1
|
Wu M, Sun C, Shi Q, Luo Y, Wang Z, Wang J, Qin Y, Cui W, Yan C, Dai H, Wang Z, Zeng J, Zhou Y, Zhu M, Liu X. Dry eye disease caused by viral infection: Past, present and future. Virulence 2024; 15:2289779. [PMID: 38047740 PMCID: PMC10761022 DOI: 10.1080/21505594.2023.2289779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023] Open
Abstract
Following viral infection, the innate immune system senses viral products, such as viral nucleic acids, to activate innate defence pathways, leading to inflammation and apoptosis, control of cell proliferation, and consequently, threat to the whole body. The ocular surface is exposed to the external environment and extremely vulnerable to viral infection. Several studies have revealed that viral infection can induce inflammation of the ocular surface and reduce tear secretion of the lacrimal gland (LG), consequently triggering ocular morphological and functional changes and resulting in dry eye disease (DED). Understanding the mechanisms of DED caused by viral infection and its potential therapeutic strategies are crucial for clinical interventional advances in DED. This review summarizes the roles of viral infection in the pathogenesis of DED, applicable diagnostic and therapeutic strategies, and potential regions of future studies.
Collapse
Affiliation(s)
- Min Wu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Cuilian Sun
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Qin Shi
- Department of General Medicine, Gongli Hospital, Shanghai, China
| | - Yalu Luo
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Ziyu Wang
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jianxiang Wang
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yun Qin
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Weihang Cui
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Chufeng Yan
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Huangyi Dai
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Zhiyang Wang
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jia Zeng
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yamei Zhou
- Department of Microbiology Laboratory, Jiaxing Center for Disease Control and Prevention, Jiaxing, Zhejiang, China
| | - Manhui Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
2
|
Cong B, Dong X, Yang Z, Yu P, Chai Y, Liu J, Zhang M, Zang Y, Kang J, Feng Y, Liu Y, Feng W, Wang D, Deng W, Li F, Song Z, Wang Z, Chen X, Qin H, Yu Q, Li Z, Liu S, Xu X, Zhong N, Ren X, Qin C, Liu L, Wang J, Cao X. Single-cell spatiotemporal analysis reveals alveolar dendritic cell-T cell immunity hubs defending against pulmonary infection. Cell Discov 2024; 10:103. [PMID: 39414763 PMCID: PMC11484931 DOI: 10.1038/s41421-024-00733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/08/2024] [Indexed: 10/18/2024] Open
Abstract
How immune cells are spatiotemporally coordinated in the lung to effectively monitor, respond to, and resolve infection and inflammation in primed form needs to be fully illustrated. Here we apply immunocartography, a high-resolution technique that integrates spatial and single-cell RNA sequencing (scRNA-seq) through deconvolution and co-localization analyses, to the SARS-CoV-2-infected Syrian hamster model. We generate a comprehensive transcriptome map of the whole process of pulmonary infection from physiological condition, infection initiation, severe pneumonia to natural recovery at organ scale and single-cell resolution, with 142,965 cells and 45 lung lobes from 25 hamsters at 5 time points. Integrative analysis identifies that alveolar dendritic cell-T cell immunity hubs, where Ccr7+Ido1+ dendritic cells, Cd160+Cd8+ T cells, and Tnfrsf4+Cd4+ T cells physiologically co-localize, rapidly expand during SARS-CoV-2 infection, eliminate SARS-CoV-2 with the aid of Slamf9+ macrophages, and then restore to physiological levels after viral clearance. We verify the presence of these cell subpopulations in the immunity hubs in normal and SARS-CoV-2-infected hACE2 mouse models, as well as in publicly available human scRNA-seq datasets, demonstrating the potential broad relevance of our findings in lung immunity.
Collapse
Affiliation(s)
- Boyi Cong
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Dong
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Zongheng Yang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Pin Yu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangyang Chai
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaqi Liu
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Meihan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | | | | | - Yu Feng
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Dehe Wang
- Changping Laboratory, Beijing, China
| | - Wei Deng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Fengdi Li
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiqi Song
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziqiao Wang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosu Chen
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hua Qin
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Qinyi Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhiqing Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Shuxun Liu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | | | - Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China.
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Xuetao Cao
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China.
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
3
|
Sabit H, Arneth B, Abdel-Ghany S, Madyan EF, Ghaleb AH, Selvaraj P, Shin DM, Bommireddy R, Elhashash A. Beyond Cancer Cells: How the Tumor Microenvironment Drives Cancer Progression. Cells 2024; 13:1666. [PMID: 39404428 PMCID: PMC11475877 DOI: 10.3390/cells13191666] [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: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Liver cancer represents a substantial global health challenge, contributing significantly to worldwide morbidity and mortality. It has long been understood that tumors are not composed solely of cancerous cells, but also include a variety of normal cells within their structure. These tumor-associated normal cells encompass vascular endothelial cells, fibroblasts, and various inflammatory cells, including neutrophils, monocytes, macrophages, mast cells, eosinophils, and lymphocytes. Additionally, tumor cells engage in complex interactions with stromal cells and elements of the extracellular matrix (ECM). Initially, the components of what is now known as the tumor microenvironment (TME) were thought to be passive bystanders in the processes of tumor proliferation and local invasion. However, recent research has significantly advanced our understanding of the TME's active role in tumor growth and metastasis. Tumor progression is now known to be driven by an intricate imbalance of positive and negative regulatory signals, primarily influenced by specific growth factors produced by both inflammatory and neoplastic cells. This review article explores the latest developments and future directions in understanding how the TME modulates liver cancer, with the aim of informing the design of novel therapies that target critical components of the TME.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt; (H.S.); (E.F.M.)
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt;
| | - Engy F. Madyan
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt; (H.S.); (E.F.M.)
| | - Ashraf H. Ghaleb
- Department of Surgery, College of Medicine, Misr University for Science and Technology, Giza P.O. Box 77, Egypt;
- Department of Surgery, College of Medicine, Cairo University, Giza 12613, Egypt
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.S.); (R.B.)
| | - Dong M. Shin
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Ramireddy Bommireddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.S.); (R.B.)
| | - Ahmed Elhashash
- Department of Biology, Texas A&M University, 3258 TAMU I, College Station, TX 77843-3258, USA
| |
Collapse
|
4
|
Liu L, Zhang L, Hao X, Wang Y, Zhang X, Ge L, Wang P, Tian B, Zhang M. Coronavirus envelope protein activates TMED10-mediated unconventional secretion of inflammatory factors. Nat Commun 2024; 15:8708. [PMID: 39379362 PMCID: PMC11461611 DOI: 10.1038/s41467-024-52818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
The precise cellular mechanisms underlying heightened proinflammatory cytokine production during coronavirus infection remain incompletely understood. Here we identify the envelope (E) protein in severe coronaviruses (SARS-CoV-2, SARS, or MERS) as a potent inducer of interleukin-1 release, intensifying lung inflammation through the activation of TMED10-mediated unconventional protein secretion (UcPS). In contrast, the E protein of mild coronaviruses (229E, HKU1, or OC43) demonstrates a less pronounced effect. The E protein of severe coronaviruses contains an SS/DS motif, which is not present in milder strains and facilitates interaction with TMED10. This interaction enhances TMED10-oligomerization, facilitating UcPS cargo translocation into the ER-Golgi intermediate compartment (ERGIC)-a pivotal step in interleukin-1 UcPS. Progesterone analogues were identified as compounds inhibiting E-enhanced release of proinflammatory factors and lung inflammation in a Mouse Hepatitis Virus (MHV) infection model. These findings elucidate a molecular mechanism driving coronavirus-induced hyperinflammation, proposing the E-TMED10 interaction as a potential therapeutic target to counteract the adverse effects of coronavirus-induced inflammation.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Lijingyao Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xinyan Hao
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaochun Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Peihui Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Boxue Tian
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Min Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China.
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
5
|
Groves AM, Paris ND, Johnston CJ, Hernady E, Finkelstein J, Lawrence P, Marples B. Mitigating Viral Impact on the Radiation Response of the Lung. Radiat Res 2024; 202:552-564. [PMID: 39048109 DOI: 10.1667/rade-24-00103.1] [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: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 07/27/2024]
Abstract
Inflammation is a key factor in both influenza and radiation-induced lung pathophysiology. This implies a commonality of response to pulmonary damage from these insults and suggests exacerbated pathology may occur after combined exposure. We therefore tested the hypothesis that past inflammation from viral infection alters the lung microenvironment and lowers tolerance for radiation injury. Mice were inoculated with influenza A virus (IAV) and three weeks later, after virus clearance, mice received total-body irradiation (TBI). Survival as well as systemic and local lung inflammation were assessed, and strategies to mitigate pulmonary injury were investigated. After IAV infection alone, body condition recovered within 3 weeks, however inflammatory pathways remained active for 15 weeks. IAV infection exacerbated subsequent TBI responses, evident by increased lethality, enhanced histologically evident lung injury and an altered lung macrophage phenotype. To mitigate this enhanced sensitivity, captopril [an angiotensin converting enzyme inhibitor (ACEi)] was administered to limit tissue inflammation, or inflammatory monocyte-derived macrophage recruitment was blocked with a C-C chemokine receptor type 2 (CCR2) inhibitor. Both treatments abrogated the changes in circulating immune cells observed 4 weeks after TBI, and attenuated pro-inflammatory phenotypes in lung alveolar macrophages, appearing to shift immune cell dynamics towards recovery. Histologically apparent lung injury was not improved by either treatment. We show that latent lung injury from viral infection exacerbates radiation morbidity and mortality. Although strategies that attenuate proinflammatory immune cell phenotypes can normalize macrophage dynamics, this does not fully mitigate lung injury. Recognizing that past viral infections can enhance lung radiosensitivity is of critical importance for patients receiving TBI, as it could increase the incidence of adverse outcomes.
Collapse
Affiliation(s)
- Angela M Groves
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Nicole D Paris
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Carl J Johnston
- Department of Pediatrics, University of Rochester, Rochester, New York
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Jacob Finkelstein
- Department of Pediatrics, University of Rochester, Rochester, New York
| | - Paige Lawrence
- Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| |
Collapse
|
6
|
Al-Mistarehi AHW, El-Akawi S, Kheirallah KA, Bani Ata EM, Zaitoun KJ, Khassawneh AB, Jarrah A, Alzoubi HM, Al-Azzam S, Karasneh RA, Altawalbeh RB, Khassawneh B. Enhanced Treatment in Severe-Critical COVID-19 With Tocilizumab, Remdesivir, Dexamethasone: A Jordanian Cohort Study. Cureus 2024; 16:e67467. [PMID: 39314607 PMCID: PMC11417280 DOI: 10.7759/cureus.67467] [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] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Several medications have been proposed to manage COVID-19, with controversial data regarding their clinical benefits. We aimed to investigate the clinical efficacy of using remdesivir (RDV) with and without tocilizumab (TCZ) and standard therapy in treating severe COVID-19. METHODS This retrospective cohort study was conducted in a Jordanian tertiary hospital (September 26th, 2020 - August 28th, 2021) and included adult COVID-19 patients requiring oxygen support. Patients were categorized into three groups based on treatment: TCZ+RDV and standard therapy; RDV and standard therapy; and standard therapy alone, which included dexamethasone, vitamins, anticoagulants, and ceftriaxone. RESULTS Of 1,556 screened, 1,244 patients (mean age 62.33, 60.8% men) were included. Distribution was 106 in TCZ+RDV, 520 in RDV, and 618 in standard therapy. No significant differences were observed in age, gender, or BMI. Mortality was lowest in TCZ+RDV (32.1%), followed by RDV (40.6%) and standard therapy (47.1%) (p=0.005). Among ICU patients, TCZ+RDV showed significantly lower mortality (51.1%) compared to RDV (75%) and standard therapy (85.8%) (p<0.001). The ICU stays and invasive mandatory ventilation (IMV) durations were significantly shorter with TCZ+RDV (4.30 and 2.69 days, respectively) compared to RDV (7.61 and 4.52 days) and standard therapy (7.98 and 5.32 days) (p<0.001 for ICU stays, p=0.025 for IMV durations). CONCLUSIONS Combining TCZ, RDV, and dexamethasone shows promise in reducing mortality and ICU/IMV duration for severe COVID-19.
Collapse
Affiliation(s)
| | - Shadi El-Akawi
- Internal Medicine, MedStar Washington Hospital Center-Georgetown University, Washington, DC, USA
| | - Khalid A Kheirallah
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, JOR
| | - Ehab M Bani Ata
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, JOR
| | - Khaled J Zaitoun
- Faculty of Medicine, Jordan University of Science and Technology, Amman, JOR
| | - Ahmad B Khassawneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, JOR
| | - Abdullah Jarrah
- Internal Medicine, Detroit Medical Center/Sinai Grace Hospital/Wayne State University, Detroit, USA
| | - Hamed M Alzoubi
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, JOR
| | - Sayer Al-Azzam
- Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, JOR
| | | | - Rana B Altawalbeh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, JOR
| | - Basheer Khassawneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, JOR
| |
Collapse
|
7
|
Martins YA, Guerra-Gomes IC, Rodrigues TS, Tapparel C, Lopez RFV. Enhancing pulmonary delivery and immunomodulation of respiratory diseases through virus-mimicking nanoparticles. J Control Release 2024; 372:417-432. [PMID: 38908758 DOI: 10.1016/j.jconrel.2024.06.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
This study introduces the nanobromhexine lipid particle (NBL) platform designed for effective pulmonary drug delivery. Inspired by respiratory virus transport mechanisms, NBL address challenges associated with mucus permeation and inflammation in pulmonary diseases. Composed of low molecular weight polyethylene glycol-coated lipid nanoparticles with bromhexine hydrochloride, NBL exhibit a size of 118 ± 24 nm, a neutral zeta potential, osmolarity of 358 ± 28 mOsmol/kg, and a pH of 6.5. Nebulizing without leakage and showing no toxicity to epithelial cells, NBL display mucoadhesive properties with a 60% mucin-binding efficiency. They effectively traverse the dense mucus layer of Calu-3 cultures in an air-liquid interface, as supported by a 55% decrease in MUC5AC density and a 29% increase in nanoparticles internalization compared to non-exposed cells. In assessing immunomodulatory effects, NBL treatment in SARS-CoV-2-infected lung cells leads to a 40-fold increase in anti-inflammatory MUC1 gene expression, a proportional reduction in pro-inflammatory IL-6 expression, and elevated anti-inflammatory IL-10 expression. These findings suggest a potential mechanism to regulate the excessive IL-6 expression triggered by virus infection. Therefore, the NBL platform demonstrates promising potential for efficient pulmonary drug delivery and immunomodulation, offering a novel approach to addressing mucus permeation and inflammation in pulmonary diseases.
Collapse
Affiliation(s)
- Yugo Araújo Martins
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil
| | - Isabel Cristina Guerra-Gomes
- Fundação Oswaldo Cruz - FIOCRUZ, Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, SP 14040-030, Brazil
| | - Tamara Silva Rodrigues
- Department of Biochemistry and Imumunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Renata Fonseca Vianna Lopez
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil.
| |
Collapse
|
8
|
Berger L, Wolf J, Kalbitz S, Kellner N, Lübbert C, Borte S. Comparative Analysis of Lymphocyte Populations in Post-COVID-19 Condition and COVID-19 Convalescent Individuals. Diagnostics (Basel) 2024; 14:1286. [PMID: 38928701 PMCID: PMC11202600 DOI: 10.3390/diagnostics14121286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Reduced lymphocyte counts in peripheral blood are one of the most common observations in acute phases of viral infections. Although many studies have already examined the impact of immune (dys)regulation during SARS-CoV-2 infection, there are still uncertainties about the long-term consequences for lymphocyte homeostasis. Furthermore, as persistent cellular aberrations have been described following other viral infections, patients with "Post-COVID-19 Condition" (PCC) may present similarly. In order to investigate cellular changes in the adaptive immune system, we performed a retrospective analysis of flow cytometric data from lymphocyte subpopulations in 106 patients with confirmed SARS-CoV-2 infection who received medical care at our institution. The patients were divided into three groups according to the follow-up date; laboratory analyses of COVID-19 patients were compared with 28 unexposed healthy controls. Regarding B lymphocyte subsets, levels of IgA + CD27+, IgG + CD27+, IgM + CD27- and switched B cells were significantly reduced at the last follow-up compared to unexposed healthy controls (UHC). Of the 106 COVID-19 patients, 56 were clinically classified as featuring PCC. Significant differences between PCC and COVID-19 convalescents compared to UHC were observed in T helper cells and class-switched B cells. However, we did not detect specific or long-lasting immune cellular changes in PCC compared to the non-post-COVID-19 condition.
Collapse
Affiliation(s)
- Luisa Berger
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
| | - Johannes Wolf
- Department of Laboratory Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
| | - Nils Kellner
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Stephan Borte
- Department of Laboratory Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| |
Collapse
|
9
|
Li Y, Tang Y, Wang X, Zhu A, Liu D, He Y, Guo H, Zheng J, Liu X, Chi F, Wang Y, Zhuang Z, Zhang Z, Liu D, Chen Z, Li F, Ran W, Yu K, Wang D, Wen L, Zhuo J, Zhang Y, Xi Y, Zhao J, Zhao J, Sun J. Characterization of humoral immune responses against SARS-CoV-2 accessory proteins in infected patients and mouse model. Virol Sin 2024; 39:414-421. [PMID: 38677713 PMCID: PMC11280257 DOI: 10.1016/j.virs.2024.04.005] [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: 11/05/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, encodes several accessory proteins that have been shown to play crucial roles in regulating the innate immune response. However, their expressions in infected cells and immunogenicity in infected humans and mice are still not fully understood. This study utilized various techniques such as luciferase immunoprecipitation system (LIPS), immunofluorescence assay (IFA), and western blot (WB) to detect accessory protein-specific antibodies in sera of COVID-19 patients. Specific antibodies to proteins 3a, 3b, 7b, 8 and 9c can be detected by LIPS, but only protein 3a antibody was detected by IFA or WB. Antibodies against proteins 3a and 7b were only detected in ICU patients, which may serve as a marker for predicting disease progression. Further, we investigated the expression of accessory proteins in SARS-CoV-2-infected cells and identified the expressions of proteins 3a, 6, 7a, 8, and 9b. We also analyzed their ability to induce antibodies in immunized mice and found that only proteins 3a, 6, 7a, 8, 9b and 9c were able to induce measurable antibody productions, but these antibodies lacked neutralizing activities and did not protect mice from SARS-CoV-2 infection. Our findings validate the expression of SARS-CoV-2 accessory proteins and elucidate their humoral immune response, providing a basis for protein detection assays and their role in pathogenesis.
Collapse
Affiliation(s)
- Yuming Li
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250117, China; Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250117, China; State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yanhong Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China; Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Hunan Normal University, Changsha, 410005, China
| | - Xiaoqian Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250117, China; Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250117, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Dongdong Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yiyun He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Hu Guo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jie Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xinzhuo Liu
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250117, China; Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250117, China
| | - Fengyu Chi
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250117, China; Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250117, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Donglan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Fang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Wei Ran
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Kuai Yu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Dong Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Liyan Wen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jianfen Zhuo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yanjun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yin Xi
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China; Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China; Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China; Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China; Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518005, China.
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
10
|
Opsteen S, Fram T, Files JK, Levitan EB, Goepfert P, Erdmann N. Impact of Chronic HIV Infection on Acute Immune Responses to SARS-CoV-2. J Acquir Immune Defic Syndr 2024; 96:92-100. [PMID: 38408318 PMCID: PMC11009054 DOI: 10.1097/qai.0000000000003399] [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: 07/23/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024]
Abstract
ABSTRACT There is mounting evidence that HIV infection is a risk factor for severe presentations of COVID-19. We hypothesized that the persistent immune activation associated with chronic HIV infection contributes to worsened outcomes during acute COVID-19. The goals of this study were to provide an in-depth analysis of immune response to acute COVID-19 and investigate relationships between immune responses and clinical outcomes in an unvaccinated, sex- and race-matched cohort of people with HIV (PWH, n = 20) and people without HIV (PWOH, n = 41). We performed flow cytometric analyses on peripheral blood mononuclear cells from PWH and PWOH experiencing acute COVID-19 (≤21-day postsymptom onset). PWH were younger (median 52 vs 65 years) and had milder COVID-19 (40% vs 88% hospitalized) compared with PWOH. Flow cytometry panels included surface markers for immune cell populations, activation and exhaustion surface markers (with and without SARS-CoV-2-specific antigen stimulation), and intracellular cytokine staining. We observed that PWH had increased expression of activation (eg, CD137 and OX40) and exhaustion (eg, PD1 and TIGIT) markers as compared to PWOH during acute COVID-19. When analyzing the impact of COVID-19 severity, we found that hospitalized PWH had lower nonclassical (CD16 + ) monocyte frequencies, decreased expression of TIM3 on CD4 + T cells, and increased expression of PDL1 and CD69 on CD8 + T cells. Our findings demonstrate that PWH have increased immune activation and exhaustion as compared to a cohort of predominately older, hospitalized PWOH and raises questions on how chronic immune activation affects acute disease and the development of postacute sequelae.
Collapse
Affiliation(s)
- Skye Opsteen
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Tim Fram
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Jacob K. Files
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Emily B. Levitan
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - Paul Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Nathaniel Erdmann
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| |
Collapse
|
11
|
Xu X, Huang J, Zhang H, Lu W, Liu J. Differential expression of lymphocyte subpopulations in the peripheral blood of patients with COVID-19: Implications for disease severity and prognosis. Immun Inflamm Dis 2024; 12:e1281. [PMID: 38780019 PMCID: PMC11112625 DOI: 10.1002/iid3.1281] [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: 12/04/2023] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
OBJECTIVE To investigate the expression patterns and clinical significance of specific lymphocyte subsets in the peripheral blood of patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. METHODS Between December 2022 and February 2023, a cohort of 165 patients from the First Affiliated Hospital of Guangzhou University of Chinese Medicine were analyzed. The participants represented various stages of coronavirus infection severity: mild, moderate, severe, and critical. Additionally, 40 healthy individuals constituted the control group. The FC 500MPL flow cytometer and associated reagents for flow cytometry. RESULTS Compared with the healthy control group, activated B lymphocytes witnessed a pronounced increase (p < .05). A significant decrease was observed in the levels of Breg, Cytotoxic T cells or Suppressor T-cell (Tc/s), late-activated T, late-activated Th, and late-activated Tc/s lymphocytes (p < .05). Th, initial Th, initial Tc/s, total Treg, natural Treg, induced Treg, early activated T, and early activated Th lymphocyte levels showed no significant difference (p > .05). As the disease progressed, there was an uptick in midterm activated T lymphocytes (p < .05), while Breg, T, Tc/s, senescent Tc/s, and total senescent T levels dwindled (p < .05). Noteworthy patterns emerged across different groups for B1, T-lymphocytes, Tc/s, B2, CD8+ Treg cells, and other subsets, highlighting variance in immune responses relative to disease severity. When juxtaposed, no significant difference was found in the expression levels of lymphocyte subsets between patients who died and those deemed critically ill (p > .05). CONCLUSION Subsets of Treg and B-cells could act as yardsticks for the trajectory of SARS-CoV-2 infection and might have potential in forecasting patient trajectories. A comprehensive evaluation of lymphocyte subsets, especially in real-time, holds the key to discerning the clinical severity in those with COVID-19. This becomes instrumental in monitoring treatment outcomes, tracking disease evolution, and formulating prognostications. Moreover, the results provide a deeper understanding of the cellular immune defense mechanisms against the novel coronavirus infection.
Collapse
Affiliation(s)
- XinQiang Xu
- The First Affiliated HospitalUniversity of Chinese MedicineGuangzhouChina
- Guangdong Clinical Research Academy of Chinese MedicineGuangzhouChina
| | - JunYuan Huang
- The First Affiliated HospitalUniversity of Chinese MedicineGuangzhouChina
- Guangdong Clinical Research Academy of Chinese MedicineGuangzhouChina
| | - Haiqi Zhang
- Guangzhou United Yijian Medical Laboratory Co., LtdGuangzhouChina
| | - Weiguo Lu
- The First Affiliated HospitalUniversity of Chinese MedicineGuangzhouChina
- Guangdong Clinical Research Academy of Chinese MedicineGuangzhouChina
| | - Jiduo Liu
- The First Affiliated HospitalUniversity of Chinese MedicineGuangzhouChina
- Guangdong Clinical Research Academy of Chinese MedicineGuangzhouChina
| |
Collapse
|
12
|
Sun Y, Liu P, Zhang L, Lei S, Gao Q. Peripheral Blood CD8+T Cell as a Prognostic Biomarker for Hospitalised COVID-19 Patients Without Antiviral Treatment. Infect Drug Resist 2024; 17:109-117. [PMID: 38230269 PMCID: PMC10790588 DOI: 10.2147/idr.s432283] [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: 09/07/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024] Open
Abstract
Background The status of T lymphocyte subset counts in patients with COVID-19 remains uncertain. This study aimed to assess alterations in peripheral blood CD3+CD8+T (CD8+T) cells among hospitalized COVID-19 patients who have not received antiviral treatment and to evaluate their prognostic value within this patient population. Methods A single-center, retrospective cohort study and a meta-analysis were conducted. The cohort study was performed at Xiangya Hospital from December 5, 2022, to January 31, 2023. We conducted a meta-analysis to explore the association between peripheral blood CD3+CD8+T cells and mortality in COVID-19 patients who did not receive antiviral therapy. All relevant studies in Embase, PubMed, Web of Science databases were systematically searched for meta-analysis. Results The retrospective cohort study included 201 patients. A significant decrease in peripheral blood CD8+ T cell count was found to be associated with an increased risk of mortality (adjusted odds ratio [aOR]: 13.88; 95% confidence interval [CI]: 3.15-61.23), after adjusting for gender, age, comorbidities, severity at admission, steroid therapy, and antibiotic therapy. The threshold value for CD8+T cell counts, determined by the receiver operating characteristic (ROC) curve analysis, was 145.5 (area under the curve [AUC]: 0.828, specificity: 90.3%, sensitivity: 72.9%, P<0.001). Additionally, A total of 7 studies with 2765 participants were included in the meta-analysis. The meta-analysis reveals a significant association between lower CD8+ T cell counts and mortality (odds ratio [OR] = 3.543, 95% CI: 1.726 to 7.272; I2=93%). Conclusion Peripheral blood CD8+ T cell can serve as a valuable prognostic biomarker for hospitalized patients who do not receive antiviral treatment.
Collapse
Affiliation(s)
- Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Engineering Research Centre of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Furong Laboratory, Central South University, Changsha, People’s Republic of China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Centre of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Peilin Liu
- Clinical Laboratory Department, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Lifang Zhang
- Department of Plastic and Cosmetic Surgery, Changsha Mylike Cosmetic Hospital, Changsha, People’s Republic of China
| | - Shaorong Lei
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Qian Gao
- Clinical Laboratory Department, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
13
|
Deng J, Moskalyk M, Zuo QK, Garcia C, Abbas U, Ramaraju HB, Rayner D, Park YJ, Heybati K, Zhou F, Lohit S. Evaluating fluvoxamine for the outpatient treatment of COVID-19: A systematic review and meta-analysis. Rev Med Virol 2024; 34:e2501. [PMID: 38148036 DOI: 10.1002/rmv.2501] [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: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 12/28/2023]
Abstract
This systematic review and meta-analysis of randomised controlled trials (RCTs) aimed to evaluate the efficacy, safety, and tolerability of fluvoxamine for the outpatient management of COVID-19. We conducted this review in accordance with the PRISMA 2020 guidelines. Literature searches were conducted in MEDLINE, EMBASE, International Pharmaceutical Abstracts, CINAHL, Web of Science, and CENTRAL up to 14 September 2023. Outcomes included incidence of hospitalisation, healthcare utilization (emergency room visits and/or hospitalisation), mortality, supplemental oxygen and mechanical ventilation requirements, serious adverse events (SAEs) and non-adherence. Fluvoxamine 100 mg twice a day was associated with reductions in the risk of hospitalisation (risk ratio [RR] 0.75, 95% confidence interval [CI] 0.58-0.97; I 2 = 0%) and reductions in the risk of healthcare utilization (RR 0.68, 95% CI 0.53-0.86; I 2 = 0%). While no increased SAEs were observed, fluvoxamine 100 mg twice a day was associated with higher treatment non-adherence compared to placebo (RR 1.61, 95% CI 1.22-2.14; I 2 = 53%). In subgroup analyses, fluvoxamine reduced healthcare utilization in outpatients with BMI ≥30 kg/m2 , but not in those with lower BMIs. While fluvoxamine offers potential benefits in reducing healthcare utilization, its efficacy may be most pronounced in high-risk patient populations. The observed non-adherence rates highlight the need for better patient education and counselling. Future investigations should reassess trial endpoints to include outcomes relating to post-COVID sequelaes. Registration: This review was prospectively registered on PROSPERO (CRD42023463829).
Collapse
Affiliation(s)
- Jiawen Deng
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Myron Moskalyk
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Qi Kang Zuo
- UBC Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cristian Garcia
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Umaima Abbas
- Schulich School of Medicine & Dentistry (Windsor), Western University, Windsor, Ontario, Canada
| | | | - Daniel Rayner
- Department of Health Research Methods, Evidence, and Impact, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Ye-Jean Park
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Kiyan Heybati
- Mayo Clinic Alix School of Medicine (Jacksonville), Mayo Clinic, Jacksonville, Florida, USA
| | - Fangwen Zhou
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Simran Lohit
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Chen Y, Guo DZ, Zhu CL, Ren SC, Sun CY, Wang Y, Wang JF. The implication of targeting PD-1:PD-L1 pathway in treating sepsis through immunostimulatory and anti-inflammatory pathways. Front Immunol 2023; 14:1323797. [PMID: 38193090 PMCID: PMC10773890 DOI: 10.3389/fimmu.2023.1323797] [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: 10/19/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Sepsis currently remains a major contributor to mortality in the intensive care unit (ICU), with 48.9 million cases reported globally and a mortality rate of 22.5% in 2017, accounting for almost 20% of all-cause mortality worldwide. This highlights the urgent need to improve the understanding and treatment of this condition. Sepsis is now recognized as a dysregulation of the host immune response to infection, characterized by an excessive inflammatory response and immune paralysis. This dysregulation leads to secondary infections, multiple organ dysfunction syndrome (MODS), and ultimately death. PD-L1, a co-inhibitory molecule expressed in immune cells, has emerged as a critical factor in sepsis. Numerous studies have found a significant association between the expression of PD-1/PD-L1 and sepsis, with a particular focus on PD-L1 expressed on neutrophils recently. This review explores the role of PD-1/PD-L1 in immunostimulatory and anti-inflammatory pathways, illustrates the intricate link between PD-1/PD-L1 and sepsis, and summarizes current therapeutic approaches against PD-1/PD-L1 in the treatment and prognosis of sepsis in preclinical and clinical studies.
Collapse
Affiliation(s)
- Yu Chen
- School of Basic Medicine, Naval Medical University, Shanghai, China
| | - De-zhi Guo
- School of Basic Medicine, Naval Medical University, Shanghai, China
| | - Cheng-long Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shi-chun Ren
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chen-yan Sun
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yi Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jia-feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
15
|
Li T, Wang D, Wei H, Xu X. Cytokine storm and translating IL-6 biology into effective treatments for COVID-19. Front Med 2023; 17:1080-1095. [PMID: 38157195 DOI: 10.1007/s11684-023-1044-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024]
Abstract
As of May 3, 2023, the Coronavirus disease 2019 (COVID-19) pandemic has resulted in more than 760 million confirmed cases and over 6.9 million deaths. Several patients have developed pneumonia, which can deteriorate into acute respiratory distress syndrome. The primary etiology may be attributed to cytokine storm, which is triggered by the excessive release of proinflammatory cytokines and subsequently leads to immune dysregulation. Considering that high levels of interleukin-6 (IL-6) have been detected in several highly pathogenic coronavirus-infected diseases, such as severe acute respiratory syndrome in 2002, the Middle East respiratory syndrome in 2012, and COVID-19, the IL-6 pathway has emerged as a key in the pathogenesis of this hyperinflammatory state. Thus, we review the history of cytokine storm and the process of targeting IL-6 signaling to elucidate the pivotal role played by tocilizumab in combating COVID-19.
Collapse
Affiliation(s)
- Tiantian Li
- Department of Geriatric Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Dongsheng Wang
- Respiratory and Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Haiming Wei
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Science and Medical Center, University of Science and Technology of China, Hefei, 230001, China
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, China
| | - Xiaoling Xu
- Respiratory and Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
16
|
Ghaempanah F, Nikouei M, Cheraghi M, Jahangiri A, Moradi Y. Does tocilizumab have an effect on the clinical outcomes in COVID-19 patients? A meta-analysis of randomized control trials. J Pharm Policy Pract 2023; 16:151. [PMID: 37986199 PMCID: PMC10658795 DOI: 10.1186/s40545-023-00662-w] [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: 01/19/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND This meta-analysis was conducted to investigate the impact of tocilizumab on clinical outcomes associated with COVID-19. METHODS A comprehensive search was conducted across Scopus, PubMed (Medline), Cochrane Library, EMBASE (Elsevier), ClinicalTrials.gov, and Web of Sciences to identify pertinent studies published until May 2022. The primary search terms included "tocilizumab" and "COVID-19". Following the formulation of the search strategy, all identified studies were screened, and the data extraction process was initiated. Subsequently, the Cochrane risk of bias checklist was employed to evaluate the risk of bias. The effects of tocilizumab were assessed utilizing the pooled risk ratio (RR) and the fixed effect model in STATA (version 17). RESULTS In this meta-analysis, we analyzed 17 clinical trial studies to assess the impact of tocilizumab on mortality in patients with COVID-19. The pooled risk ratio (RR) for mortality was 0.93 (RR: 0.93; 95% CI: 0.86, 1.00; I2: 72.39%; P value: 0.001). The findings indicated that tocilizumab use was associated with a 4% increase in ICU hospitalization (RR: 1.04; 95% CI: 0.90, 1.20; I2: 0.00%; P value: 0.65). Additionally, tocilizumab administration was linked to a 2% reduction in the requirement for a ventilator (RR: 0.98; 95% CI: 0.90, 1.08; I2: 26.87%; P value: 0.16). CONCLUSION The administration of tocilizumab during the COVID-19 pandemic, prescribed to patients with the virus, exerted a noteworthy impact on reducing outcomes associated with COVID-19.
Collapse
Affiliation(s)
- Faezeh Ghaempanah
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maziar Nikouei
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mojtaba Cheraghi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Arman Jahangiri
- Department of Emergency Medicine, School of Medicine, Besat Hospital, Hamadan University of Medical Sciences, Hamedan, Iran
| | - Yousef Moradi
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, 66179-13446, Iran.
| |
Collapse
|
17
|
Di Spigna G, Spalletti Cernia D, Covelli B, Vargas M, Rubino V, Iacovazzo C, Napolitano F, Postiglione L. Interleukin-6 and Its Soluble Receptor Complex in Intensive Care Unit COVID-19 Patients: An Analysis of Second Wave Patients. Pathogens 2023; 12:1264. [PMID: 37887780 PMCID: PMC10610043 DOI: 10.3390/pathogens12101264] [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: 07/26/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
In December 2019, a SARS-CoV-2 virus, coined Coronavirus Disease 2019 (COVID-19), discovered in Wuhan, China, affected the global population, causing more than a million and a half deaths. Since then, many studies have shown that the hyperinflammatory response of the most severely affected patients was primarily related to a higher concentration of the pro-inflammatory cytokine interleukin-6, which directly correlated with disease severity and high mortality. Our study analyzes IL-6 and its soluble receptor complex (sIL-6R and sgp130) in critically ill COVID-19 patients who suffered severe respiratory failure from the perspective of the second COVID wave of 2020. A chemiluminescent immunoassay was performed for the determination of IL6 in serum together with an enzyme-linked immunosorbent assay to detect serum levels of sIL-6R and sgp130, which confirmed that the second wave's serum levels of IL-6 were significantly elevated in the more severe patients, as with the first 2019 COVID-19 wave, resulting in adverse clinical outcomes. At present, considering that no specific treatment for severe COVID-19 cases in its later stages exists, these molecules could be considered promising markers for disease progression, illness severity, and risk of mortality.
Collapse
Affiliation(s)
- Gaetano Di Spigna
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (D.S.C.); (V.R.); (F.N.); (L.P.)
| | - Daniela Spalletti Cernia
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (D.S.C.); (V.R.); (F.N.); (L.P.)
| | - Bianca Covelli
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (D.S.C.); (V.R.); (F.N.); (L.P.)
| | - Maria Vargas
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.)
| | - Valentina Rubino
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (D.S.C.); (V.R.); (F.N.); (L.P.)
| | - Carmine Iacovazzo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.)
| | - Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (D.S.C.); (V.R.); (F.N.); (L.P.)
| | - Loredana Postiglione
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (D.S.C.); (V.R.); (F.N.); (L.P.)
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples “Federico II”, 80131 Naples, Italy
| |
Collapse
|
18
|
Yu S, Lin Y, Li Y, Chen S, Zhou L, Song H, Yang C, Zhang H, Zhou J, Sun S, Li Y, Chen J, Feng R, Qiao N, Xie Y, Zhang R, Yin T, Chen S, Li Q, Zhu J, Qu J. Systemic immune profiling of Omicron-infected subjects inoculated with different doses of inactivated virus vaccine. Cell 2023; 186:4615-4631.e16. [PMID: 37769658 DOI: 10.1016/j.cell.2023.08.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 07/03/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023]
Abstract
SARS-CoV-2 primary strain-based vaccination exerts a protective effect against Omicron variants-initiated infection, symptom occurrence, and disease severity in a booster-dependent manner. Yet, the underlying mechanisms remain unclear. During the 2022 Omicron outbreak in Shanghai, we enrolled 122 infected adults and 50 uninfected controls who had been unvaccinated or vaccinated with two or three doses of COVID-19 inactive vaccines and performed integrative analysis of 41-plex CyTOF, RNA-seq, and Olink on their peripheral blood samples. The frequencies of HLA-DRhi classical monocytes, non-classical monocytes, and Th1-like Tem tended to increase, whereas the frequency of Treg was reduced by booster vaccine, and they influenced symptom occurrence in a vaccine dose-dependent manner. Intercorrelation and mechanistic analysis suggested that the booster vaccination induced monocytic training, which would prime monocytic activation and maturation rather than differentiating into myeloid-derived suppressive cells upon Omicron infections. Overall, our study provides insights into how booster vaccination elaborates protective immunity across SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Shanhe Yu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Yingni Lin
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Yong Li
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Shijun Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Lina Zhou
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Hejie Song
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Cuiping Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 201801, China
| | - Haiqing Zhang
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Jianping Zhou
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Shunchang Sun
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 201801, China
| | - Yanan Li
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Juan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Ruixue Feng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Niu Qiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Yinyin Xie
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Ruihong Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Tong Yin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Saijuan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China.
| | - Qingyun Li
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China.
| | - Jiang Zhu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China.
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China; National Research Center for Translational Medicine at Shanghai, Shanghai 200025, China.
| |
Collapse
|
19
|
Balestrieri E, Corinaldesi E, Fabi M, Cipriani C, Giudice M, Conti A, Minutolo A, Petrone V, Fanelli M, Miele MT, Andreozzi L, Guida F, Filice E, Meli M, Grelli S, Rasi G, Toschi N, Torcetta F, Matteucci C, Lanari M, Sinibaldi-Vallebona P. Preliminary Evidence of the Differential Expression of Human Endogenous Retroviruses in Kawasaki Disease and SARS-CoV-2-Associated Multisystem Inflammatory Syndrome in Children. Int J Mol Sci 2023; 24:15086. [PMID: 37894766 PMCID: PMC10606856 DOI: 10.3390/ijms242015086] [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: 08/23/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a postinfectious sequela of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with some clinical features overlapping with Kawasaki disease (KD). Our research group and others have highlighted that the spike protein of SARS-CoV-2 can trigger the activation of human endogenous retroviruses (HERVs), which in turn induces inflammatory and immune reactions, suggesting HERVs as contributing factors in COVID-19 immunopathology. With the aim to identify new factors involved in the processes underlying KD and MIS-C, we analysed the transcriptional levels of HERVs, HERV-related genes, and immune mediators in children during the acute and subacute phases compared with COVID-19 paediatric patients and healthy controls. The results showed higher levels of HERV-W, HERV-K, Syn-1, and ASCT-1/2 in KD, MIS-C, and COV patients, while higher levels of Syn-2 and MFSD2A were found only in MIS-C patients. Moreover, KD and MIS-C shared the dysregulation of several inflammatory and regulatory cytokines. Interestingly, in MIS-C patients, negative correlations have been found between HERV-W and IL-10 and between Syn-2 and IL-10, while positive correlations have been found between HERV-K and IL-10. In addition, HERV-W expression positively correlated with the C-reactive protein. This pilot study supports the role of HERVs in inflammatory diseases, suggesting their interplay with the immune system in this setting. The elevated expression of Syn-2 and MFSD2A seems to be a distinctive trait of MIS-C patients, allowing to distinguish them from KD ones. The understanding of pathological mechanisms can lead to the best available treatment for these two diseases, limiting complications and serious outcomes.
Collapse
Affiliation(s)
- Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Elena Corinaldesi
- Pediatric Unit, Ramazzini Hospital, 41012 Carpi, Italy; (E.C.); (F.T.)
| | - Marianna Fabi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Chiara Cipriani
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Martina Giudice
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Allegra Conti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (N.T.)
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Laura Andreozzi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Fiorentina Guida
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Emanuele Filice
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Matteo Meli
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Guido Rasi
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (N.T.)
- Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA 02129, USA
| | | | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
| | - Marcello Lanari
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy; (L.A.); (F.G.); (E.F.); (M.M.); (M.L.)
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (M.G.); (A.M.); (V.P.); (M.F.); (M.T.M.); (S.G.); (G.R.); (C.M.); (P.S.-V.)
- National Research Council, Institute of Translational Pharmacology, 00133 Rome, Italy
| |
Collapse
|
20
|
Zarpoosh M, Amirian P. COVID-associated arthritis after severe and non-severe COVID-19: A systematic review. Immun Inflamm Dis 2023; 11:e1035. [PMID: 37904701 PMCID: PMC10566449 DOI: 10.1002/iid3.1035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/03/2023] [Accepted: 09/22/2023] [Indexed: 11/01/2023] Open
Abstract
AIM Since the coronavirus outbreak became a global health emergency in 2020, various immune-based effects, such as inflammatory arthritis (IA), have been recorded. This study aimed to determine the role of COVID-19 severity on post-COVID arthritis. METHODS We systematically reviewed 95 patients who developed arthritis after severe and non-severe COVID-19 infection by searching the databases, including PubMed, SCOPUS, and EMBASE. We used the term "COVID-associated arthritis" because there was no definite diagnostic method for classifying arthritides after COVID-19 infection, and the diagnosed arthritis types were based on the authors' viewpoints. RESULTS After evaluating the data between the two severe and non-severe COVID-19-infected groups of patients, the results showed that the COVID-19 severity may affect the pattern of joint involvement in IA. In both groups, combination therapy, including oral nonsteroidal anti-inflammatory drugs with different types of corticosteroids, was the most common treatment. In addition, the mean age and comorbidities rate was higher in the severe COVID-19 group. Even though the patients in the severe COVID-19 group developed more serious COVID-19 symptoms, they experienced milder arthritis with better outcomes and more delayed onsets that required less aggressive therapy. CONCLUSION We conclude that there may be an inverse relationship between COVID-19 severity and arthritis severity, possibly due to weaker immunity conditions following immunosuppressant treatments in patients with severe COVID-19.
Collapse
Affiliation(s)
- Mahsa Zarpoosh
- Kermanshah University of Medical Science (KUMS)KermanshahIran
| | - Parsa Amirian
- Kermanshah University of Medical Science (KUMS)KermanshahIran
| |
Collapse
|
21
|
Muyayalo KP, Gong GS, Kiyonga Aimeé K, Liao AH. Impaired immune response against SARS-CoV-2 infection is the major factor indirectly altering reproductive function in COVID-19 patients: a narrative review. HUM FERTIL 2023; 26:778-796. [PMID: 37811836 DOI: 10.1080/14647273.2023.2262757] [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: 02/28/2023] [Accepted: 05/25/2023] [Indexed: 10/10/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease affecting multiple systems and organs, including the reproductive system. SARS-CoV-2, the virus that causes COVID-19, can damage reproductive organs through direct (angiotensin converting enzyme-2, ACE-2) and indirect mechanisms. The immune system plays an essential role in the homeostasis and function of the male and female reproductive systems. Therefore, an altered immune response related to infectious and inflammatory diseases can affect reproductive function and fertility in both males and females. This narrative review discussed the dysregulation of innate and adaptive systems induced by SARS-CoV-2 infection. We reviewed the evidence showing that this altered immune response in COVID-19 patients is the major indirect mechanism leading to adverse reproduction outcomes in these patients. We summarized studies reporting the long-term effect of SARS-CoV-2 infection on women's reproductive function and proposed the chronic inflammation and chronic autoimmunity characterizing long COVID as potential underlying mechanisms. Further studies are needed to clarify the role of autoimmunity and chronic inflammation (long COVID) in altered female reproduction function in COVID-19.
Collapse
Affiliation(s)
- Kahindo P Muyayalo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Department of Obstetrics and Gynecology, University of Kinshasa, Kinshasa, D. R. Congo
| | - Guang-Shun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Kahindo Kiyonga Aimeé
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, People's Republic of China
- Department of Tropical Medicine Infectious and Parasitic Diseases, University of Kinshasa, Kinshasa, D. R. Congo
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| |
Collapse
|
22
|
Sun Y, Dian Y, Gao Q, Deng G. Immunophenotyping characteristics and outcome of COVID-19 patients: peripheral blood CD8+T cell as a prognostic biomarker for patients with Nirmatrelvir. Front Immunol 2023; 14:1227905. [PMID: 37799722 PMCID: PMC10547864 DOI: 10.3389/fimmu.2023.1227905] [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: 05/24/2023] [Accepted: 09/01/2023] [Indexed: 10/07/2023] Open
Abstract
Background Nirmatrelvir has been authorized for the treatment of both hospitalized and non-hospitalized COVID-19 patients. However, the association between T lymphocyte subsets and the outcome of hospitalized COVID-19 patients treated with oral Nirmatrelvir has not been investigated. The objective of this study was to examine whether lymphocyte subsets could serve as biomarkers to assess the risk of mortality in COVID-19 patients undergoing Nirmatrelvir treatment, with the aim of enhancing medication management for COVID-19 patients. Methods We conducted a retrospective cohort study at the Xiangya Hospital of Central South University in China between December 5, 2022 and January 31, 2023. The study reported demographic, clinical, T lymphocyte subsets, and inflammatory cytokine data of COVID-19 patients. We evaluated the associations of T lymphocyte subsets on admission with the composite outcome or death of patients using univariate and multivariable Cox regression analyses with hazards ratios (HRs) and 95% confidence intervals (CIs). Results We identified 2118 hospitalized COVID-19 patients during the study period, and conducted a follow-up of up to 38 days. Of these, 131 patients received Nirmatrelvir, with 56 (42.7%) in the composite outcome group, and 75 (57.3%) in the non-composite outcome group. Additionally, 101 (77.1%) patients were discharged, while 30 (22.9%) died. Our results showed a significant decrease in the CD3+, CD4+, and CD8+ T cell counts of patients in the composite outcome group and mortality group compared to the non-composite outcome group and discharged group, respectively. Multivariate Cox regression analysis showed that the significant decrease in CD8+ T cell count in peripheral blood was independently associated with the composite outcome in COVID-19 patients treated with Nirmatrelvir, with an HR of 1.96 (95%CI: 1.01-3.80). The significant decrease in CD4+ and CD8+ T cell counts in peripheral blood increased the hazard of developing mortality, with HRs of 6.48 (95%CI: 1.47-28.63) and 3.75 (95%CI: 1.27-11.11), respectively. Conclusion Our study revealed a significant positive correlation between a decrease in CD8+ T cell counts and progression and mortality of hospitalized COVID-19 patients treated with Nirmatrelvir. Lower counts (/μL) of CD8+ T cell (<201) were associated with a higher risk of in-hospital severity and death. Our findings may provide valuable references for physicians in optimizing the use of Nirmatrelvir.
Collapse
Affiliation(s)
- Yuming Sun
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Centre of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Centre of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Centre of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Centre of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Qian Gao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Centre of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Centre of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Clinical Laboratory Department, Xiangya Hospital, Central South University, Changsha, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Centre of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Centre of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Changsha, China
| |
Collapse
|
23
|
Isazadeh A, Heris JA, Shahabi P, Mohammadinasab R, Shomali N, Nasiri H, Valedkarimi Z, Khosroshahi AJ, Hajazimian S, Akbari M, Sadeghvand S. Pattern-recognition receptors (PRRs) in SARS-CoV-2. Life Sci 2023; 329:121940. [PMID: 37451397 DOI: 10.1016/j.lfs.2023.121940] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Pattern recognition receptors (PRRs) are specific sensors that directly recognize various molecules derived from viral or bacterial pathogens, senescent cells, damaged cells, and apoptotic cells. These sensors act as a bridge between nonspecific and specific immunity in humans. PRRs in human innate immunity were classified into six types: toll-like receptors (TLR), C-type lectin receptors (CLRs), nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs), absent in melanoma 2 (AIM2)-like receptors (ALRs), retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), and cyclic GMP-AMP (cGAMP) synthase (cGAS). Numerous types of PRRs are responsible for recognizing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, which is immensely effective in prompting interferon responses. Detection of SARS-CoV-2 infection by PRRs causes the initiation of an intracellular signaling cascade and subsequently the activation of various transcription factors that stimulate the production of cytokines, chemokines, and other immune-related factors. Therefore, it seems that PRRs are a promising potential therapeutic approach for combating SARS-CoV-2 infection and other microbial infections. In this review, we have introduced the current knowledge of various PRRs and related signaling pathways in response to SARS-CoV-2.
Collapse
Affiliation(s)
- Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Shomali
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Nasiri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Valedkarimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saba Hajazimian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Shahram Sadeghvand
- Department of Pediatrics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Yan C, Hu M, Dai R. Safety and efficacy of mesenchymal stem cells in COVID-19 patients: A systematic review and meta-analysis. Immun Inflamm Dis 2023; 11:e1000. [PMID: 37773722 PMCID: PMC10515507 DOI: 10.1002/iid3.1000] [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: 06/26/2023] [Revised: 08/03/2023] [Accepted: 08/22/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Coronavirus disease-19 (COVID-19) is a zoonotic disease that has become a global pandemic. The fast evolution of the COVID-19 pandemic and persist problems make COVID-19 highly infectious; publicly accessible literature and other sources of information continue to expand in volume. The mesenchymal stem cells (MSCs) therapy efficacy for COVID-19 is debatable. OBJECTIVE This systematic review and meta-analysis (SRMA) aimed to evaluate the usefulness of MSCs in treating COVID-19. METHODS Relevant publications were retrieved from databases up to April 30, 2022. In the case of dichotomous data, the 95% confidence intervals (CIs) and pooled risk ratio (RR) were estimated with a random effects model (REM) or fixed effects model (FEM). The pooled mean difference (MD) and 95% CIs were calculated with REM or FEM in continuous data. In the outcomes, studies with insufficient or unusable data were reported descriptively. RESULTS A total of eight randomized controlled trials (RCTs) with 464 people were chosen for this SRMA. Relative to the control group, mortality was significantly lower in the MSCs group (RR: 0.66, 95% CI: 0.44, 0.99, Z = 2.01, p = .04); other secondary outcomes, such as the clinical symptom improvement rate improved in the MSCs group (RR: 1.44, 95% CI: 1.05, 1.99, Z = 2.24, p = .03), clinical symptom improvement time (MD: -4.01, 95% CI: -6.33, -1.68, Z = 3.38, p = .0007), C-reactive protein (CRP) (MD: -39.16, 95% CI: -44.39, -33.94, Z = 14.70, p < .00001) and days to hospital discharge (MD: -3.83, 95% CI: -6.19, -1.48, Z = 3.19, p = .001) reduced significantly in MSCs group. However, the adverse reaction incidence did not change significantly. CONCLUSIONS MSCs are a viable therapy option for COVID-19 because of their safety and potential efficacy. With no significant adverse effects, MSCs can reduce mortality, clinical symptom improvement time, and days to hospital discharge, improve clinical symptoms, and reduce inflammatory cytokines CRP in COVID-19. However, further high-quality clinical studies are required to confirm these results.
Collapse
Affiliation(s)
- Cai Yan
- Xiangtan Central HospitalDepartment of Infectious diseasesXiangtanHunan provincePeople's Republic of China
| | - Minjie Hu
- The First Affiliated Hospital, Department of Cardiothoracic Surgery, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan provincePeople's Republic of China
| | - Rongjuan Dai
- The First Affiliated Hospital, Department of Infectious Diseases, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan provincePeople's Republic of China
| |
Collapse
|
25
|
Ahmadi S, Khaledi S. Brain Renin-Angiotensin System: From Physiology to Pathology in Neuronal Complications Induced by SARS-CoV-2. Anal Cell Pathol (Amst) 2023; 2023:8883492. [PMID: 37575318 PMCID: PMC10421715 DOI: 10.1155/2023/8883492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/26/2023] [Accepted: 07/15/2023] [Indexed: 08/15/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2), a key enzyme in the renin-angiotensin system (RAS), is expressed in various tissues and organs, including the central nervous system (CNS). The spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for coronavirus disease-2019 (COVID-19), binds to ACE2, which raises concerns about the potential for viral infection in the CNS. There are numerous reports suggesting a link between SARS-CoV-2 infection and neurological manifestations. This study aimed to present an updated review of the role of brain RAS components, especially ACE2, in neurological complications induced by SARS-CoV-2 infection. Several routes of SARS-CoV-2 entry into the brain have been proposed. Because an anosmia condition appeared broadly in COVID-19 patients, the olfactory nerve route was suggested as an early pathway for SARS-CoV-2 entry into the brain. In addition, a hematogenous route via disintegrations in the blood-brain barrier following an increase in systemic cytokine and chemokine levels and retrograde axonal transport, especially via the vagus nerve innervating lungs, have been described. Common nonspecific neurological symptoms in COVID-19 patients are myalgia, headache, anosmia, and dysgeusia. However, more severe outcomes include cerebrovascular diseases, cognitive impairment, anxiety, encephalopathy, and stroke. Alterations in brain RAS components such as angiotensin II (Ang II) and ACE2 mediate neurological manifestations of SARS-CoV-2 infection, at least in part. Downregulation of ACE2 due to SARS-CoV-2 infection, followed by an increase in Ang II levels, leads to hyperinflammation and oxidative stress, which in turn accelerates neurodegeneration in the brain. Furthermore, ACE2 downregulation in the hypothalamus induces stress and anxiety responses by increasing corticotropin-releasing hormone. SARS-CoV-2 infection may also dysregulate the CNS neurotransmission, leading to neurological complications observed in severe cases of COVID-19. It can be concluded that the neurological manifestations of COVID-19 may be partially associated with changes in brain RAS components.
Collapse
Affiliation(s)
- Shamseddin Ahmadi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Shiler Khaledi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| |
Collapse
|
26
|
Yang MY, Zheng MH, Meng XT, Ma LW, Liang HY, Fan HY. Role of toll-like receptors in the pathogenesis of COVID-19: Current and future perspectives. Scand J Immunol 2023; 98:e13275. [PMID: 38441378 DOI: 10.1111/sji.13275] [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: 10/22/2022] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 03/07/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic underlines a persistent threat of respiratory tract infectious diseases and warrants preparedness for a rapid response. At present, COVID-19 has had a serious social impact and imposed a heavy global burden on public health. The exact pathogenesis of COVID-19 has not been fully elucidated. Since the outbreak of COVID-19, a renewed attention has been brought to Toll-like receptors (TLRs). Available data and new findings have demonstrated that the interaction of human TLRs and SARS-CoV-2 is a vital mediator of COVID-19 immunopathogenesis. TLRs such as TLR2, 4, 7 and 8 are potentially important in viral combat and activation of immunity in patients with COVID-19. Therapeutics targeting TLRs are currently considered promising options against the pandemic. A number of TLR-targeting immunotherapeutics are now being investigated in preclinical studies and different phases of clinical trials. In addition, innovative vaccines based on TLRs under development could be a promising approach for building a new generation of vaccines to solve the current challenges. In this review, we summarize recent progress in the role of TLRs in COVID-19, focusing the new candidate drugs targeting TLRs, the current technology and potential paths forward for employing TLR agonists as vaccine adjuvants.
Collapse
Affiliation(s)
- Ming-Yan Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Mei-Hua Zheng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Xiang-Ting Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Le-Wei Ma
- Ruikang Pharmaceutical Group Co. Ltd., Yantai, China
| | - Hai-Yue Liang
- Yantai Center for Food and Drug Control, Yantai, China
| | - Hua-Ying Fan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
27
|
Starshinova A, Kudryavtsev I, Rubinstein A, Malkova A, Dovgaluk I, Kudlay D. Tuberculosis and COVID-19 Dually Affect Human Th17 Cell Immune Response. Biomedicines 2023; 11:2123. [PMID: 37626620 PMCID: PMC10452633 DOI: 10.3390/biomedicines11082123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19 infection not only profoundly impacts the detection of tuberculosis infection (Tbc) but also affects modality in tuberculosis patient immune response. It is important to determine immune response alterations in latent tuberculosis infection as well as in SARS-CoV-2-infected tuberculosis patients. Such changes may have underlying effects on the development and course of further tuberculosis. Here, we aimed to review the characteristics of immune response in TB patients or convalescent COVID-19 patients with latent TB infection (LTBI). MATERIALS AND METHODS We analyzed the features of immune response in tuberculosis and COVID-19 patients. For this, we analyzed publications released from December 2019 to March 2023; those which were published in accessible international databases ("Medline", "PubMed", "Scopus") and with keywords such as "COVID-19", "SARS-CoV-2", "tuberculosis", "pulmonary tuberculosis", "latent tuberculosis infection", "Treg", "follicular Treg", and "Treg subsets", we considered. RESULTS Through our analysis, we found that tuberculosis patients who had been infected with COVID-19 previously and elevated Th1 and Th2 cell levels. High levels of Th1 and Th2 cells may serve as a positive marker, characterizing activated immune response during TB infection. COVID-19 or post-COVID-19 subjects showed decreased Th17 levels, indicating a lack of tuberculosis development. Moreover, the typical course of tuberculosis is associated with an increase in Treg level, but COVID-19 contributes to a hyperinflammatory response. CONCLUSION According to the data obtained, the course of tuberculosis proceeds in a dissimilar way due to the distinct immune response, elicited by SARS-CoV-2. Importantly, the development of active tuberculosis with a severe course is associated with a decline in Treg levels. Both pathogens lead to disturbed immune responses, increasing the risk of developing severe TB. The insights and findings of this paper may be used to improve the future management of individuals with latent and active tuberculosis.
Collapse
Affiliation(s)
- Anna Starshinova
- Almazov National Medical Research Centre, 197341 St-Petersburg, Russia
| | - Igor Kudryavtsev
- Department of Immunology, Institution of Experimental Medicine, 197022 St-Petersburg, Russia; (I.K.); (A.R.)
| | - Artem Rubinstein
- Department of Immunology, Institution of Experimental Medicine, 197022 St-Petersburg, Russia; (I.K.); (A.R.)
| | - Anna Malkova
- Faculty of Natural Sciences, Ariel University, Ariel 40700, Israel;
| | - Irina Dovgaluk
- Phthisiopulmonology Department, Research Institute of Phthisiopulmonology, 191036 St-Petersburg, Russia;
| | - Dmitry Kudlay
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia;
- Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
| |
Collapse
|
28
|
Antunes MDSM, Sugiyama FHC, Gravina HD, Castro RC, Mercado FJR, de Lima JO, Fontanari C, Frantz FG. COVID-19 inactivated and non-replicating viral vector vaccines induce regulatory training phenotype in human monocytes under epigenetic control. Front Cell Infect Microbiol 2023; 13:1200789. [PMID: 37520439 PMCID: PMC10382685 DOI: 10.3389/fcimb.2023.1200789] [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: 04/05/2023] [Accepted: 05/26/2023] [Indexed: 08/01/2023] Open
Abstract
Background Trained immunity is the enhanced innate immune response resulting from exposure to pathogens or vaccines against an unrelated pathogen stimulus. Certain vaccines induce a memory like response in monocytes and NK cells, leading to modulation in cytokine production, metabolic changes, and modifications in histone patterns. Here, we hypothesized that vaccination against SARS-CoV-2 could induce the training of monocytes in addition to stimulating the adaptive immune response. Methods Therefore, we aimed to investigate the immunophenotyping, cytokine and metabolic profile of monocytes from individuals who were completely immunized with two doses of inactivated COVID-19 vaccine or non-replicating viral vector vaccine. Subsequently, we investigated the epigenetic mechanisms underlying monocyte immune training. As a model of inflammatorychallenge, to understand if the monocytes were trained by vaccination and how they were trained, cells were stimulated in vitro with the endotoxin LPS, an unrelated stimulus that would provoke the effects of training. Results When challenged in vitro, monocytes from vaccinated individuals produced less TNF-α and those who received inactivated vaccine produced less IL-6, whereas vaccination with non-replicating viral vector vaccine induced more IL-10. Inactivated vaccine increased classical monocyte frequency, and both groups showed higher CD163 expression, a hallmark of trained immunity. We observed increased expression of genes involved in glycolysis and reduced IRG1 expression in vaccinated subjects, a gene associated with the tolerance phenotype in monocytes. We observed that both vaccines reduced the chromatin accessibility of genes associated with the inflammatory response, the inactivated COVID-19 vaccine trained monocytes to a regulatory phenotype mediated by histone modifications in the IL6 and IL10 genes, while the non-replicating viral vector COVID-19 vaccine trained monocytes to a regulatory phenotype, mediated by histone modifications in the IL6, IL10, TNF, and CCL2 genes. Conclusions Our findings support the recognized importance of adopting vaccination against SARS CoV-2, which has been shown to be effective in enhancing the adaptive immune response against the virus and reducing mortality and morbidity rates. Here, we provide evidence that vaccination also modulates the innate immune response by controlling the detrimental inflammatory response to unrelated pathogen stimulation.
Collapse
|
29
|
Mabrey FL, Nian H, Yu C, Barnes EM, Malhotra U, Mikacenic C, Goldstein J, O'Mahony DS, Garcia-Diaz J, Finn P, Voelker K, Morrell ED, Self WH, Becker PM, Martin TR, Wurfel MM. Phase 2, randomized, double-blind, placebo-controlled multi-center trial of the clinical and biological effects of anti-CD14 treatment in hospitalized patients with COVID-19 pneumonia. EBioMedicine 2023; 93:104667. [PMID: 37336058 DOI: 10.1016/j.ebiom.2023.104667] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Severe COVID-19 is associated with innate immunopathology, and CD14, a proximal activator of innate immunity, has been suggested as a potential therapeutic target. METHODS We conducted the COVID-19 anti-CD14 Treatment Trial (CaTT), a Phase II randomized, double-blind, placebo-controlled trial at 5 US-sites between April 12, 2021 and November 30, 2021 (NCT04391309). Hospitalized adults with COVID-19 requiring supplemental oxygen (<30 LPM) were randomized 1:1 to receive 4 daily doses of intravenous IC14, an anti-CD14 monoclonal antibody, or placebo. All participants received remdesivir. The primary outcome was time-to-resolution of illness, defined as improvement on the 8-point NIH-Ordinal COVID-19 Scale to category ≤3. Secondary endpoints were safety and exploratory endpoints were pro-inflammatory and antiviral mediators in serum on days 0-5 & 7. The trial was stopped after 40 patients were randomized and treated due to slow enrollment. FINDINGS 40 participants were randomized and treated with IC14 (n = 20) or placebo (n = 20). The median time-to-recovery was 6 days (95% CI, 5-11) in the IC14 group vs. 5 days (95% CI, 4-10) in the Placebo group (recovery rate ratio: 0.77 (95% CI, 0.40, 1.48) (log-rank p = 0.435). The number of adverse events was similar in each group, and no IC14-attributable secondary infections occurred. In repeated-measures mixed-effects analyses, IC14 treatment increased serum sCD14 concentrations, an expected pharmacodynamic effect. Pre-planned, exploratory analyses suggested that IC14 treatment decreased the trajectories of circulating MIP-1β and TNF-α. INTERPRETATION IC14 treatment did not improve time-to-resolution of illness in hypoxemic patients with COVID-19 in this small trial. Results of exploratory analyses suggested IC14 had biologic effects that warrant future clinical investigation. FUNDING National Institute of Allergy and Infectious Diseases.
Collapse
Affiliation(s)
- F Linzee Mabrey
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Hui Nian
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chang Yu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth M Barnes
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Uma Malhotra
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA; Virginia Mason Franciscan Health, Seattle, WA, USA
| | - Carmen Mikacenic
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA; Virginia Mason Franciscan Health, Seattle, WA, USA
| | - Julia Goldstein
- National Institute of Allergy and Infectious Diseases, National Institute of Health, USA
| | - D Shane O'Mahony
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, WA, USA
| | | | - Patricia Finn
- University of Illinois Hospital and Health Sciences System, University of Illinois College of Medicine, Chicago, IL, USA
| | - Kirk Voelker
- Sarasota Memorial Healthcare System, Sarasota, FL, USA
| | - Eric D Morrell
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Wesley H Self
- Vanderbilt Institute for Clinical and Translational Research and Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Patrice M Becker
- National Institute of Allergy and Infectious Diseases, National Institute of Health, USA
| | - Thomas R Martin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
30
|
Opsteen S, Files JK, Fram T, Erdmann N. The role of immune activation and antigen persistence in acute and long COVID. J Investig Med 2023; 71:545-562. [PMID: 36879504 PMCID: PMC9996119 DOI: 10.1177/10815589231158041] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/09/2023] [Accepted: 01/24/2023] [Indexed: 03/08/2023]
Abstract
In late 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered the global coronavirus disease 2019 (COVID-19) pandemic. Although most infections cause a self-limited syndrome comparable to other upper respiratory viral pathogens, a portion of individuals develop severe illness leading to substantial morbidity and mortality. Furthermore, an estimated 10%-20% of SARS-CoV-2 infections are followed by post-acute sequelae of COVID-19 (PASC), or long COVID. Long COVID is associated with a wide variety of clinical manifestations including cardiopulmonary complications, persistent fatigue, and neurocognitive dysfunction. Severe acute COVID-19 is associated with hyperactivation and increased inflammation, which may be an underlying cause of long COVID in a subset of individuals. However, the immunologic mechanisms driving long COVID development are still under investigation. Early in the pandemic, our group and others observed immune dysregulation persisted into convalescence after acute COVID-19. We subsequently observed persistent immune dysregulation in a cohort of individuals experiencing long COVID. We demonstrated increased SARS-CoV-2-specific CD4+ and CD8+ T-cell responses and antibody affinity in patients experiencing long COVID symptoms. These data suggest a portion of long COVID symptoms may be due to chronic immune activation and the presence of persistent SARS-CoV-2 antigen. This review summarizes the COVID-19 literature to date detailing acute COVID-19 and convalescence and how these observations relate to the development of long COVID. In addition, we discuss recent findings in support of persistent antigen and the evidence that this phenomenon contributes to local and systemic inflammation and the heterogeneous nature of clinical manifestations seen in long COVID.
Collapse
Affiliation(s)
- Skye Opsteen
- Division of Infectious Diseases, Department
of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jacob K Files
- Division of Infectious Diseases, Department
of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tim Fram
- Division of Infectious Diseases, Department
of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nathan Erdmann
- Division of Infectious Diseases, Department
of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
31
|
Qudus MS, Tian M, Sirajuddin S, Liu S, Afaq U, Wali M, Liu J, Pan P, Luo Z, Zhang Q, Yang G, Wan P, Li Y, Wu J. The roles of critical pro-inflammatory cytokines in the drive of cytokine storm during SARS-CoV-2 infection. J Med Virol 2023; 95:e28751. [PMID: 37185833 DOI: 10.1002/jmv.28751] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/17/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023]
Abstract
In patients with severe COVID-19, acute respiratory distress syndrome (ARDS), multiple organ dysfunction syndrome (MODS), and even mortality can result from cytokine storm, which is a hyperinflammatory medical condition caused by the excessive and uncontrolled release of pro-inflammatory cytokines. High levels of numerous crucial pro-inflammatory cytokines, such as interleukin-1 (IL-1), IL-2, IL-6, tumor necrosis factor-α, interferon (IFN)-γ, IFN-induced protein 10 kDa, granulocyte-macrophage colony-stimulating factor, monocyte chemoattractant protein-1, and IL-10 and so on, have been found in severe COVID-19. They participate in cascade amplification pathways of pro-inflammatory responses through complex inflammatory networks. Here, we review the involvements of these critical inflammatory cytokines in SARS-CoV-2 infection and discuss their potential roles in triggering or regulating cytokine storm, which can help to understand the pathogenesis of severe COVID-19. So far, there is rarely effective therapeutic strategy for patients with cytokine storm besides using glucocorticoids, which is proved to result in fatal side effects. Clarifying the roles of key involved cytokines in the complex inflammatory network of cytokine storm will help to develop an ideal therapeutic intervention, such as neutralizing antibody of certain cytokine or inhibitor of some inflammatory signal pathways.
Collapse
Affiliation(s)
- Muhammad Suhaib Qudus
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Mingfu Tian
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Summan Sirajuddin
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Uzair Afaq
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Muneeba Wali
- Department of Allied Health Sciences, CECOS University of IT and Emerging Sciences, Peshawar, Pakistan
| | - Jinbiao Liu
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Pan Pan
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Zhen Luo
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Qiwei Zhang
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Ge Yang
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Pin Wan
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Yongkui Li
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| |
Collapse
|
32
|
Zornikova KV, Sheetikov SA, Rusinov AY, Iskhakov RN, Bogolyubova AV. Architecture of the SARS-CoV-2-specific T cell repertoire. Front Immunol 2023; 14:1070077. [PMID: 37020560 PMCID: PMC10067759 DOI: 10.3389/fimmu.2023.1070077] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/08/2023] [Indexed: 03/22/2023] Open
Abstract
The T cell response plays an indispensable role in the early control and successful clearance of SARS-CoV-2 infection. However, several important questions remain about the role of cellular immunity in COVID-19, including the shape and composition of disease-specific T cell repertoires across convalescent patients and vaccinated individuals, and how pre-existing T cell responses to other pathogens—in particular, common cold coronaviruses—impact susceptibility to SARS-CoV-2 infection and the subsequent course of disease. This review focuses on how the repertoire of T cell receptors (TCR) is shaped by natural infection and vaccination over time. We also summarize current knowledge regarding cross-reactive T cell responses and their protective role, and examine the implications of TCR repertoire diversity and cross-reactivity with regard to the design of vaccines that confer broader protection against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ksenia V. Zornikova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Saveliy A. Sheetikov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander Yu Rusinov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Rustam N. Iskhakov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Apollinariya V. Bogolyubova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- *Correspondence: Apollinariya V. Bogolyubova,
| |
Collapse
|
33
|
Liu W, Han F, Wan M, Yang XZ. Integrated bioinformatics analysis identifies shared immune changes between ischemic stroke and COVID 19. Front Immunol 2023; 14:1102281. [PMID: 36969251 PMCID: PMC10030956 DOI: 10.3389/fimmu.2023.1102281] [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: 11/18/2022] [Accepted: 02/23/2023] [Indexed: 03/10/2023] Open
Abstract
Although COVID-19 is primarily a respiratory disease, its neurological complications, such as ischemic stroke (IS), have aroused growing concerns and reports. However, the molecular mechanisms that underlie IS and COVID-19 are not well understood. Therefore, we implemented transcriptomic analysis from eight GEO datasets consist of 1191 samples to detect common pathways and molecular biomarkers in IS and COVID-19 that help understand the linkage between them. Differentially expressed genes (DEGs) were detected for IS and COVID-19 separately for finding shared mechanisms and we found that immune-related pathways were outlined with statistical significance. JAK2, which was identified as a hub gene, was supposed to be a potential therapeutic gene targets during the immunological process of COVID-19 and IS. Besides, we found a decrease in the proportion of CD8+ T and T helper 2 cells in the peripheral circulation of both COVID and IS patients, and NCR3 expression was significantly correlated with this change. In conclusion, we demonstrated that transcriptomic analyses reported in this study could make a deeper understanding of the common mechanism and might be promising for effective therapeutic for IS and COVID-19.
Collapse
Affiliation(s)
- Wenhao Liu
- Eight-year program of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyao Wan
- Eight-year program of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Zhuang Yang
- Medical Research Center, State Key laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Xin-Zhuang Yang,
| |
Collapse
|
34
|
Eryılmaz-Eren E, Köker MY, Ulu-Kılıç A, Hürmet-Öz HT, Ay-Altıntop Y, Saatçi E, Özsoy S, Kılınç-Toker A, Topaloğlu US, Yüksel RC, Avcılar H, Beştepe-Dursun Z, Çelik İ. CD4 + T-Cell Depression is Linked to the Severity of COVID-19 and Predicts Mortality. INFECTIOUS DISEASES & CLINICAL MICROBIOLOGY 2023; 5:23-30. [PMID: 38633909 PMCID: PMC10985810 DOI: 10.36519/idcm.2023.190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/02/2022] [Indexed: 04/19/2024]
Abstract
Objective Most patients with coronavirus disease (COVID-19) have abnormalities of lymphocyte subsets. This study aimed to determine the distribution of lymphocytes in patients with various severity levels of COVID-19 and to describe the relationship between the CD4+ T helper and prognosis. Materials and Methods Adult (>18 years old) patients with COVID-19 who followed up in a tertiary hospital were included in the study prospectively. Demographic and clinical characteristics of the patients were obtained from the hospital records. Peripheral flow cytometry was studied in patients with different severity of COVID-19 and different prognoses. Next, we analyzed the characteristics and predictive values of lymphocyte subsets in COVID-19 patients. Results Totally 86 patients were included in the study, of which 21 (24.4%) had asymptomatic, 23 (26.7%) had mild/moderate, and 42 (48.8%) had severe/critical COVID-19. Severe/critical patients had lower lymphocyte levels and older age than asymptomatic patients (p<0.001 and p<0.001, respectively). We determined that decreased CD4+ T cell ratio (p<0.001) and CD4+ /CD8+ ratio (p<0.001) were indicative of the severity of the disease. CD4+ T cell ratio on admission (odds ratio [OR]=0.858; p=0.033), day seven CD4+ T cell ratio (OR=0.840; p=0.029), and C-reactive protein (CRP) levels (OR=1.014; p=0.043) were prognostic factors for mortality. According to receiver operating characteristics (ROC) curve analysis, the area under the curve was greater than 0.9 for decreased CD4 + T cell ratio on admission and the seventh day. Conclusion A low CD4+ T helper ratio predicts a poor prognosis. In combination with CRP, it can be used in clinical follow-up.
Collapse
Affiliation(s)
- Esma Eryılmaz-Eren
- Department of Infectious Diseases and Clinical Microbiology,
Kayseri City Education and Research Hospital, Kayseri, Turkey
| | - Mustafa Yavuz Köker
- Department of Immunology, Erciyes University School of Medicine,
Kayseri, Turkey
| | - Aysegul Ulu-Kılıç
- Department of Infectious Diseases and Clinical Microbiology,
Erciyes University School of Medicine, Kayseri, Turkey
| | - Hatice Tuna Hürmet-Öz
- Department of Medical Microbiology, Kayseri City Education and
Research Hospital, Kayseri, Turkey
| | - Yasemin Ay-Altıntop
- Department of Medical Microbiology, Kayseri City Education and
Research Hospital, Kayseri, Turkey
| | - Esma Saatçi
- Department of Medical Microbiology, Kayseri City Education and
Research Hospital, Kayseri, Turkey
| | - Sevil Özsoy
- Department of Immunology, Erciyes University School of Medicine,
Kayseri, Turkey
| | - Ayşin Kılınç-Toker
- Department of Internal Medicine, Kayseri City Education and
Research Hospital, Kayseri, Turkey
| | - Ulaş Serkan Topaloğlu
- Department of Internal Medicine and Intensive Care, Erciyes
University School of Medicine, Kayseri, Turkey
| | - Recep Civan Yüksel
- Department of Infectious Diseases and Clinical Microbiology,
Kayseri City Education and Research Hospital, Kayseri, Turkey
| | - Hüseyin Avcılar
- Department of Immunology, Erciyes University School of Medicine,
Kayseri, Turkey
| | - Zehra Beştepe-Dursun
- Department of Infectious Diseases and Clinical Microbiology,
Kayseri City Education and Research Hospital, Kayseri, Turkey
| | - İlhami Çelik
- Department of Infectious Diseases and Clinical Microbiology,
Kayseri City Education and Research Hospital, Kayseri, Turkey
| |
Collapse
|
35
|
Hattab D, Amer MFA, Mohd Gazzali A, Chuah LH, Bakhtiar A. Current status in cellular-based therapies for prevention and treatment of COVID-19. Crit Rev Clin Lab Sci 2023:1-25. [PMID: 36825325 DOI: 10.1080/10408363.2023.2177605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) outbreaks that resulted in a catastrophic threat to global health, with more than 500 million cases detected and 5.5 million deaths worldwide. Patients with a COVID-19 infection presented with clinical manifestations ranging from asymptomatic to severe symptoms, resulting in acute lung injury, acute respiratory distress syndrome, and even death. Immune dysregulation through delayed innate immune response or impairment of the adaptive immune response is the key contributor to the pathophysiology of COVID-19 and SARS-CoV-2-induced cytokine storm. Symptomatic and supportive therapy is the fundamental strategy in treating COVID-19 infection, including antivirals, steroid-based therapies, and cell-based immunotherapies. Various studies reported substantial effects of immune-based therapies for patients with COVID-19 to modulate the over-activated immune system while simultaneously refining the body's ability to destroy the virus. However, challenges may arise from the complexity of the disease through the genetic variance of the virus itself and patient heterogeneity, causing increased transmissibility and heightened immune system evasion that rapidly change the intervention and prevention measures for SARS-CoV-2. Cell-based therapy, utilizing stem cells, dendritic cells, natural killer cells, and T cells, among others, are being extensively explored as other potential immunological approaches for preventing and treating SARS-CoV-2-affected patients the similar process was effectively proven in SARS-CoV-1 and MERS-CoV infections. This review provides detailed insights into the innate and adaptive immune response-mediated cell-based immunotherapies in COVID-19 patients. The immune response linking towards engineered autologous or allogenic immune cells for either treatment or preventive therapies is subsequently highlighted in an individual study or in combination with several existing treatments. Up-to-date data on completed and ongoing clinical trials of cell-based agents for preventing or treating COVID-19 are also outlined to provide a guide that can help in treatment decisions and future trials.
Collapse
Affiliation(s)
- Dima Hattab
- Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mumen F A Amer
- Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Amirah Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Lay Hong Chuah
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
| | - Athirah Bakhtiar
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
| |
Collapse
|
36
|
Simultaneous determination of twenty-nine active compounds in fuzhengjiedu granules by HPLC-QQQ-MS/MS. Heliyon 2023; 9:e13675. [PMID: 36873156 PMCID: PMC9975104 DOI: 10.1016/j.heliyon.2023.e13675] [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: 11/28/2022] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
As an empirical medicine of traditional Chinese medicine, Fuzhengjiedu Granules have shown an effect against COVID-19 in clinical and inflammatory animal models. It is formulated with eight herbs, including Aconiti Lateralis Radix Praeparata, Zingiberis Rhizoma, Glycyrrhizae Radix Et Rhizoma, Lonicerae Japonicae Flos, Gleditsiae Spina, Fici Radix, Pogostemonis Herba, and Citri Reticulatae Pericarpium. This study established a high-performance liquid chromatography-triple quadrupole mass spectrometry (HPLC-QQQ-MS/MS) method by simultaneously determining 29 active compounds in the granules with significant content differences. Separation by gradient elution using acetonitrile and water (0.1% formic acid) as mobile phases was performed on a Waters Acquilty UPLC T3 column (2.1 mm × 100 mm, 1.7 μm). A triple quadrupole mass spectrometer, operating in positive and negative ionization modes, was used for multiple reaction monitoring to detect the 29 compounds. All calibration curves showed good linear regression (r2 > 0.998). RSDs of precision, reproducibility, and stability of active compounds were all lower than 5.0%. The recovery rates were 95.4-104.9%, with RSDs< 5.0%. This method was successfully used to analyze the samples, and the results showed that 26 representative active components from 8 herbs were detected in the granules. While aconitine, mesaconitine, and hypaconitine were not detected, indicating that the existing samples were safe. The granules had the maximum and minimum content of hesperidin (27.3 ± 0.375 mg/g) and benzoylaconine (38.2 ± 0.759 ng/g). To conclude, a fast, accurate, sensitive, and reliable HPLC-QQQ-MS/MS method was established, which can simultaneously detect 29 active compounds that have a considerable difference in the content of Fuzhengjiedu Granules. This study can be used to control the quality and safety of Fuzhengjiedu Granules and provide a basis and guarantee for further experimental research and clinical application.
Collapse
|
37
|
Abstract
Coronavirus disease (COVID-19) is an infectious airborne viral pneumonia caused by a novel virus belonging to the family coronaviridae. On February 11, 2019, the Internal Committee on Taxonomy of Virus (ICTV) announced the name of the novel virus as "severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). One of the proteins present on its membrane i.e. the Spike protein is responsible for the attachment of the virus to the host. It spreads through the salivary droplets released when an infected person sneezes or coughs. The best way to slow down the disease is by protecting self by washing hands and using the disinfectant. Most of the infected people experience mild to moderate breathing issues. Serious illness might develop in people with underlying cardiovascular problems, diabetes and other immuno-compromised diseases. To date, there is no effective medicine available in the market which is effective in COVID-19. However, healthcare professionals are using ritonavir, flavipiravir, lopinavir, hydroxychloroquine and remdesivir. Along with the medicines, some countries are using convalescent plasma and mesenchymal stem cells for treatment. Till date, it has claimed millions of death worldwide. In this detailed review, we have discussed the structure of SARS-CoV-2, essential proteins, its lifecycle, transmission, symptoms, pathology, clinical features, diagnosis, prevention, treatment and epidemiology of the disease.
Collapse
Affiliation(s)
- Heena Rehman
- Department of Biochemistry, Jamia Hamdard, New Delhi, India
| | - Md Iftekhar Ahmad
- Department of Pharmaceutics, Shri Gopichand College of Pharmacy, Baghpat, India
| |
Collapse
|
38
|
Ongoing Treatment with a Spore-Based Probiotic Containing Five Strains of Bacillus Improves Outcomes of Mild COVID-19. Nutrients 2023; 15:nu15030488. [PMID: 36771194 PMCID: PMC9920365 DOI: 10.3390/nu15030488] [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: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Spore-based Bacillus probiotic treatment improves intestinal health. The intestinal microbiota influences both the innate and adaptive immune responses. As such, the influence of ongoing spore-based probiotic treatment (five probiotic strains of Bacillus) on the clinical outcomes of mild COVID-19 was evaluated in this retrospective, observational study. Demographics, medical history, probiotic use, and COVID-19 symptom information were collected. The study included 120 patients with a PCR-confirmed SARS-CoV-2 infection and mild COVID-19 symptoms. The probiotic group (n = 60) comprised patients with ongoing probiotic treatment (≥1 month); the control group comprised patients not taking probiotics (n = 60). The primary outcome was time to symptom resolution; secondary outcomes included time to fever resolution and presence of digestive symptoms. The probiotic group had a significantly shorter time to symptom resolution (mean (95% confidence interval) days: control group, 8.48 (6.56, 10.05); probiotic group, 6.63 (5.56; 6.63); p = 0.003) and resolution of fever (control group, 2.67 (1.58, 3.61); probiotic group, 1.48 (1.21, 2.03); p < 0.001). More patients in the probiotic group (n = 53) than in the control group (n = 34) did not have digestive symptoms (p < 0.001). Among adults with mild COVID-19, participants receiving ongoing probiotic treatment had a shorter clinical course, and fewer had digestive symptoms compared with those not taking probiotics.
Collapse
|
39
|
Al-Rifai RH, Alhosani F, Abuyadek R, Atef S, Donnelly JG, Leinberger-Jabari A, Ahmed LA, Altrabulsi B, Alatoom A, Alsuwaidi AR, AbdelWareth L. Evaluation of post-vaccination immunoglobulin G antibodies and T-cell immune response after inoculation with different types and doses of SARS-CoV-2 vaccines: A retrospective cohort study. Front Med (Lausanne) 2023; 9:1092646. [PMID: 36703898 PMCID: PMC9871809 DOI: 10.3389/fmed.2022.1092646] [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: 11/08/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction The induction and speed of production of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) immune biomarkers may vary by type and number of inoculated vaccine doses. This study aimed to explore variations in SARS-CoV-2 anti-spike (anti-S), anti-nucleocapsid (anti-N), and neutralizing immunoglobulin G (IgG) antibodies, and T-cell response by type and number of SARS-CoV-2 vaccine doses received. Methods In a naturally exposed and SARS-CoV-2-vaccinated population, we quantified the anti-S, anti-N, and neutralizing IgG antibody concentration and assessed T-cell response. Data on socio-demographics, medical history, and history of SARS-CoV-2 infection and vaccination were collected. Furthermore, nasal swabs were collected to test for SARS-CoV-2 infection. Confounder-adjusted association between having equal or more than a median concentration of the three IgG antibodies and T-cell response by number and type of the inoculated vaccines was quantified. Results We surveyed 952 male participants with a mean age of 35.5 years ± 8.4 standard deviations. Of them, 52.6% were overweight/obese, and 11.7% had at least one chronic comorbidity. Of the participants, 1.4, 0.9, 20.2, 75.2, and 2.2% were never vaccinated, primed with only one dose, primed with two doses, boosted with only one dose, and boosted with two doses, respectively. All were polymerase chain reaction-negative to SARS-CoV-2. BBIBP-CorV (Sinopharm) was the most commonly used vaccine (92.1%), followed by rAd26-S + rAd5-S (Sputnik V Gam-COVID-Vac) (1.5%) and BNT162b2 (Pfizer-BioNTech) (0.3%). Seropositivity to anti-S, anti-N, and neutralizing IgG antibodies was detected in 99.7, 99.9, and 99.3% of the study participants, respectively. The T-cell response was detected in 38.2% of 925 study participants. Every additional vaccine dose was significantly associated with increased odds of having ≥median concentration of anti-S [adjusted odds ratio (aOR), 1.34; 95% confidence interval (CI): 1.02-1.76], anti-N (aOR, 1.35; 95% CI: 1.03-1.75), neutralizing IgG antibodies (aOR, 1.29; 95% CI: 1.00-1.66), and a T-cell response (aOR, 1.48; 95% CI: 1.12-1.95). Compared with boosting with only one dose, boosting with two doses was significantly associated with increased odds of having ≥median concentration of anti-S (aOR, 13.8; 95% CI: 1.78-106.5), neutralizing IgG antibodies (aOR, 13.2; 95% CI: 1.71-101.9), and T-cell response (aOR, 7.22; 95% CI: 1.99-26.5) although not with anti-N (aOR, 0.41; 95% CI: 0.16-1.08). Compared with priming and subsequently boosting with BBIBP-CorV, all participants who were primed with BBIBP-CorV and subsequently boosted with BNT162b2 had ≥median concentration of anti-S and neutralizing IgG antibodies and 14.6-time increased odds of having a T-cell response (aOR, 14.63; 95% CI: 1.78-120.5). Compared with priming with two doses, boosting with the third dose was not associated, whereas boosting with two doses was significantly associated with having ≥median concentration of anti-S (aOR, 14.20; 95% CI: 1.85-109.4), neutralizing IgG (aOR, 13.6; 95% CI: 1.77-104.3), and T-cell response (aOR, 7.62; 95% CI: 2.09-27.8). Conclusion Achieving and maintaining a high blood concentration of protective immune biomarkers that predict vaccine effectiveness is very critical to limit transmission and contain outbreaks. In this study, boosting with only one dose or with only BBIBP-CorV after priming with BBIBP-CorV was insufficient, whereas boosting with two doses, particularly boosting with the mRNA-based vaccine, was shown to be associated with having a high concentration of anti-S, anti-N, and neutralizing IgG antibodies and producing an efficient T-cell response.
Collapse
Affiliation(s)
- Rami H. Al-Rifai
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates,*Correspondence: Rami H. Al-Rifai,
| | - Farida Alhosani
- Abu Dhabi Public Health Center–ADPHC, Abu Dhabi, United Arab Emirates
| | - Rowan Abuyadek
- Abu Dhabi Public Health Center–ADPHC, Abu Dhabi, United Arab Emirates,High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Shereen Atef
- National Reference Laboratory, Abu Dhabi, United Arab Emirates,Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Luai A. Ahmed
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Basel Altrabulsi
- National Reference Laboratory, Abu Dhabi, United Arab Emirates,Pathology and Laboratory Medicine Institute (PLMI), Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Adnan Alatoom
- National Reference Laboratory, Abu Dhabi, United Arab Emirates,Pathology and Laboratory Medicine Institute (PLMI), Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Ahmed R. Alsuwaidi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Laila AbdelWareth
- National Reference Laboratory, Abu Dhabi, United Arab Emirates,Pathology and Laboratory Medicine Institute (PLMI), Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
40
|
Mirsharif ES, Chenary MR, Bozorgmehr M, Mohammadi S, Hashemi SM, Ardestani SK, Beigmohammadi M, Abdollahi A, Sadeghipour A, Kariminia A, Tuserkani F, Ghazanfari T. Immunophenotyping characteristics of COVID-19 patients: Peripheral blood CD8+ HLA-DR+ T cells as a biomarker for mortality outcome. J Med Virol 2023; 95:e28192. [PMID: 36192361 PMCID: PMC9874930 DOI: 10.1002/jmv.28192] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 08/19/2022] [Accepted: 09/27/2022] [Indexed: 01/27/2023]
Abstract
INTRODUCTION The goal of this study was to identify biomarker(s) to assign risk of mortality in COVID-19 patients to improve intensive care unit (ICU) and coronary care unit management. A total of 100 confirmed COVID-19 patients admitted at Imam Khomeini Hospital in Tehran, were compared to 70 control subjects. Peripheral blood leukocyte was studied using staining reagents included CD3, CD4, CD8, HLA-DR, CD19, CD16, and CD56. The immunophenotyping analysis was evaluated using the FACSCalibur instrument. To investigate the cell density of lung infiltrating T cells, postmortem slides of needle necropsies taken from the lung tissue of 3 critical patients were evaluated by immunohistochemistry staining. The number of lymphocyte subpopulations was significantly lower in COVID-19 patients than in the control group. Regarding the disease severity, the absolute count of T, NK, and HLA-DR+ T cells were significantly reduced in severe patients compared to the moderate ones. The critical patients had a significantly lower count of CD8-HLA-DR+ T cells than the moderate cases. Regarding the disease mortality, based on univariate analysis, the count of HLA-DR+ T, CD8- HLA-DR+ T, and CD8+ HLA-DR+ T cells was associated with mortality in COVID-19 patients. Receiver operating characteristic curve analysis showed the count of CD8+ HLA-DR+ T cells is the best candidate as a biomarker for mortality outcome. Furthermore, pulmonary infiltration of T cells in the lung tissue showed only slight infiltrations of CD3+ T cells, with an equal percentage of CD4+ and CD8+ T cell subpopulation in the lung tissue. These findings suggest that close monitoring of the value of CD8+ HLA-DR+ T cells in COVID-19 patients may be helpful to identify high-risk patients. However, further studies with larger sample size are needed.
Collapse
Affiliation(s)
| | | | | | - Saeed Mohammadi
- Hematology‐Oncology and Stem Cell Transplantation Research CenterTehran University of Medical SciencesTehranIran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | | - Mohammad‐Taghi Beigmohammadi
- Department of Anesthesiology and Intensive Care, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Alireza Abdollahi
- Department of Pathology, School of Medicine, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Alireza Sadeghipour
- Medicine Department of Pathology, School of Medicine, Oncopathology Research CenterIran University of Medical SciencesTehranIran
| | - Amina Kariminia
- School of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | | | - Tooba Ghazanfari
- Immunoregulation Research CenterShahed UniversityTehranIran,Simorgh Clinical LaboratoryTehranIran
| |
Collapse
|
41
|
Shapiro JR, Roberts CW, Arcovio K, Reade L, Klein SL, Dhakal S. Effects of Biological Sex and Pregnancy on SARS-CoV-2 Pathogenesis and Vaccine Outcomes. Curr Top Microbiol Immunol 2023; 441:75-110. [PMID: 37695426 DOI: 10.1007/978-3-031-35139-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
SARS-CoV-2 is the causative agent of COVID-19 in humans and has resulted in the death of millions of people worldwide. Similar numbers of infections have been documented in males and females; males, however, are more likely than females to be hospitalized, require intensive care unit, or die from COVID-19. The mechanisms that account for this are multi-factorial and are likely to include differential expression of ACE2 and TMPRSS2 molecules that are required for viral entry into hosts cells and sex differences in the immune response, which are due to modulation of cellular functions by sex hormones and differences in chromosomal gene expression. Furthermore, as comorbidities are also associated with poorer outcomes to SARS-CoV-2 infection and several comorbidities are overrepresented in males, these are also likely to contribute to the observed sex differences. Despite their relative better prognosis following infection with SARS-CoV-2, females do have poorer outcomes during pregnancy. This is likely to be due to pregnancy-induced changes in the immune system that adversely affect viral immunity and disruption of the renin-angiotensin system. Importantly, vaccination reduces the severity of disease in males and females, including pregnant females, and there is no evidence that vaccination has any adverse effects on the outcomes of pregnancy.
Collapse
Affiliation(s)
- Janna R Shapiro
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Craig W Roberts
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Kasandra Arcovio
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Lisa Reade
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Sabra L Klein
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
42
|
Lundstrom K, Hromić-Jahjefendić A, Bilajac E, Aljabali AAA, Baralić K, Sabri NA, Shehata EM, Raslan M, Ferreira ACBH, Orlandi L, Serrano-Aroca Á, Tambuwala MM, Uversky VN, Azevedo V, Alzahrani KJ, Alsharif KF, Halawani IF, Alzahrani FM, Redwan EM, Barh D. COVID-19 signalome: Pathways for SARS-CoV-2 infection and impact on COVID-19 associated comorbidity. Cell Signal 2023; 101:110495. [PMID: 36252792 PMCID: PMC9568271 DOI: 10.1016/j.cellsig.2022.110495] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has been the focus of research the past two years. The major breakthrough was made by discovering pathways related to SARS-CoV-2 infection through cellular interaction by angiotensin-converting enzyme (ACE2) and cytokine storm. The presence of ACE2 in lungs, intestines, cardiovascular tissues, brain, kidneys, liver, and eyes shows that SARS-CoV-2 may have targeted these organs to further activate intracellular signalling pathways that lead to cytokine release syndrome. It has also been reported that SARS-CoV-2 can hijack coatomer protein-I (COPI) for S protein retrograde trafficking to the endoplasmic reticulum-Golgi intermediate compartment (ERGIC), which, in turn, acts as the assembly site for viral progeny. In infected cells, the newly synthesized S protein in endoplasmic reticulum (ER) is transported first to the Golgi body, and then from the Golgi body to the ERGIC compartment resulting in the formation of specific a motif at the C-terminal end. This review summarizes major events of SARS-CoV-2 infection route, immune response following host-cell infection as an important factor for disease outcome, as well as comorbidity issues of various tissues and organs arising due to COVID-19. Investigations on alterations of host-cell machinery and viral interactions with multiple intracellular signaling pathways could represent a major factor in more effective disease management.
Collapse
Affiliation(s)
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Esma Bilajac
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan.
| | - Katarina Baralić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Nagwa A Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11865, Egypt.
| | - Eslam M Shehata
- Drug Research Center, Clinical Research and Bioanalysis Department, Cairo 11865, Egypt.
| | - Mohamed Raslan
- Drug Research Center, Clinical Research and Bioanalysis Department, Cairo 11865, Egypt.
| | - Ana Cláudia B H Ferreira
- Campinas State University, Campinas, São Paulo, Brazil; University Center of Lavras (UNILAVRAS), Lavras, Minas Gerais, Brazil.
| | - Lidiane Orlandi
- University Center of Lavras (UNILAVRAS), Lavras, Minas Gerais, Brazil.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain.
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Vasco Azevedo
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Khalaf F Alsharif
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Ibrahim F Halawani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Fuad M Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah, Saudi Arabia.
| | - Debmalya Barh
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur 721172, India.
| |
Collapse
|
43
|
Lei S, Chen X, Wu J, Duan X, Men K. Small molecules in the treatment of COVID-19. Signal Transduct Target Ther 2022; 7:387. [PMID: 36464706 PMCID: PMC9719906 DOI: 10.1038/s41392-022-01249-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 12/11/2022] Open
Abstract
The outbreak of COVID-19 has become a global crisis, and brought severe disruptions to societies and economies. Until now, effective therapeutics against COVID-19 are in high demand. Along with our improved understanding of the structure, function, and pathogenic process of SARS-CoV-2, many small molecules with potential anti-COVID-19 effects have been developed. So far, several antiviral strategies were explored. Besides directly inhibition of viral proteins such as RdRp and Mpro, interference of host enzymes including ACE2 and proteases, and blocking relevant immunoregulatory pathways represented by JAK/STAT, BTK, NF-κB, and NLRP3 pathways, are regarded feasible in drug development. The development of small molecules to treat COVID-19 has been achieved by several strategies, including computer-aided lead compound design and screening, natural product discovery, drug repurposing, and combination therapy. Several small molecules representative by remdesivir and paxlovid have been proved or authorized emergency use in many countries. And many candidates have entered clinical-trial stage. Nevertheless, due to the epidemiological features and variability issues of SARS-CoV-2, it is necessary to continue exploring novel strategies against COVID-19. This review discusses the current findings in the development of small molecules for COVID-19 treatment. Moreover, their detailed mechanism of action, chemical structures, and preclinical and clinical efficacies are discussed.
Collapse
Affiliation(s)
- Sibei Lei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xiaohua Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
44
|
Widyasari K, Jang J, Lee S, Kang T, Kim S. Evaluation of the T cell and B cell response following the administration of COVID-19 vaccines in Korea. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:1013-1024. [PMID: 36261313 PMCID: PMC9516877 DOI: 10.1016/j.jmii.2022.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND The coronavirus disease (COVID-19) has been a worldwide concern since 2019. Vaccines are predicted to be crucial in preventing further outbreaks. The development and kinetics of immune responses determine the efficacy of COVID-19 vaccines. METHODS We measured interferon-gamma (IFN-γ) levels upon administering homologous adenovirus vector-based (ChAdOx1-S [AZ], Ad26.COV2.S [JAN]), mRNA-based (BNT162b2 [PF]; mRNA-1273 [MO]), and heterologous (AZ/PF) vaccines in healthy Korean individuals using two IFN-γ release assays: the Covi-FERON ELISA and T-SPOT Discovery SARS-CoV-2 assay. B cell responses were evaluated by assessing the production of neutralizing antibodies by surrogate virus neutralization assay. The immune response among the vaccine groups was compared after adjusting the vaccination dose and interactions between each group. RESULTS AZ triggered the highest T cell response after the first dose but showed instability after the second. PF and MO yielded stable and higher increments of T and B cell responses compared to AZ. MO yielded a higher immune response than PF. JAN yielded T and B cell responses at lower levels than the other vaccines. The booster dose triggered significant increases in the T and B cell responses and is therefore needed to protect against SARS-CoV-2 given the possibility of waning immune responses. CONCLUSION Administering two doses of mRNA vaccines provides the most effective results among the administered vaccines in triggering the immune response specific to SARS-CoV-2 in healthy Korean individuals. Administration of booster doses demonstrated a significant increase in the immune response and may provide longer protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Kristin Widyasari
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon, 51472, South Korea
| | - Jieun Jang
- Gyeongnam Center for Infectious Disease Control and Prevention, Changwon 51154, South Korea
| | - Seungjun Lee
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon, 51472, South Korea,Gyeongsang National University College of Medicine, Gyeongsang Institute of Health Sciences, Jinju 52727, South Korea
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, South Korea,School of Pharmacy, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Sunjoo Kim
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon, 51472, South Korea,Gyeongnam Center for Infectious Disease Control and Prevention, Changwon 51154, South Korea,Gyeongsang National University College of Medicine, Gyeongsang Institute of Health Sciences, Jinju 52727, South Korea,Corresponding author. Department of Laboratory Medicine, Gyeongsang University Changwon Hospital, Samjungja-ro 11, Seongsan-gu, Changwon-si, Gyeongnam 51472, South Korea. Fax: +82 55 214 3087
| |
Collapse
|
45
|
Mehta A, Andrew Awuah W, Yarlagadda R, Kalmanovich J, Huang H, Kundu M, Nansubuga EP, Lopes L, Ghosh B, Hasan MM. Investigating thyroid dysfunction in the context of COVID-19 infection. Ann Med Surg (Lond) 2022; 84:104806. [PMID: 36339111 PMCID: PMC9621589 DOI: 10.1016/j.amsu.2022.104806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/23/2022] [Indexed: 11/07/2022] Open
Abstract
COVID-19 is a contagious viral infection caused by severe acute respiratory syndrome coronavirus 2 (Sars-CoV-2). One of the key features of COVID-19 infection is inflammation. There is increasing evidence pointing to an association between cytokine storm and autoimmunity. One autoimmune disease of interest in connection to COVID-19 is hyperthyroidism. COVID-19 has been shown to decrease TSH levels and induce thyrotoxicosis, destructive thyroiditis, and de novo Graves' disease. It has also been suggested that the immune response against SARS-CoV-2 antigens following vaccination can cross-react through a mechanism called molecular mimicry which can elicit autoimmune reactivity, potentially leading to potential thyroid disease post vaccine. However, if the COVID-19 vaccine is linked to reduced COVID-19 related serious disease, it could potentially play a protective role against post COVID-19 hyperthyroidism (de novo disease and exacerbations). Further studies investigating the complex interplay between COVID-19 or COVID-19 vaccine and thyroid dysfunction can help provide substantial evidence and potential therapeutic targets that can alter prognosis and improve COVID-19 related outcomes in individuals with or without preexisting thyroid disease.
Collapse
Affiliation(s)
- Aashna Mehta
- University of Debrecen-Faculty of Medicine, Debrecen, 4032, Hungary
| | | | - Rohan Yarlagadda
- Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | | | - Helen Huang
- Royal College of Surgeons in Ireland, University of Medicine and Health Science, Dublin, Ireland
| | - Mrinmoy Kundu
- Institute of Medical Sciences and SUM Hospital, Bhubaneswar, India
| | | | - Leilani Lopes
- Western University of Health Sciences, College of Osteopathic Medicine of the Pacific-Northwest, Lebanon, OR, USA
| | | | - Mohammad Mehedi Hasan
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| |
Collapse
|
46
|
Satapathy P, Ratho RK, Sethi S. Immunopathogenesis in SARS-CoV-2 and Mycobacterium tuberculosis: The danger of overlapping crises. Front Pharmacol 2022; 13:1065124. [DOI: 10.3389/fphar.2022.1065124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
|
47
|
He J, Zhou L, Huang G, Shen J, Chen W, Wang C, Kim A, Zhang Z, Cheng W, Dai S, Ding F, Chen P. Enhanced Label-Free Nanoplasmonic Cytokine Detection in SARS-CoV-2 Induced Inflammation Using Rationally Designed Peptide Aptamer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:48464-48475. [PMID: 36281943 PMCID: PMC9627400 DOI: 10.1021/acsami.2c14748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/13/2022] [Indexed: 06/12/2023]
Abstract
Rapid and precise serum cytokine quantification provides immense clinical significance in monitoring the immune status of patients in rapidly evolving infectious/inflammatory disorders, examplified by the ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. However, real-time information on predictive cytokine biomarkers to guide targetable immune pathways in pathogenic inflammation is critically lacking, because of the insufficient detection range and detection limit in current label-free cytokine immunoassays. In this work, we report a highly sensitive localized surface plasmon resonance imaging (LSPRi) immunoassay for label-free Interleukin 6 (IL-6) detection utilizing rationally designed peptide aptamers as the capture interface. Benefiting from its characteristically smaller dimension and direct functionalization on the sensing surface via Au-S bonding, the peptide-aptamer-based LSPRi immunoassay achieved enhanced label-free serum IL-6 detection with a record-breaking limit of detection down to 4.6 pg/mL, and a wide dynamic range of ∼6 orders of magnitude (values from 4.6 to 1 × 106 pg/mL were observed). The immunoassay was validated in vitro for label-free analysis of SARS-CoV-2 induced inflammation, and further applied in rapid quantification of serum IL-6 profiles in COVID-19 patients. Our peptide aptamer LSPRi immunoassay demonstrates great potency in label-free cytokine detection with unprecedented sensing capability to provide accurate and timely interpretation of the inflammatory status and disease progression, and determination of prognosis.
Collapse
Affiliation(s)
- Jiacheng He
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Lang Zhou
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Gangtong Huang
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina29634, United States
| | - Jialiang Shen
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Wu Chen
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama36849, United States
| | - Chuanyu Wang
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Albert Kim
- Center for Medicine, Health, and Society, Vanderbilt University, Nashville, Tennessee37235, United States
| | - Zhuoyu Zhang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, New York11201, United States
- Department of Biomedical Engineering, New York University, Brooklyn, New York11201, United States
| | - Weiqiang Cheng
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, New York11201, United States
- Department of Biomedical Engineering, New York University, Brooklyn, New York11201, United States
| | - Siyuan Dai
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina29634, United States
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| |
Collapse
|
48
|
Abadi B, Aarabi Jeshvaghani AH, Fathalipour H, Dehghan L, Rahimi Sirjani K, Forootanfar H. Therapeutic Strategies in the Fight against COVID-19: From Bench to Bedside. IRANIAN JOURNAL OF MEDICAL SCIENCES 2022; 47:517-532. [PMID: 36380976 PMCID: PMC9652495 DOI: 10.30476/ijms.2021.92662.2396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/17/2021] [Accepted: 12/10/2021] [Indexed: 06/16/2023]
Abstract
In December 2019, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in China. This virus rapidly spread worldwide and was declared a global pandemic by the World Health Organization (WHO) in March 2020. High incidence, long incubation period, and diverse clinical signs of the disease posed a huge challenge globally. The efforts of health systems have been focused on repurposing existing drugs or developing innovative therapies to reduce the morbidity and mortality associated with SARS-CoV-2. In addition, most of the large pharmaceutical companies are intensely working on vaccine development to swiftly deliver safe and effective vaccines to prevent further spread of the virus. In this review, we will discuss the latest data on therapeutic strategies undergoing clinical trials. Additionally, we will provide a summary of vaccines currently under development.
Collapse
Affiliation(s)
- Banafshe Abadi
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Brain Cancer Research Core, Universal Scientific Education and Research Network, Tehran, Iran
| | | | - Hadis Fathalipour
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Leili Dehghan
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Hamid Forootanfar
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
49
|
Vitali L, Merlini A, Galvagno F, Proment A, Sangiolo D. Biological and Exploitable Crossroads for the Immune Response in Cancer and COVID-19. Biomedicines 2022; 10:2628. [PMID: 36289890 PMCID: PMC9599827 DOI: 10.3390/biomedicines10102628] [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: 07/28/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 12/15/2022] Open
Abstract
The outbreak of novel coronavirus disease 2019 (COVID-19) has exacted a disproportionate toll on cancer patients. The effects of anticancer treatments and cancer patients' characteristics shared significant responsibilities for this dismal outcome; however, the underlying immunopathological mechanisms are far from being completely understood. Indeed, despite their different etiologies, SARS-CoV-2 infection and cancer unexpectedly share relevant immunobiological connections. In the pathogenesis and natural history of both conditions, there emerges the centrality of the immune response, orchestrating the timed appearance, functional and dysfunctional roles of multiple effectors in acute and chronic phases. A significant number (more than 600) of observational and interventional studies have explored the interconnections between COVID-19 and cancer, focusing on aspects as diverse as psychological implications and prognostic factors, with more than 4000 manuscripts published so far. In this review, we reported and discussed the dynamic behavior of the main cytokines and immune system signaling pathways involved in acute vs. early, and chronic vs. advanced stages of SARS-CoV-2 infection and cancer. We highlighted the biological similarities and active connections within these dynamic disease scenarios, exploring and speculating on possible therapeutic crossroads from one setting to the other.
Collapse
Affiliation(s)
- Letizia Vitali
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Alessandra Merlini
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Federica Galvagno
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Alessia Proment
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Dario Sangiolo
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| |
Collapse
|
50
|
Yoga, Meditation, Breathing Exercises, and Inflammatory Biomarkers with Possible Implications in COVID-19: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3523432. [PMID: 36248417 PMCID: PMC9568285 DOI: 10.1155/2022/3523432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/01/2022] [Indexed: 01/08/2023]
Abstract
Introduction COVID-19, a multisystem disease, has implications for various immunity and infection biomarkers. Yoga (Y), meditation (M), and pranayama (P), and their combinations have shown positive changes on those biomarkers among other than COVID-19 patients and healthy people. So, we aimed to document the evidence of possible implication in a systematic way. Materials and Methods We screened 84 full texts, published in the last ten years, from three databases, from which only 44 met the eligibility criteria, and then extracted the data related to demographic characteristics, intervention, results, and strengths and limitations in two MS-Excel grids, and then presented them in tables and figures. Furthermore, we carried out meta-analysis including subgroup and sensitivity analysis using a random effects model of 11 RCTs and reported the mean difference, heterogeneity, and p value with 95% CI and presented them with forest and funnel plots and the tables. Results Twenty-five biomarkers of 4023 participants (range, 15-413) from 13 countries, healthy and clinical, from both sexes above 18 years, and from mainly clinical settings, were reported. YMP intervention, in solitary or in different possible combinations with varied durations among clinical and pregnant (range, 960-4800 minutes) and healthy (960-8400 minutes, excluding two studies of 20 minutes only) participants, was reported. It was revealed that 25 biomarkers, nine among the apparently healthy, 14 among the patients, and two among the pregnant, changed favourably (p < 0.05). Furthermore, either in meta- or subgroup-analysis, mean differences of IL-6 (-1.44 pg/ml) (95% CI) (-2.33, -0.55), (p = 0.002, I 2 = 82%), Cortisol (-40.75 pg/ml) (95% CI) (-64.13, -17.38), (p = 0.0006, I 2 = 87%), and TNF-α (-3.40 pg/ml) (95% CI) (-4.83, -1.98), (p < 0.0001, I 2 = 79%) showed statistically significant changes. Nonetheless, considerable heterogeneity and publication bias were observed among the studies. Conclusion Although more than two dozens of biomarkers in individual studies showed favourable changes, only IL-6, Cortisol, and TNF-α produced significant combined results, even then with much less certainty. Further meta-analysis of biomarkers of COVID-19 patients is highly recommended. Registration: CRD42021283894.
Collapse
|