. Our findings suggest that ACE2/angiotensin-(1-7)/Mas axis may serve as a therapeutic target to control inflammatory response triggered by SARS-CoV-2 SP.
Collapse
Affiliation(s)
- Yi-Luen Shen
- Division of Chest Medicine, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan, ROC
| | - Yi-An Hsieh
- Division of Chest Medicine, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan, ROC
| | - Po-Wei Hu
- Division of Chest Medicine, Department of Internal Medicine, National Yang Ming Chiao Tung University Hospital, Taiwan, ROC; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Po-Chun Lo
- Taipei Veterans General Hospital, Fenglin Branch, Hualien, Taiwan, ROC; Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Han Hsiao
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hsin-Kuo Ko
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Fang-Chi Lin
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chien-Wen Huang
- Division of Chest Medicine, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan, ROC; Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan, ROC
| | - Kang-Cheng Su
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
| | - Diahn-Warng Perng
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
| |
Collapse
|
41
|
Dey A, Vaishak K, Deka D, Radhakrishnan AK, Paul S, Shanmugam P, Daniel AP, Pathak S, Duttaroy AK, Banerjee A. Epigenetic perspectives associated with COVID-19 infection and related cytokine storm: an updated review. Infection 2023; 51:1603-1618. [PMID: 36906872 PMCID: PMC10008189 DOI: 10.1007/s15010-023-02017-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/27/2023] [Indexed: 03/13/2023]
Abstract
PURPOSE The COVID-19 pandemic caused by the novel Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) has put the world in a medical crisis for the past three years; nearly 6.3 million lives have been diminished due to the virus outbreak. This review aims to update the recent findings on COVID-19 infections from an epigenetic scenario and develop future perspectives of epi-drugs to treat the disease. METHODS Original research articles and review studies related to COVID-19 were searched and analyzed from the Google Scholar/PubMed/Medline databases mainly between 2019 and 2022 to brief the recent work. RESULTS Numerous in-depth studies of the mechanisms used by SARS-CoV-2 have been going on to minimize the consequences of the viral outburst. Angiotensin-Converting Enzyme 2 receptors and Transmembrane serine protease 2 facilitate viral entry to the host cells. Upon internalization, it uses the host machinery to replicate viral copies and alter the downstream regulation of the normal cells, causing infection-related morbidities and mortalities. In addition, several epigenetic regulations such as DNA methylation, acetylation, histone modifications, microRNA, and other factors (age, sex, etc.) are responsible for the regulations of viral entry, its immune evasion, and cytokine responses also play a major modulatory role in COVID-19 severity, which has been discussed in detail in this review. CONCLUSION Findings of epigenetic regulation of viral pathogenicity open a new window for epi-drugs as a possible therapeutical approach against COVID-19.
Collapse
Affiliation(s)
- Amit Dey
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India
| | - K Vaishak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India
| | - Dikshita Deka
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India
| | - Arun Kumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, TN, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No.500 Fracc., CP 76130, San Pablo, Querétaro, Mexico
| | - Priyadarshini Shanmugam
- Department of Microbiology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, TN, 603103, India
| | - Alice Peace Daniel
- Department of Microbiology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, TN, 603103, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, TN, 603103, India.
| |
Collapse
|
42
|
Felkle D, Zięba K, Kaleta K, Czaja J, Zyzdorf A, Sobocińska W, Jarczyński M, Bryniarski K, Nazimek K. Overreactive macrophages in SARS-CoV-2 infection: The effects of ACEI. Int Immunopharmacol 2023; 124:110858. [PMID: 37708705 DOI: 10.1016/j.intimp.2023.110858] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
Among various factors influencing the course of SARS-CoV-2 infection in humans, macrophage overactivation is considered the main cause of the cytokine storm that leads to severe complications of COVID-19. Moreover, the increased expression of angiotensin converting enzyme 2 (ACE2), an obligatory entry receptor of the coronavirus, caused by treatment with ACE inhibitors (ACEI) lowered overall confidence in the safety of these drugs. However, analysis of the course of coronavirus infection in patients treated with ACEI does not support these concerns. Instead, the beneficial effect of ACEI on macrophages has increasingly been emphasized. This includes their anti-inflammatory activation and the consequent reduction in the risk of severe disease and life-threatening complications. Herein, we summarize the current knowledge and understanding of the dual role of macrophages in SARS-CoV-2 infection, with a special focus on the postulated mechanisms underlying the beneficial effects of macrophage targeting by ACEI. These seem to involve the stimulation of macrophage angiotensin II type 2 and Mas receptors by angiotensin 1-7, intensively produced due to the up-regulation of ACE2 expression on macrophages, as well as the direct inhibition of macrophage hyper-responsiveness by ACEI. The impact of ACEI on macrophages may also lead to the activation of an effective antiviral response due to the increased expression of ACE2.
Collapse
Affiliation(s)
- Dominik Felkle
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Katarzyna Zięba
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Konrad Kaleta
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Julia Czaja
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Amanda Zyzdorf
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Wiktoria Sobocińska
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Mateusz Jarczyński
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Katarzyna Nazimek
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland.
| |
Collapse
|
43
|
Bahgat MM, Nadeem R, Nasraa MH, Amer K, Hassan WA, ELGarhy FM, Reda S, Abd-Elshafy DN. Proinflammatory Cytokine Profiles in Both Mild Symptomatic and Asymptomatic SARS-CoV-2-Infected Egyptian Individuals and a Proposed Relationship to Post-COVID-19 Sequela. Viral Immunol 2023; 36:600-609. [PMID: 37831918 DOI: 10.1089/vim.2023.0060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023] Open
Abstract
Severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) infection is associated with proinflammatory cytokine release as mediators of host antiviral response to the infection. Cytokine persistent elevation leads to post-Coronavirus disease-2019 (COVID-19) post-COVID-19 sequela (PCS) reported in about 60% of patients affecting individual's normal life after recovery. This study evaluates relationship of cytokines and chemokines pattern during and postinfection to PCS events. Serum samples collected from 82 individuals with symptomatic, asymptomatic, or no SARS-CoV-2 infection were classified as recently or formerly infected groups according to levels of anti-2019nCoV Immunoglobulin G/Immunoglobulin M. Levels of interleukin (IL)-1α, IL-1β, IL-6, IL-8, interferon alpha (IFN-α), tumor necrosis factor alpha (TNF-α), granulocyte macrophage colony-stimulating factor (GM-CSF), and monocyte chemoattractant protein-1 were assessed via ELISA for each individual. All asymptomatic groups showed nonsignificant differences in cytokines' levels than control group. Significant elevation of IFN-α, TNF-α, and GM-CSF levels were observed in recent symptomatic, while IFN-α and TNF-α levels were significant in former symptomatic groups. We observed an association between fever with IL-1α and IFN-α levels, fatigue with TNF-α and GM-CSF, dyspnea with IFN-α, TNF-α, and GM-CSF, and chest-wheezing with GM-CSF. Individuals were surveyed 12 months postsampling for PCS events. Among 35 responders to survey, 8 (22.8%) reported PCS events, 6 of which were females. Upon studying PCS events, IL-8, IFN-α, TNF-α, and GM-CSF levels showed significant elevation in active infection, that was not seen in a resolved state of infection. Cytokines patterns suggest that either a persistent elevation in levels or damage caused during infection contributes to PCS. Although with the limited sample size, our study emphasizes the importance to conduct medical approaches targeting the associated cytokines to improve the PCS symptoms.
Collapse
Affiliation(s)
- Mahmoud M Bahgat
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, The National Research Centre, Cairo, Egypt
- Immune- and Bio-markers for Infection Research Group, The Center of Excellence for Advanced Sciences, The National Research Centre, Cairo, Egypt
| | - Rola Nadeem
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, The National Research Centre, Cairo, Egypt
- Immune- and Bio-markers for Infection Research Group, The Center of Excellence for Advanced Sciences, The National Research Centre, Cairo, Egypt
| | - Mohamed H Nasraa
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, The National Research Centre, Cairo, Egypt
- Immune- and Bio-markers for Infection Research Group, The Center of Excellence for Advanced Sciences, The National Research Centre, Cairo, Egypt
| | - Khaled Amer
- Egypt Center for Research and Regenerative Medicine, Cairo, Egypt
| | - Wael A Hassan
- Egypt Center for Research and Regenerative Medicine, Cairo, Egypt
| | - Fadya M ELGarhy
- Egypt Center for Research and Regenerative Medicine, Cairo, Egypt
| | - Salem Reda
- Egypt Center for Research and Regenerative Medicine, Cairo, Egypt
| | - Dina N Abd-Elshafy
- Immune- and Bio-markers for Infection Research Group, The Center of Excellence for Advanced Sciences, The National Research Centre, Cairo, Egypt
- Department of Water Pollution Research, Environmental and Climate Change Research Institute, The National Research Centre, Cairo, Egypt
| |
Collapse
|
44
|
Bello-Perez M, Hurtado-Tamayo J, Mykytyn AZ, Lamers MM, Requena-Platek R, Schipper D, Muñoz-Santos D, Ripoll-Gómez J, Esteban A, Sánchez-Cordón PJ, Enjuanes L, Haagmans BL, Sola I. SARS-CoV-2 ORF8 accessory protein is a virulence factor. mBio 2023; 14:e0045123. [PMID: 37623322 PMCID: PMC10653805 DOI: 10.1128/mbio.00451-23] [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: 03/01/2023] [Accepted: 07/04/2023] [Indexed: 08/26/2023] Open
Abstract
IMPORTANCE The relevance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ORF8 in the pathogenesis of COVID-19 is unclear. Virus natural isolates with deletions in ORF8 were associated with wild milder disease, suggesting that ORF8 might contribute to SARS-CoV-2 virulence. This manuscript shows that ORF8 is involved in inflammation and in the activation of macrophages in two experimental systems: humanized K18-hACE2 transgenic mice and organoid-derived human airway cells. These results identify ORF8 protein as a potential target for COVID-19 therapies.
Collapse
Affiliation(s)
- M. Bello-Perez
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - J. Hurtado-Tamayo
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - A. Z. Mykytyn
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - M. M. Lamers
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - R. Requena-Platek
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - D. Schipper
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - D. Muñoz-Santos
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - J. Ripoll-Gómez
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - A. Esteban
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - P. J. Sánchez-Cordón
- Veterinary Pathology Department, Animal Health Research Center (CISA), National Institute of Research, Agricultural and Food Technology, Valdeolmos, Spain
| | - L. Enjuanes
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - B. L. Haagmans
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - I. Sola
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
45
|
Chujan S, Nakareangrit W, Suriyo T, Satayavivad J. Integrated Transcriptomics and Network Analysis of Potential Mechanisms and Health Effects of Convalescent COVID-19 Patients. Bioinform Biol Insights 2023; 17:11779322231206684. [PMID: 37881207 PMCID: PMC10594973 DOI: 10.1177/11779322231206684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/22/2023] [Indexed: 10/27/2023] Open
Abstract
Coronaviral disease 2019 (COVID-19) is a recent pandemic disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Currently, there are still cases of COVID-19 around the world that can develop into persistent symptoms after discharge. The constellation of symptoms, termed long COVID, persists for months and can lead to various diseases such as lung inflammation and cardiovascular disease, which may lead to considerable financial burden and possible risk to human health. Moreover, the molecular mechanisms underlying the post-pandemic syndrome of COVID-19 remain unclear. In this study, we aimed to explore the molecular mechanism, disease association, and possible health risks in convalescent COVID-19 patients. Gene expression data from a human convalescent COVID-19 data set was compared with a data set from healthy normal individuals in order to identify differentially expressed genes (DEGs). To determine biological function and potential pathway alterations, the GO and KEGG databases were used to analyze the DEGs. Disease association, tissue, and organ-specific analyses were used to identify possible health effects. A total of 250 DEGs were identified between healthy and convalescent COVID-19 subjects. The biological function alterations identified revealed cytokine interactions and increased inflammation through NF-κB1, RELA, JUN, STAT3, and SP1. Interestingly, the most significant pathways were cytokine-cytokine receptor interaction, altered lipid metabolism, and atherosclerosis that play a crucial role in convalescent COVID-19. In addition, we also found pneumonitis, dermatitis, and autoimmune diseases. Based on our study, convalescent COVID-19 is associated with inflammation in a variety of organs that could lead to autoimmune and inflammatory diseases, as well as atherosclerosis. These findings are a first step toward fully exploring the disease mechanisms in depth to understand the relationship between post-COVID-19 infection and potential health risks. This is necessary for the development of appropriate strategies for the prevention and treatment of long COVID.
Collapse
Affiliation(s)
- Suthipong Chujan
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| | | | - Tawit Suriyo
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| |
Collapse
|
46
|
Xie J, Kuppermann N, Florin TA, Tancredi DJ, Funk AL, Kim K, Salvadori MI, Yock-Corrales A, Shah NP, Breslin KA, Chaudhari PP, Bergmann KR, Ahmad FA, Nebhrajani JR, Mintegi S, Gangoiti I, Plint AC, Avva UR, Gardiner MA, Malley R, Finkelstein Y, Dalziel SR, Bhatt M, Kannikeswaran N, Caperell K, Campos C, Sabhaney VJ, Chong SL, Lunoe MM, Rogers AJ, Becker SM, Borland ML, Sartori LF, Pavlicich V, Rino PB, Morrison AK, Neuman MI, Poonai N, Simon NJE, Kam AJ, Kwok MY, Morris CR, Palumbo L, Ambroggio L, Navanandan N, Eckerle M, Klassen TP, Payne DC, Cherry JC, Waseem M, Dixon AC, Ferre IB, Freedman SB. Impact of SARS-CoV-2 Infection on the Association Between Laboratory Tests and Severe Outcomes Among Hospitalized Children. Open Forum Infect Dis 2023; 10:ofad485. [PMID: 37869403 PMCID: PMC10588618 DOI: 10.1093/ofid/ofad485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/28/2023] [Indexed: 10/24/2023] Open
Abstract
Background To assist clinicians with identifying children at risk of severe outcomes, we assessed the association between laboratory findings and severe outcomes among severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected children and determined if SARS-CoV-2 test result status modified the associations. Methods We conducted a cross-sectional analysis of participants tested for SARS-CoV-2 infection in 41 pediatric emergency departments in 10 countries. Participants were hospitalized, had laboratory testing performed, and completed 14-day follow-up. The primary objective was to assess the associations between laboratory findings and severe outcomes. The secondary objective was to determine if the SARS-CoV-2 test result modified the associations. Results We included 1817 participants; 522 (28.7%) SARS-CoV-2 test-positive and 1295 (71.3%) test-negative. Seventy-five (14.4%) test-positive and 174 (13.4%) test-negative children experienced severe outcomes. In regression analysis, we found that among SARS-CoV-2-positive children, procalcitonin ≥0.5 ng/mL (adjusted odds ratio [aOR], 9.14; 95% CI, 2.90-28.80), ferritin >500 ng/mL (aOR, 7.95; 95% CI, 1.89-33.44), D-dimer ≥1500 ng/mL (aOR, 4.57; 95% CI, 1.12-18.68), serum glucose ≥120 mg/dL (aOR, 2.01; 95% CI, 1.06-3.81), lymphocyte count <1.0 × 109/L (aOR, 3.21; 95% CI, 1.34-7.69), and platelet count <150 × 109/L (aOR, 2.82; 95% CI, 1.31-6.07) were associated with severe outcomes. Evaluation of the interaction term revealed that a positive SARS-CoV-2 result increased the associations with severe outcomes for elevated procalcitonin, C-reactive protein (CRP), D-dimer, and for reduced lymphocyte and platelet counts. Conclusions Specific laboratory parameters are associated with severe outcomes in SARS-CoV-2-infected children, and elevated serum procalcitonin, CRP, and D-dimer and low absolute lymphocyte and platelet counts were more strongly associated with severe outcomes in children testing positive compared with those testing negative.
Collapse
Affiliation(s)
| | - Nathan Kuppermann
- Davis School of Medicine, University of California, Sacramento, California, USA
| | - Todd A Florin
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Northwestern University, Chicago, Illinois, USA
| | - Daniel J Tancredi
- Davis School of Medicine, University of California, Sacramento, California, USA
| | - Anna L Funk
- University of Calgary, Calgary, Alberta, Canada
| | - Kelly Kim
- University of Calgary, Calgary, Alberta, Canada
| | | | | | - Nipam P Shah
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | - Fahd A Ahmad
- Washington University School of Medicine, St.Louis, Missouri, USA
| | | | - Santiago Mintegi
- University of the Basque Country, UPV/EHU Bilbao, Basque Country, Spain
| | - Iker Gangoiti
- University of the Basque Country, UPV/EHU Bilbao, Basque Country, Spain
| | - Amy C Plint
- University of Ottawa, Ottawa, Ontario, Canada
| | - Usha R Avva
- Montefiore-Nyack Hospital, Nyack, NewYork, New York, USA
| | | | | | | | | | - Maala Bhatt
- Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | | | | | - Carmen Campos
- Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | | - Shu-Ling Chong
- Duke-NUS Medical School, SingHealth Duke-NUS Global Health Institute, Singapore
| | - Maren M Lunoe
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Sarah M Becker
- Primary Children’s Hospital, Intermountain Healthcare, Salt Lake City, Utah, USA
| | | | - Laura F Sartori
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Pedro B Rino
- Hospital de Pediatría “Prof. Dr. Juan P. Garrahan,” RIDEPLA, Buenos Aires, Argentina
| | | | | | - Naveen Poonai
- Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Norma-Jean E Simon
- Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - April J Kam
- McMaster Children’s Hospital, Hamilton, Ontario, Canada
| | - Maria Y Kwok
- Columbia University Irving Medical Center, NewYork, New York, USA
| | - Claudia R Morris
- Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Laura Palumbo
- ASST Spedali Civili di Brescia—Pronto Soccorso Pediatrico, Brescia, Italy
| | | | | | - Michelle Eckerle
- University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Daniel C Payne
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | | | | | | | | |
Collapse
|
47
|
Laughey W, Lodhi I, Pennick G, Smart L, Sanni O, Sandhu S, Charlesworth B. Ibuprofen, other NSAIDs and COVID-19: a narrative review. Inflammopharmacology 2023; 31:2147-2159. [PMID: 37603158 PMCID: PMC10518289 DOI: 10.1007/s10787-023-01309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/22/2023]
Abstract
At the start of the coronavirus disease 2019 (COVID-19) pandemic (March 2020), there was speculation that non-steroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen, used to manage some of the symptoms of COVID-19, could increase the susceptibility to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and negatively impact clinical outcomes. In the absence of any robust mechanistic and clinical evidence, this speculation led to confusion about the safety of ibuprofen, contributing to the so-called 'infodemic' surrounding COVID-19. A wealth of evidence has been generated in subsequent years, and this narrative review aims to consider the body of in vitro and in vivo research, observational studies, systematic reviews and meta-analyses on the use of NSAIDs, including ibuprofen, in COVID-19. Overall, the direction of evidence supports that NSAIDs do not increase susceptibility to infection, nor worsen disease outcomes in patients with COVID-19. Neither do they impact the immune response to COVID-19 vaccines. There is no basis to limit the use of NSAIDs, and doing so may deprive patients of effective self-care measures to control symptoms.
Collapse
Affiliation(s)
- William Laughey
- Reckitt Health Care UK Ltd, Hull, UK.
- Hull York Medical School, University of York, York, UK.
| | | | | | | | | | | | | |
Collapse
|
48
|
Francis ME, Jansen EB, Yourkowski A, Selim A, Swan CL, MacPhee BK, Thivierge B, Buchanan R, Lavender KJ, Darbellay J, Rogers MB, Lew J, Gerdts V, Falzarano D, Skowronski DM, Sjaarda C, Kelvin AA. Previous infection with seasonal coronaviruses does not protect male Syrian hamsters from challenge with SARS-CoV-2. Nat Commun 2023; 14:5990. [PMID: 37752151 PMCID: PMC10522707 DOI: 10.1038/s41467-023-41761-1] [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: 02/22/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
SARS-CoV-2 variants and seasonal coronaviruses continue to cause disease and coronaviruses in the animal reservoir pose a constant spillover threat. Importantly, understanding of how previous infection may influence future exposures, especially in the context of seasonal coronaviruses and SARS-CoV-2 variants, is still limited. Here we adopted a step-wise experimental approach to examine the primary immune response and subsequent immune recall toward antigenically distinct coronaviruses using male Syrian hamsters. Hamsters were initially inoculated with seasonal coronaviruses (HCoV-NL63, HCoV-229E, or HCoV-OC43), or SARS-CoV-2 pango B lineage virus, then challenged with SARS-CoV-2 pango B lineage virus, or SARS-CoV-2 variants Beta or Omicron. Although infection with seasonal coronaviruses offered little protection against SARS-CoV-2 challenge, HCoV-NL63-infected animals had an increase of the previously elicited HCoV-NL63-specific neutralizing antibodies during challenge with SARS-CoV-2. On the other hand, primary infection with HCoV-OC43 induced distinct T cell gene signatures. Gene expression profiling indicated interferon responses and germinal center reactions to be induced during more similar primary infection-challenge combinations while signatures of increased inflammation as well as suppression of the antiviral response were observed following antigenically distant viral challenges. This work characterizes and analyzes seasonal coronaviruses effect on SARS-CoV-2 secondary infection and the findings are important for pan-coronavirus vaccine design.
Collapse
Affiliation(s)
- Magen E Francis
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ethan B Jansen
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Anthony Yourkowski
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Alaa Selim
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Cynthia L Swan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Brian K MacPhee
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Brittany Thivierge
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Rachelle Buchanan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kerry J Lavender
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Joseph Darbellay
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Matthew B Rogers
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jocelyne Lew
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Danuta M Skowronski
- BC Centre for Disease Control, Immunization Programs and Vaccine Preventable Diseases Service, Vancouver, BC, Canada
- University of British Columbia, School of Population and Public Health, Vancouver, BC, Canada
| | - Calvin Sjaarda
- Department of Psychiatry, Queen's University, Kingston, ON, Canada
- Queen's Genomics Lab at Ongwanada (Q-GLO), Ongwanada Resource Centre, Kingston, ON, Canada
| | - Alyson A Kelvin
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
49
|
HOSAMIRUDSARI HADISEH, ALIMOHAMADI YOUSEF, AKRAMI MAJID, ZAHRA MAHAK, FARAJI NEDA. Is single point HbA1c a reliable predictor for death in severe COVID-19? JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2023; 64:E298-E303. [PMID: 38125992 PMCID: PMC10730054 DOI: 10.15167/2421-4248/jpmh2023.64.3.3032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/05/2023] [Indexed: 12/23/2023]
Abstract
Introduction The severity of COVID-19 infection is affected by several risk factors such as Diabetes Mellitus (DM). The current study aimed to determine the effect of single-point HbA1c on the severity and mortality of hospitalized COVID-19 patients. Methods This cross-sectional study was conducted among hospitalized moderate and severe COVID-19 patients in Baharloo Hospital in Iran between December 23rd and February 23rd, 2021. The patients have been diagnosed by Polymerase Chain Reaction (PCR) and Chest Computed Tomography (CT) imaging as COVID-19. Demographic data, clinical presentation, laboratory results, and treatments along with the HbA1c data were included. Results 165 COVID-19 cases were included in this study; 126 (76.4%) of which were severe cases. 89 (53.9%) patients were male, with a mean age of 59.89 ± 16.59 years. Severe COVID-19 patients were more prone to a longer hospital stay, and a higher level of inflammatory mediators, compared to the moderate COVID-19 patients (p < 0.05). No significant association was found between single point HbA1c, FBS, and severity and mortality of COVID-19 cases (p > 0.05). Conclusions Single point HbA1c was not a reliable mediator for the prediction of severity or death in hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- HADISEH HOSAMIRUDSARI
- Department of Infectious Diseases, Baharloo Hospital, Railway Square, Tehran University of Medical Sciences, Tehran, Iran
| | - YOUSEF ALIMOHAMADI
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - MAJID AKRAMI
- Department of Anesthesiology, Baharloo Hospital, Railway Square; Tehran University of Medical Sciences, Tehran, Iran
| | - MAHAK ZAHRA
- Department of Infectious Diseases, Baharloo Hospital, Railway Square, Tehran University of Medical Sciences, Tehran, Iran
| | - NEDA FARAJI
- Department of Internal Medicine, Baharloo Hospital, Railway Square, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Radkhah H, Mansouri ES, Rahimipour Anaraki S, Gholizadeh Mesgarha M, Sheikhy A, Khadembashiri MM, Khadembashiri MA, Eslami M, Mahmoodi T, Inanloo B, Pour Mohammad A. Predictive value of hematological indices on incidence and severity of pulmonary embolism in COVID-19 patients. Immun Inflamm Dis 2023; 11:e1012. [PMID: 37773719 PMCID: PMC10540144 DOI: 10.1002/iid3.1012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/28/2023] [Accepted: 08/31/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Pulmonary thromboembolism (PTE) is a common complication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which raises the COVID-19 disease's fatality rate from 3% to 45%. Nevertheless, due to fairly indistinguishable clinical symptoms and a lack of validated clinical prediction models, PTE diagnosis in COVID-19 patients is challenging. This study aims to investigate the applicability of hematological indices to predict PTE incidence and its severity in SARS-CoV-2 patients. METHODS A retrospective cohort study was conducted on hospitalized patients with a confirmed diagnosis of SARS-CoV-2 infection who underwent CT angiography to assess probable PTE in them. The correlation between complete blood count parameters 1 day before CT angiography and CT angiography outcomes, and simplified pulmonary embolism severity index (s-PESI) was investigated. RESULTS We discovered that among individuals with a probable PTE, males and those with higher platelet-to-lymphocyte (PLR) and neutrophil-to-lymphocyte (NLR) ratios had a greater likelihood of PTE incidence (p < .001, .027, and .037, respectively). PLR was a significant and independent predictor of PTE with a p value of .045. Moreover, a higher neutrophil count was associated with a higher s-PESI score in COVID-19 patients developing PTE (p: .038). CONCLUSIONS Among hematological indices, NLR and more precisely PLR are cost-effective and simply calculable markers that can assist physicians in determining whether or not COVID-19 patients with clinically probable PTE require CT angiography and the higher neutrophil count can be employed as an indicator of PTE severity in COVID-19 patients. Further large multicenter and prospective studies are warranted to corroborate these observations.
Collapse
Affiliation(s)
- Hanieh Radkhah
- Department of Internal Medicine, School of Medicine, Sina HospitalTehran University of Medical Sciences (TUMS)TehranIran
| | - Ensieh Sadat Mansouri
- Department of Internal Medicine, School of Medicine, Sina HospitalTehran University of Medical Sciences (TUMS)TehranIran
| | | | | | - Ali Sheikhy
- Students' Scientific Research CenterTehran University of Medical Sciences (TUMS)TehranIran
| | | | | | - Mohamad Eslami
- Students' Scientific Research CenterTehran University of Medical Sciences (TUMS)TehranIran
| | - Tara Mahmoodi
- Students' Scientific Research CenterTehran University of Medical Sciences (TUMS)TehranIran
| | - Behnaz Inanloo
- Sina HospitalTehran University of Medical Sciences (TUMS)TehranIran
| | | |
Collapse
|