1
|
Petry J, Shoykhet M, Weiser T, Griesbaum L, Bashiri Dezfouli A, Verschoor A, Wollenberg B. SARS-CoV-2 S1 protein induces IgG-mediated platelet activation and is prevented by 1.8-cineole. Biomed Pharmacother 2025; 187:118100. [PMID: 40306177 DOI: 10.1016/j.biopha.2025.118100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/12/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025] Open
Abstract
COVID-19 patients face an increased risk of thromboembolic complications, yet the exact pathophysiological role of platelets in the disease remains unclear. Considering the multifaceted nature of COVID-19 symptoms, including platelet hyperactivation and inflammation, the development of compounds that simultaneously target both represents a promising therapeutic strategy. The monoterpene 1.8-cineole (CNL-1976) is known for its anti-inflammatory and anti-aggregatory effects. Thus, understanding the mechanism behind platelet hyperactivation and the effect of 1.8-cineole during COVID-19 is crucial when aiming for a reduction of disease severity. In this study, we investigated the mechanism of platelet activation triggered by the SARS-CoV-2 S1 spike protein (S1). Utilizing S1-coupled beads, we discovered that platelet activation and aggregation were dependent on plasma components, particularly S1-specific IgG antibodies. The formation of immune complexes through IgG binding to S1 facilitated the crosslinking of the platelet expressed FcγRIIa receptor, initiating platelet activation and aggregation, as well as formation of platelet-leukocyte aggregates (PLAs). Importantly, treatment with 1.8-cineole significantly inhibited S1-bead-induced platelet activity and PLA formation. These findings strongly suggest that antibody-mediated platelet activation via FcγRIIa directly contributes to the well-recognized prothrombotic environment during COVID-19. Moreover, our data indicate that 1.8-cineole can serve as a potential therapeutic compound, alleviating platelet-driven thromboinflammatory complications associated with COVID-19 and post-acute sequelae of SARS-CoV-2 (PASC).
Collapse
Affiliation(s)
- Julie Petry
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany
| | - Maria Shoykhet
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany
| | - Tobias Weiser
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany
| | - Lena Griesbaum
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany
| | - Ali Bashiri Dezfouli
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany; Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Department of Radiation Oncology, TUM University Hospital, Germany
| | - Admar Verschoor
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany; University of Lübeck, Department of Dermatology, University Clinic Schleswig-Holstein (UKSH), Germany
| | - Barbara Wollenberg
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany.
| |
Collapse
|
2
|
Fiore NT, Willcox KF, Grieco AR, Dayani D, Zuberi YA, Heijnen CJ, Grace PM. Autoreactive IgG levels and Fc receptor γ subunit upregulation drive mechanical allodynia after nerve constriction or crush injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.22.644748. [PMID: 40196481 PMCID: PMC11974762 DOI: 10.1101/2025.03.22.644748] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
B cells contribute to the development of pain after sciatic nerve chronic constriction injury (CCI) via binding of immunoglobulin G (IgG) to Fc gamma receptors (FcγRs) in the lumbar dorsal root ganglia (DRG) and spinal cord. Yet the contribution of B cells to pain after different types of peripheral nerve injury is uncertain. Using male and female mice, we demonstrate a divergent role for B cell-IgG-FcγR signaling underlying mechanical allodynia between CCI, nerve crush (NC), spared nerve injury (SNI), and spinal nerve ligation (SNL). Depletion (monoclonal anti-CD20) or genetic deletion (muMT mice) of B cells prevented development of allodynia following NC and CCI, but not SNI or SNL. In apparent contradiction, circulating levels of autoreactive IgG and circulating immune complexes were increased in all models, though more prominent following NC and CCI. Passive transfer of IgG from SNI donor mice induced allodynia in CCI muMT recipient mice, demonstrating that IgG secreted after SNI is pronociceptive. To investigate why pronociceptive IgG did not contribute to mechanical allodynia after SNI, we evaluated levels of the Fc receptor γ subunit. SNI or SNL did not increase γ subunit levels in the DRG and spinal cord, whereas CCI and NC did, in agreement with B cell-dependent allodynia in these models. Together, the results suggest that traumatic peripheral nerve injury drives secretion of autoreactive IgG from B cells. However, levels of cognate FcγRs are increased following sciatic nerve constriction and crush, but not transection, to differentially regulate pain through the B cell-IgG-FcγR axis.
Collapse
Affiliation(s)
- Nathan T. Fiore
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center; Houston, USA
| | - Kendal F. Willcox
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center; Houston, USA
| | - Anamaria R. Grieco
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center; Houston, USA
| | - Dorsa Dayani
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center; Houston, USA
| | - Younus A. Zuberi
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center; Houston, USA
| | - Cobi J. Heijnen
- Department of Psychological Sciences, Rice University; Houston, USA
| | - Peter M. Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center; Houston, USA
| |
Collapse
|
3
|
Low ZXB, Yong SJ, Alrasheed HA, Al-Subaie MF, Al Kaabi NA, Alfaresi M, Albayat H, Alotaibi J, Al Bshabshe A, Alwashmi ASS, Sabour AA, Alshiekheid MA, Almansour ZH, Alharthi H, Al Ali HA, Almoumen AA, Alqasimi NA, AlSaihati H, Rodriguez-Morales AJ, Rabaan AA. Serotonergic psychedelics as potential therapeutics for post-COVID-19 syndrome (or Long COVID): A comprehensive review. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111279. [PMID: 39909170 DOI: 10.1016/j.pnpbp.2025.111279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
RATIONALE In our ongoing battle against the coronavirus 2019 (COVID-19) pandemic, a major challenge is the enduring symptoms that continue after acute infection. Also known as Long COVID, post-COVID-19 syndrome (PCS) often comes with debilitating symptoms like fatigue, disordered sleep, olfactory dysfunction, and cognitive issues ("brain fog"). Currently, there are no approved treatments for PCS. Recent research has uncovered that the severity of PCS is inversely linked to circulating serotonin levels, highlighting the potential of serotonin-modulating therapeutics for PCS. Therefore, we propose that serotonergic psychedelics, acting mainly via the 5-HT2A serotonin receptor, hold promise for treating PCS. OBJECTIVES Our review aims to elucidate potential mechanisms by which serotonergic psychedelics may alleviate the symptoms of PCS. RESULTS Potential mechanisms through which serotonergic psychedelics may alleviate PCS symptoms are discussed, with emphasis on their effects on inflammation, neuroplasticity, and gastrointestinal function. Additionally, this review explores the potential of serotonergic psychedelics in mitigating endothelial dysfunction, a pivotal aspect of PCS pathophysiology implicated in organ dysfunction. This review also examines the potential role of serotonergic psychedelics in alleviating specific PCS symptoms, which include olfactory dysfunction, cognitive impairment, sleep disturbances, and mental health challenges. CONCLUSIONS Emerging evidence suggests that serotonergic psychedelics may alleviate PCS symptoms. However, further high-quality research is needed to thoroughly assess their safety and efficacy in treating patients with PCS.
Collapse
Affiliation(s)
- Zhen Xuen Brandon Low
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Shin Jie Yong
- School of Medical and Life Sciences, Sunway University, Selangor, Malaysia.
| | - Hayam A Alrasheed
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Maha F Al-Subaie
- Research Center, Dr. Sulaiman Alhabib Medical Group, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Nawal A Al Kaabi
- College of Medicine and Health Science, Khalifa University, Abu Dhabi, United Arab Emirates; Sheikh Khalifa Medical City, Abu Dhabi Health Services Company, Abu Dhabi, United Arab Emirates
| | - Mubarak Alfaresi
- Department of Microbiology, National Reference Laboratory, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates; Department of Pathology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Hawra Albayat
- Infectious Disease Department, King Saud Medical City, Riyadh, Saudi Arabia
| | - Jawaher Alotaibi
- Infectious Diseases Unit, Department of Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ali Al Bshabshe
- Adult Critical Care Department of Medicine, Division of Adult Critical Care, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ameen S S Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Amal A Sabour
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Maha A Alshiekheid
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Zainab H Almansour
- Biological Science Department, College of Science, King Faisal University, Hofuf, Saudi Arabia
| | - Huda Alharthi
- Clinical Pharmacist, Pharmaceutical Care Department, King Faisal Medical Complex, Taif Health Cluster, Ministry of Health, Taif, Saudi Arabia
| | - Hani A Al Ali
- Pediatrics Department, Maternity & Children Hospital, Dammam, Saudi Arabia
| | - Adel A Almoumen
- Pediatrics Department, Maternity & Children Hospital, Dammam, Saudi Arabia
| | - Nabil A Alqasimi
- Pediatrics Department, Maternity & Children Hospital, Dammam, Saudi Arabia
| | - Hajir AlSaihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Alfonso J Rodriguez-Morales
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima, Peru; Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Ali A Rabaan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan.
| |
Collapse
|
4
|
Wu Y, Liu J, Xiao Y, Zhang S, Li L. CoupleVAE: coupled variational autoencoders for predicting perturbational single-cell RNA sequencing data. Brief Bioinform 2025; 26:bbaf126. [PMID: 40178283 PMCID: PMC11966612 DOI: 10.1093/bib/bbaf126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/21/2025] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
With the rapid advances in single-cell sequencing technology, it is now feasible to conduct in-depth genetic analysis in individual cells. Study on the dynamics of single cells in response to perturbations is of great significance for understanding the functions and behaviors of living organisms. However, the acquisition of post-perturbation cellular states via biological experiments is frequently cost-prohibitive. Predicting the single-cell perturbation responses poses a critical challenge in the field of computational biology. In this work, we propose a novel deep learning method called coupled variational autoencoders (CoupleVAE), devised to predict the postperturbation single-cell RNA-Seq data. CoupleVAE is composed of two coupled VAEs connected by a coupler, initially extracting latent features for controlled and perturbed cells via two encoders, subsequently engaging in mutual translation within the latent space through two nonlinear mappings via a coupler, and ultimately generating controlled and perturbed data by two separate decoders to process the encoded and translated features. CoupleVAE facilitates a more intricate state transformation of single cells within the latent space. Experiments in three real datasets on infection, stimulation and cross-species prediction show that CoupleVAE surpasses the existing comparative models in effectively predicting single-cell RNA-seq data for perturbed cells, achieving superior accuracy.
Collapse
Affiliation(s)
- Yahao Wu
- School of Mathematics and Statistics, Xi’an Jiaotong University, No. 28 Xianning West Road, Xi’an, Shaanxi 710049, China
| | - Jing Liu
- School of Mathematics and Statistics, Xi’an Jiaotong University, No. 28 Xianning West Road, Xi’an, Shaanxi 710049, China
| | - Yanni Xiao
- School of Mathematics and Statistics, Xi’an Jiaotong University, No. 28 Xianning West Road, Xi’an, Shaanxi 710049, China
| | - Shuqin Zhang
- School of Mathematical Sciences, Center for Applied Mathematics, Research Institute of Intelligent Complex Systems, and Shanghai Key Laboratory for Contemporary Applied Mathematics, Fudan University, 220 Handan Road, 200433 Shanghai, China
| | - Limin Li
- School of Mathematics and Statistics, Xi’an Jiaotong University, No. 28 Xianning West Road, Xi’an, Shaanxi 710049, China
| |
Collapse
|
5
|
Sun DS, Lien TS, Chang HH. Virus-Induced Pathogenic Antibodies: Lessons from Long COVID and Dengue Hemorrhage Fever. Int J Mol Sci 2025; 26:1898. [PMID: 40076527 PMCID: PMC11899886 DOI: 10.3390/ijms26051898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Virus-induced antibodies represent a dual-edged sword in the immune response to viral infections. While antibodies are critical for neutralizing pathogens, some can paradoxically exacerbate disease severity through mechanisms such as antibody-dependent enhancement (ADE), autoantibody, and prolonged inflammation. Long coronavirus disease (COVID) and dengue hemorrhagic fever (DHF) exemplify conditions where pathogenic antibodies play a pivotal role in disease progression. Long COVID is associated with persistent immune dysregulation and autoantibody production, leading to chronic symptoms and tissue damage. In DHF, pre-existing antibodies against dengue virus contribute to ADE, amplifying viral replication, immune activation, and vascular permeability. This review explores the mechanisms underlying these pathogenic antibody responses, highlighting the shared pathways of immune dysregulation and comparing the distinct features of both conditions. By examining these studies, we identify key lessons for therapeutic strategies, vaccine design, and future research aimed at mitigating the severe outcomes of viral infections.
Collapse
Grants
- 104-2320-B-320 -009 -MY3, 107-2311-B-320-002-MY3, 111-2320-B320-006-MY3, 112-2320-B-320-007 National Science and Technology Council, Taiwan
- TCMMP104-06, TCMMP108-04, TCMMP 111-01, TCAS111-02, TCAS-112-02, TCAS113-04, TCRD112-033, TCRD113-041 Tzu-Chi Medical Foundation
Collapse
Affiliation(s)
| | | | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (D.-S.S.); (T.-S.L.)
| |
Collapse
|
6
|
Moriishi M, Takazono T, Hashizume J, Aibara N, Kutsuna YJ, Okamoto M, Sawai T, Hoshino T, Mori Y, Fukuda Y, Awaya Y, Yamanashi H, Furusato Y, Yanagihara T, Miyamoto H, Sato K, Kodama Y, Mizukami S, Sakamoto N, Yamamoto K, Sakamoto K, Yanagihara K, Izumikawa K, Maeda T, Nakashima M, Fukushima K, Mukae H, Ohyama K. Immune complexome analysis reveals an autoimmune signature predictive of COVID-19 severity. Clin Biochem 2025; 135:110865. [PMID: 39689808 DOI: 10.1016/j.clinbiochem.2024.110865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/24/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND The factors contributing to the development of severe coronavirus disease 2019 (COVID-19) following infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain unclear. Although the presence of immune complexes (ICs), formed between antibodies and their antigens, has been linked to COVID-19 severity, their role requires further investigation, and the antigens within these ICs are yet to be characterized. METHOD Here, a C1q enzyme-liked immunosorbent assay and immune complexome analysis were used to determine IC concentrations and characterize IC antigens, respectively, in the sera of 64 unvaccinated COVID-19 patients with PCR-confirmed SARS-CoV-2 infection, enrolled at seven participating centers in 2020. For the analysis, the patients were split into the severe (n = 35) and non-severe (n = 28) groups on the basis of their COVID-19 symptoms. RESULTS We found that neither serum IC concentration nor IC antigen number was associated with COVID-19 severity. However, we identified six IC antigens, which were significantly enriched in the severe versus non-severe group. These IC antigens were all derived from human proteins, namely haptoglobin, the serum amyloid A-2 protein, the serum amyloid A-1 protein, clusterin, and lipopolysaccharide-binding protein, and complement-factor-H-related protein 3. Meanwhile, we found no association between COVID-19 severity and IC antigens derived from SARS-CoV-2 proteins. Collectively, the six IC antigens predicted COVID-19 severity with a moderate degree of accuracy (area under the receiver operating characteristic curve = 0.90, sensitivity = 94 %, specificity = 79 %). CONCLUSIONS The IC antigen signature identified in this study may have important implications for the diagnosis and treatment of severe COVID-19.
Collapse
Affiliation(s)
- Marino Moriishi
- Department of Pharmacy Practice, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takahiro Takazono
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Junya Hashizume
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Nozomi Aibara
- Department of Pharmacy Practice, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuki Jimbayashi Kutsuna
- Department of Molecular Pathochemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Masaki Okamoto
- Department of Respirology, NHO Kyushu Medical Center, Fukuoka, Japan; Division of Respiratory, Neurology and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toyomitsu Sawai
- Department of Respiratory Medicine, Nagasaki Harbor Medical Center, Nagasaki, Japan
| | - Teppei Hoshino
- Department of Internal Medicine, Kitakyushu Municipal Yahata Hospital, Kitakyushu, Fukuoka, Japan
| | - Yusuke Mori
- Department of Internal Medicine, Kitakyushu Municipal Yahata Hospital, Kitakyushu, Fukuoka, Japan
| | - Yuichi Fukuda
- Department of Respiratory Medicine, Sasebo City General Hospital, Sasebo, Japan
| | - Yukikazu Awaya
- Division of Respiratory Medicine, Itabashi Chuo Medical Center, Itabashi-ku, Tokyo, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | | | - Toyoshi Yanagihara
- Department of Respiratory Medicine, NHO Fukuoka National Hospital, Fukuoka, Japan
| | - Hirotaka Miyamoto
- Department of Pharmaceutics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kayoko Sato
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Yukinobu Kodama
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan; Department of Molecular Pathochemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shusaku Mizukami
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Noriho Sakamoto
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuko Yamamoto
- First Department of Internal Medicine, Division of Infectious, Respiratory, and Digestive Medicine, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Kei Sakamoto
- Department of Microbiology and Immunology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Katsunori Yanagihara
- Division of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Koichi Izumikawa
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Infection Control and Education Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Takahiro Maeda
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mikiro Nakashima
- Department of Pharmacy Practice, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kiyoyasu Fukushima
- Department of Respiratory Medicine, Japanese Red Cross Nagasaki Genbaku Isahaya Hospital, Isahaya, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kaname Ohyama
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan; Department of Molecular Pathochemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| |
Collapse
|
7
|
Yaugel-Novoa M, Noailly B, Jospin F, Pizzorno A, Traversier A, Pozzetto B, Waeckel L, Longet S, Pillet S, Botelho-Nevers E, Rosa-Calatrava M, Bourlet T, Paul S. Impaired mucosal IgA response in patients with severe COVID-19. Emerg Microbes Infect 2024; 13:2401940. [PMID: 39358866 PMCID: PMC11451292 DOI: 10.1080/22221751.2024.2401940] [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: 06/12/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Several studies have investigated the antibody response to SARS-CoV-2, focusing particularly on the systemic humoral immune response and the production of immunoglobulin G (IgG) antibodies. IgA antibodies play a crucial role in protecting against respiratory viral infections but have also been associated with the pathophysiology of COVID-19. We performed a prospective study of 169 COVID-19 patients - 50 with critical/severe (ICU), 47 with moderate (Non-ICU), and 72 with asymptomatic COVID-19 - to explore the humoral immune response to SARS-CoV-2 infection. We found that the early systemic IgA response strongly induced in patients with severe disease did not block IgG neutralization functions and activated FcRs more effectively than IgG. However, even if SIgA levels were high, mucosal IgA antibodies could not control the infection effectively in patients with severe disease. Our findings highlight the complexity of the immune response to SARS-CoV-2 exhibiting high systemic levels of IgA with strong neutralizing capacity in severe cases, together with higher levels of IgA-FcR activation than in asymptomatic patients. They also suggest the need for further research to fully understand the role of IgA and its structural alterations in mucosal tissues in cases of severe disease and the impact of these antibodies on disease progression.
Collapse
Affiliation(s)
- Melyssa Yaugel-Novoa
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Blandine Noailly
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Fabienne Jospin
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Andrés Pizzorno
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Aurélien Traversier
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Bruno Pozzetto
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Louis Waeckel
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphanie Longet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Sylvie Pillet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Diseases Department, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Manuel Rosa-Calatrava
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Thomas Bourlet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Lead contact
| |
Collapse
|
8
|
Andreeva I, Kolb P, Rodon L, Blank N, Lorenz HM, Merkt W. Fcγ-receptor-IIIA bioactivity of circulating and synovial immune complexes in rheumatoid arthritis. RMD Open 2024; 10:e004190. [PMID: 39209371 PMCID: PMC11367361 DOI: 10.1136/rmdopen-2024-004190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Previous technical limitations prevented the proof of Fcγ-receptor (FcγR)-activation by soluble immune complexes (sICs) in patients. FcγRIIIa (CD16) is a risk factor in rheumatoid arthritis (RA). We aimed at determining the presence of CD16-activating sICs in RA and control diseases. METHODS Sera from an exploratory cohort (n=50 patients with RA) and a validation cohort (n=106 patients with RA, 20 patients with psoriasis arthritis (PsA), 22 patients with systemic lupus erythematosus (SLE) and 31 healthy controls) were analysed using a new reporter cell assay. Additionally, 26 synovial fluid samples were analysed, including paired serum/synovial samples. RESULTS For the first time using a reliable and sensitive functional assay, the presence of sICs in RA sera was confirmed. sICs possess an intrinsic capacity to activate CD16 and can be found in both synovial fluid and in blood. In low experimental dilutions, circulating sICs were also detected in a subset of healthy people and in PsA. However, we report a significantly increased frequency of bioactive circulating sICs in RA. While the bioactivity of circulating sICs was low and did not correlate with clinical parameters, synovial sICs were highly bioactive and correlated with serum autoantibody levels. Receiver operator curves indicated that sICs bioactivity in synovial fluid could be used to discriminate immune complex-associated arthritis from non-associated forms. Finally, circulating sICs were more frequently found in SLE than in RA. The degree of CD16 bioactivity showed strong donor-dependent differences, especially in SLE. CONCLUSIONS RA is characterised by the presence of circulating and synovial sICs that can engage and activate CD16.
Collapse
Affiliation(s)
- Ivana Andreeva
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp Kolb
- Institute of Virology, Medical Center—University of Freiburg, Freiburg, Germany
- Albert-Ludwigs-Universitat Freiburg Medizinische Fakultat, Freiburg, Germany
| | - Lea Rodon
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Norbert Blank
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Merkt
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
- Hiller Forschungszentrum, Department of Rheumatology, University Hospital of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
9
|
Baig AM, Rosko S, Jaeger B, Gerlach J, Rausch H. Unraveling the enigma of long COVID: novel aspects in pathogenesis, diagnosis, and treatment protocols. Inflammopharmacology 2024; 32:2075-2090. [PMID: 38771409 DOI: 10.1007/s10787-024-01483-2] [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: 02/27/2024] [Accepted: 04/17/2024] [Indexed: 05/22/2024]
Abstract
Long COVID, now unmistakably identified as a syndromic entity encompassing a complex spectrum of symptoms, demands immediate resolution of its elusive pathogenic underpinnings. The intricate interplay of diverse factors presents a complex puzzle, difficult to resolve, and thus poses a substantial challenge. As instances of long COVID manifest by repeated infections of SARS-CoV-2 and genetic predisposition, a detailed understanding in this regard is needed. This endeavor is a comprehensive exploration and analysis of the cascading pathogenetic events driven by viral persistence and replication. Beyond its morbidity, long COVID, more disabling than fatal, exacts one of the most substantial tolls on public health in contemporary times, with the potential to cripple national economies. The paper introduces a unified theory of long COVID, detailing a novel pathophysiological framework that interlinks persistent SARS-CoV-2 infection, autoimmunity, and systemic vascular pathology. We posit a model where viral reservoirs, immune dysregulation, and genetic predispositions converge to perpetuate disease. It challenges prevailing hypotheses with new evidence, suggesting innovative diagnostic and therapeutic approaches. The paper aims to shift the paradigm in long COVID research by providing an integrative perspective that encapsulates the multifaceted nature of the condition. We explain the immunological mechanisms, hypercoagulability states, and viral reservoirs in the skull that feed NeuroCOVID in patients with long COVID. Also, this study hints toward a patient approach and how to prioritize treatment sequences in long COVID patients in hospitals and clinics.
Collapse
Affiliation(s)
| | - Sandy Rosko
- Clinicum St. George, Rosenheimer Str. 6-8, Bad Aibling, Germany
| | - Beate Jaeger
- Clinicum St. George, Rosenheimer Str. 6-8, Bad Aibling, Germany
| | - Joachim Gerlach
- Clinicum St. George, Rosenheimer Str. 6-8, Bad Aibling, Germany
| | - Hans Rausch
- Clinicum St. George, Rosenheimer Str. 6-8, Bad Aibling, Germany
| |
Collapse
|
10
|
Severa M, Etna MP, Andreano E, Ricci D, Cairo G, Fiore S, Canitano A, Cara A, Stefanelli P, Rappuoli R, Palamara AT, Coccia EM. Functional diversification of innate and inflammatory immune responses mediated by antibody fragment crystallizable activities against SARS-CoV-2. iScience 2024; 27:109703. [PMID: 38706870 PMCID: PMC11068556 DOI: 10.1016/j.isci.2024.109703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/25/2024] [Accepted: 04/06/2024] [Indexed: 05/07/2024] Open
Abstract
Monoclonal antibodies (mAb) targeting the SARS-CoV-2 Spike (S) glycoprotein have been exploited for the treatment of severe COVID-19. In this study, we evaluated the immune-regulatory features of two neutralizing anti-S mAbs (nAbs), named J08 and F05, with wild-type (WT) conformation or silenced Fc functions. In the presence of D614G SARS-CoV-2, WT nAbs enhance intracellular viral uptake in immune cells and amplify antiviral type I Interferon and inflammatory cytokine and chemokine production without viral replication, promoting the differentiation of CD16+ inflammatory monocytes and innate/adaptive PD-L1+ and PD-L1+CD80+ plasmacytoid Dendritic Cells. In spite of a reduced neutralizing property, WT J08 nAb still promotes the IL-6 production and differentiation of CD16+ monocytes once binding Omicron BA.1 variant. Fc-mediated regulation of antiviral and inflammatory responses, in the absence of viral replication, highlighted in this study, might positively tune immune response during SARS-CoV-2 infection and be exploited also in mAb-based therapeutic and prophylactic strategies against viral infections.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marilena Paola Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Emanuele Andreano
- Monoclonal Antibody Discovery Lab, Fondazione Toscana Life Sciences, 53100 Siena, Italy
| | - Daniela Ricci
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
- Department of Sciences, Roma Tre University, 00154 Rome, Italy
| | - Giada Cairo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Stefano Fiore
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Rino Rappuoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Fondazione Biotecnopolo di Siena, 53100 Siena, Italy
| | - Anna Teresa Palamara
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Eliana Marina Coccia
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
11
|
Lučiūnaitė A, Mašalaitė K, Plikusiene I, Maciulis V, Juciute S, Norkienė M, Žvirblienė A. Structural properties of immune complexes formed by viral antigens and specific antibodies shape the inflammatory response of macrophages. Cell Biosci 2024; 14:53. [PMID: 38664730 PMCID: PMC11046781 DOI: 10.1186/s13578-024-01237-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/20/2024] [Indexed: 04/28/2024] Open
Abstract
Data on the course of viral infections revealed severe inflammation as a consequence of antiviral immune response. Despite extensive research, there are insufficient data on the role of innate immune cells in promoting inflammation mediated by immune complexes (IC) of viral antigens and their specific antibodies. Recently, we demonstrated that antigens of human polyomaviruses (PyVs) induce an inflammatory response in macrophages. Here, we investigated macrophage activation by IC. We used primary murine macrophages as a cell model, virus-like particles (VLPs) of PyV capsid protein as antigens, and a collection of murine monoclonal antibodies (mAbs) of IgG1, IgG2a, IgG2b subclasses. The inflammatory response was investigated by analysing inflammatory chemokines and activation of NLRP3 inflammasome. We observed a diverse pattern of chemokine secretion in macrophages treated with different IC compared to VLPs alone. To link IC properties with cell activation status, we characterised the IC by advanced optical and acoustic techniques. Ellipsometry provided precise real-time kinetics of mAb-antigen interactions, while quartz crystal microbalance measurements showed changes in conformation and viscoelastic properties during IC formation. These results revealed differences in mAb-antigen interaction and mAb binding parameters of the investigated IC. We found that IC-mediated cell activation depends more on IC characteristics, including mAb affinity, than on mAb affinity for the activating Fc receptor. IC formed by the highest affinity mAb showed a significant enhancement of inflammasome activation. This may explain the hyperinflammation related to viral infection and vaccination. Our findings demonstrate that IC promote the viral antigen-induced inflammatory response depending on antibody properties.
Collapse
Affiliation(s)
- Asta Lučiūnaitė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, 10257, Vilnius, Lithuania.
| | - Kristina Mašalaitė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, 10257, Vilnius, Lithuania
| | - Ieva Plikusiene
- State Research Institute Center for Physical Sciences and Technology, Vilnius, Lithuania
- Pharmacy and Pharmacology Center, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Vincentas Maciulis
- State Research Institute Center for Physical Sciences and Technology, Vilnius, Lithuania
| | - Silvija Juciute
- NanoTechnas - Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Vilnius, Lithuania
| | - Milda Norkienė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, 10257, Vilnius, Lithuania
| | - Aurelija Žvirblienė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, 10257, Vilnius, Lithuania
| |
Collapse
|
12
|
Hsieh KH, Chao CH, Cheng YL, Lai YC, Chuang YC, Wang JR, Chang SY, Hung YP, Chen YMA, Liu WL, Chuang WJ, Yeh TM. Enhancement of NETosis by ACE2-cross-reactive anti-SARS-CoV-2 RBD antibodies in patients with COVID-19. J Biomed Sci 2024; 31:39. [PMID: 38637878 PMCID: PMC11027296 DOI: 10.1186/s12929-024-01026-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/26/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND High levels of neutrophil extracellular trap (NET) formation or NETosis and autoantibodies are related to poor prognosis and disease severity of COVID-19 patients. Human angiotensin-converting enzyme 2 (ACE2) cross-reactive anti-severe acute respiratory syndrome coronavirus 2 spike protein receptor-binding domain (SARS-CoV-2 RBD) antibodies (CR Abs) have been reported as one of the sources of anti-ACE2 autoantibodies. However, the pathological implications of CR Abs in NET formation remain unknown. METHODS In this study, we first assessed the presence of CR Abs in the sera of COVID-19 patients with different severity by serological analysis. Sera and purified IgG from CR Abs positive COVID-19 patients as well as a mouse monoclonal Ab (mAb 127) that can recognize both ACE2 and the RBD were tested for their influence on NETosis and the possible mechanisms involved were studied. RESULTS An association between CR Abs levels and the severity of COVID-19 in 120 patients was found. The CR Abs-positive sera and IgG from severe COVID-19 patients and mAb 127 significantly activated human leukocytes and triggered NETosis, in the presence of RBD. This NETosis, triggered by the coexistence of CR Abs and RBD, activated thrombus-related cells but was abolished when the interaction between CR Abs and ACE2 or Fc receptors was disrupted. We also revealed that CR Abs-induced NETosis was suppressed in the presence of recombinant ACE2 or the Src family kinase inhibitor, dasatinib. Furthermore, we found that COVID-19 vaccination not only reduced COVID-19 severity but also prevented the production of CR Abs after SARS-CoV-2 infection. CONCLUSIONS Our findings provide possible pathogenic effects of CR Abs in exacerbating COVID-19 by enhancing NETosis, highlighting ACE2 and dasatinib as potential treatments, and supporting the benefit of vaccination in reducing disease severity and CR Abs production in COVID-19 patients.
Collapse
Affiliation(s)
- Kun-Han Hsieh
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiao-Hsuan Chao
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory and Regenerative Medicine, MacKay Medical College, New Taipei, Taiwan
| | - Yi-Ling Cheng
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Chung Lai
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Yung-Chun Chuang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Leadgene Biomedical, Inc, Tainan, Taiwan
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Jen-Ren Wang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Diseases and Vaccinology, National Institute of Infectious National Health Research Institutes, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yuan-Pin Hung
- Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University, Medical College and Hospital, Tainan, Taiwan
| | - Yi-Ming Arthur Chen
- Laboratory of Important Infectious Diseases and Cancer, Department of Medicine, School of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan
- Diseases and Vaccinology, National Institute of Infectious National Health Research Institutes, Miaoli County, 350, Taiwan
| | - Wei-Lun Liu
- School of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan
- Department of Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, 243, Taiwan
- Data Science Center, College of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan
| | - Woei-Jer Chuang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Trai-Ming Yeh
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
13
|
Du Q, Yu J, Chen Q, Chen X, Jiang Q, Deng L, Li A, Xiong Y. Clinical characteristics and influencing factors of severe fever with thrombocytopenia syndrome complicated by viral myocarditis: a retrospective study. BMC Infect Dis 2024; 24:240. [PMID: 38389047 PMCID: PMC10885462 DOI: 10.1186/s12879-024-09096-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the clinical characteristics of severe fever with thrombocytopenia syndrome complicated by viral myocarditis (SFTS-VM) and analyze relevant influencing factors. METHODS Retrospective analysis was conducted on clinical data from 79 SFTS-VM patients, categorized into common (SFTS-CVM, n = 40) and severe groups (SFTS-SVM, n = 39). Clinical manifestations, laboratory results, cardiac ultrasonography, and electrocardiogram features were analyzed. Univariate and multivariate analyses identified significant indicators, which were further assessed using ROC curves to predict SFTS-SVM. RESULTS SFTS-SVM group exhibited higher rates of hypotension, shock, abdominal pain, cough with sputum, and consciousness disorders compared to SFTS-CVM group. Laboratory findings showed elevated platelet count, ALT, AST, amylase, lipase, LDH, D-dimer, procalcitonin, TNI, and NT-proBNP in SFTS-SVM. Abnormal electrocardiograms, especially atrial fibrillation, were more prevalent in SFTS-SVM (P < 0.05). Multivariate analysis identified elevated LDH upon admission (OR = 1.004, 95% CI: 1-1.008, P = 0.050), elevated NT-proBNP (OR = 1.005, 95% CI: 1.001-1.008, P = 0.007), and consciousness disorders (OR = 112.852, 95% CI: 3.676 ~ 3464.292, P = 0.007) as independent risk factors for SFTS-SVM. LDH and NT-proBNP had AUCs of 0.728 and 0.744, respectively, in predicting SFTS-SVM. Critical values of LDH (> 978.5U/L) and NT-proBNP (> 857.5pg/ml)) indicated increased likelihood of SFTS progression into SVM. CONCLUSION Elevated LDH, NT-proBNP, and consciousness disorders independently correlate with SFTS-SVM. LDH and NT-proBNP can aid in early identification of SFTS-SVM development when above specified thresholds.
Collapse
Affiliation(s)
- Qian Du
- Department of Infectious Disease, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Jin Yu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Qianhui Chen
- Department of Infectious Disease, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Xiaoping Chen
- Department of Infectious Disease, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Qunqun Jiang
- Department of Infectious Disease, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Liping Deng
- Department of Infectious Disease, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China.
| | - Anling Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China.
| | - Yong Xiong
- Department of Infectious Disease, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China.
| |
Collapse
|
14
|
Geyer CE, Chen HJ, Bye AP, Manz XD, Guerra D, Caniels TG, Bijl TP, Griffith GR, Hoepel W, de Taeye SW, Veth J, Vlaar AP, Vidarsson G, Bogaard HJ, Aman J, Gibbins JM, van Gils MJ, de Winther MP, den Dunnen J. Identification of new drugs to counteract anti-spike IgG-induced hyperinflammation in severe COVID-19. Life Sci Alliance 2023; 6:e202302106. [PMID: 37699657 PMCID: PMC10497933 DOI: 10.26508/lsa.202302106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Previously, we and others have shown that SARS-CoV-2 spike-specific IgG antibodies play a major role in disease severity in COVID-19 by triggering macrophage hyperactivation, disrupting endothelial barrier integrity, and inducing thrombus formation. This hyperinflammation is dependent on high levels of anti-spike IgG with aberrant Fc tail glycosylation, leading to Fcγ receptor hyperactivation. For development of immune-regulatory therapeutics, drug specificity is crucial to counteract excessive inflammation whereas simultaneously minimizing the inhibition of antiviral immunity. We here developed an in vitro activation assay to screen for small molecule drugs that specifically counteract antibody-induced pathology. We identified that anti-spike-induced inflammation is specifically blocked by small molecule inhibitors against SYK and PI3K. We identified SYK inhibitor entospletinib as the most promising candidate drug, which also counteracted anti-spike-induced endothelial dysfunction and thrombus formation. Moreover, entospletinib blocked inflammation by different SARS-CoV-2 variants of concern. Combined, these data identify entospletinib as a promising treatment for severe COVID-19.
Collapse
Affiliation(s)
- Chiara E Geyer
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Hung-Jen Chen
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Alexander P Bye
- Institute for Cardiovascular and Metabolic Research, and School of Biological Sciences, University of Reading, Reading, UK
- Molecular and Clinical Sciences Research Institute, St George's University, London, UK
- School of Pharmacy, University of Reading, Reading, UK
| | - Xue D Manz
- Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Denise Guerra
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Tom G Caniels
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Tom Pl Bijl
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Guillermo R Griffith
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Willianne Hoepel
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Steven W de Taeye
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jennifer Veth
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Alexander Pj Vlaar
- Department of Intensive Care Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Gestur Vidarsson
- Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Harm Jan Bogaard
- Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jurjan Aman
- Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, and School of Biological Sciences, University of Reading, Reading, UK
| | - Marit J van Gils
- Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Menno Pj de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jeroen den Dunnen
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
15
|
Fiorelli D, Caruso V, Belardi R, Bernardini S, Nuccetelli M. Evaluation of autoantibody profile in healthy subjects after mRNA vaccination against COVID-19. Int Immunopharmacol 2023; 122:110592. [PMID: 37413933 DOI: 10.1016/j.intimp.2023.110592] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/16/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND SARS-CoV-2 severe acute respiratory syndrome has rapidly spread worldwide since 2019. All scientific and technological forces have concentrated towards the formulation of vaccines to contain the disease. In less than one year (December 2020) a first messenger RNA vaccine (Comirnaty, BioNTech/Pfizer) was authorized. However, the research community has wondered about possible side effects on the immune system, given the vaccines administration in phase 4. AIM This study aims to evaluate the mRNA vaccine impact on the development of possible positive autoantibody profile in healthcare workers without any previous underlying pathology, after first, second and booster dose of Pfizer vaccine, by determining: circulating immune complexes concentrations (CIC); anti-myeloperoxidase (MPO) and anti-proteinase 3 (PR3) autoantibodies, the presence of antinuclear antibodies (ANA) and subsequent second level tests (extractable nuclear antigen (ENA) screen, double-strand DNA, extractable nuclear antigen (ANA) profile). METHODS The subjects were divided according to anti-SARS-CoV-2 IgG RBD antibodies increasing concentrations in: Group I < 10 BAU/ml (N = 114); Group II > 1000 BAU/ml (N = 112); Group III > 2500 BAU/ml (N = 78). RESULTS Our data show no autoreactive response changes over time in healthy subjects after vaccination. In fact, evaluation of ANA, CIC, anti-MPO, anti-PR3 and the detection of specific autoantigens, did not display significant variations. CONCLUSIONS The results suggest the exclusion of a correlation between the administration of the vaccine and the possible onset of autoimmune disorders. Nevertheless, further investigations will be needed to test for any long-term side effects on an ever-growing population.
Collapse
Affiliation(s)
- Denise Fiorelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy.
| | - Vincenza Caruso
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Riccardo Belardi
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| |
Collapse
|
16
|
Skrzydlewska E, Łuczaj W, Biernacki M, Wójcik P, Jarocka-Karpowicz I, Orehovec B, Baršić B, Tarle M, Kmet M, Lukšić I, Marušić Z, Bauer G, Žarković N. Preliminary Comparison of Molecular Antioxidant and Inflammatory Mechanisms Determined in the Peripheral Blood Granulocytes of COVID-19 Patients. Int J Mol Sci 2023; 24:13574. [PMID: 37686388 PMCID: PMC10488240 DOI: 10.3390/ijms241713574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
The aim of this study was to evaluate selected parameters of redox signaling and inflammation in the granulocytes of COVID-19 patients who recovered and those who died. Upon admission, the patients did not differ in terms of any relevant clinical parameter apart from the percentage of granulocytes, which was 6% higher on average in those patients who died. Granulocytes were isolated from the blood of 15 healthy people and survivors and 15 patients who died within a week, and who were selected post hoc for analysis according to their matching gender and age. They differed only in the lethal outcome, which could not be predicted upon arrival at the hospital. The proteins level (respective ELISA), antioxidant activity (spectrophotometry), and lipid mediators (UPUPLC-MS) were measured in the peripheral blood granulocytes obtained via gradient centrifugation. The levels of Nrf2, HO-1, NFκB, and IL-6 were higher in the granulocytes of COVID-19 patients who died within a week, while the activity of cytoplasmic Cu,Zn-SOD and mitochondrial Mn-SOD and IL-2/IL-10 were lower in comparison to the levels observed in survivors. Furthermore, in the granulocytes of those patients who died, an increase in pro-inflammatory eicosanoids (PGE2 and TXB2), together with elevated cannabinoid receptors 1 and 2 (associated with a decrease in the anti-inflammatory 15d-PGJ2), were found. Hence, this study suggests that by triggering transcription factors, granulocytes activate inflammatory and redox signaling, leading to the production of pro-inflammatory eicosanoids while reducing cellular antioxidant capacity through SOD, thus expressing an altered response to COVID-19, which may result in the onset of systemic oxidative stress, ARDS, and the death of the patient.
Collapse
Affiliation(s)
- Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Wojciech Łuczaj
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Michał Biernacki
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Piotr Wójcik
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Iwona Jarocka-Karpowicz
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Biserka Orehovec
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
| | - Bruno Baršić
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
| | - Marko Tarle
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
| | - Marta Kmet
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
| | - Ivica Lukšić
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
- School of Medicine, University of Zagreb, HR-10000 Zagreb, Croatia
| | - Zlatko Marušić
- Division of Pathology, Clinical Hospital Centre Zagreb, HR-10000 Zagreb, Croatia;
| | - Georg Bauer
- Institute of Virology, Medical Center–University of Freiburg, 79104 Freiburg, Germany;
| | - Neven Žarković
- Laboratory for Oxidative Stress (LabOS), Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| |
Collapse
|
17
|
Root-Bernstein R, Huber J, Ziehl A, Pietrowicz M. SARS-CoV-2 and Its Bacterial Co- or Super-Infections Synergize to Trigger COVID-19 Autoimmune Cardiopathies. Int J Mol Sci 2023; 24:12177. [PMID: 37569555 PMCID: PMC10418384 DOI: 10.3390/ijms241512177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Autoimmune cardiopathies (AC) following COVID-19 and vaccination against SARS-CoV-2 occur at significant rates but are of unknown etiology. This study investigated the possible roles of viral and bacterial mimicry, as well as viral-bacterial co-infections, as possible inducers of COVID-19 AC using proteomic methods and enzyme-linked immunoadsorption assays. BLAST and LALIGN results of this study demonstrate that SARS-CoV-2 shares a significantly greater number of high quality similarities to some cardiac protein compared with other viruses; that bacteria such as Streptococci, Staphylococci and Enterococci also display very significant similarities to cardiac proteins but to a different set than SARS-CoV-2; that the importance of these similarities is largely validated by ELISA experiments demonstrating that polyclonal antibodies against SARS-CoV-2 and COVID-19-associated bacteria recognize cardiac proteins with high affinity; that to account for the range of cardiac proteins targeted by autoantibodies in COVID-19-associated autoimmune myocarditis, both viral and bacterial triggers are probably required; that the targets of the viral and bacterial antibodies are often molecularly complementary antigens such as actin and myosin, laminin and collagen, or creatine kinase and pyruvate kinase, that are known to bind to each other; and that the corresponding viral and bacterial antibodies recognizing these complementary antigens also bind to each other with high affinity as if they have an idiotype-anti-idiotype relationship. These results suggest that AC results from SARS-CoV-2 infections or vaccination complicated by bacterial infections. Vaccination against some of these bacterial infections, such as Streptococci and Haemophilus, may therefore decrease AC risk, as may the appropriate and timely use of antibiotics among COVID-19 patients and careful screening of vaccinees for signs of infection such as fever, diarrhea, infected wounds, gum disease, etc.
Collapse
Affiliation(s)
- Robert Root-Bernstein
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (J.H.); (A.Z.); (M.P.)
| | | | | | | |
Collapse
|
18
|
Müller S, Schultze JL. Systems analysis of human innate immunity in COVID-19. Semin Immunol 2023; 68:101778. [PMID: 37267758 PMCID: PMC10201327 DOI: 10.1016/j.smim.2023.101778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/13/2023] [Accepted: 05/13/2023] [Indexed: 06/04/2023]
Abstract
Recent developments in sequencing technologies, the computer and data sciences, as well as increasingly high-throughput immunological measurements have made it possible to derive holistic views on pathophysiological processes of disease and treatment effects directly in humans. We and others have illustrated that incredibly predictive data for immune cell function can be generated by single cell multi-omics (SCMO) technologies and that these technologies are perfectly suited to dissect pathophysiological processes in a new disease such as COVID-19, triggered by SARS-CoV-2 infection. Systems level interrogation not only revealed the different disease endotypes, highlighted the differential dynamics in context of disease severity, and pointed towards global immune deviation across the different arms of the immune system, but was already instrumental to better define long COVID phenotypes, suggest promising biomarkers for disease and therapy outcome predictions and explains treatment responses for the widely used corticosteroids. As we identified SCMO to be the most informative technologies in the vest to better understand COVID-19, we propose to routinely include such single cell level analysis in all future clinical trials and cohorts addressing diseases with an immunological component.
Collapse
Affiliation(s)
- Sophie Müller
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany; Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Genomics & Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany; Genomics & Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn, Bonn, Germany.
| |
Collapse
|
19
|
Zhao J, Zhu J, Huang C, Zhu X, Zhu Z, Wu Q, Yuan R. Uncovering the information immunology journals transmitted for COVID-19: A bibliometric and visualization analysis. Front Immunol 2022; 13:1035151. [PMID: 36405695 PMCID: PMC9670819 DOI: 10.3389/fimmu.2022.1035151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/17/2022] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Since the global epidemic of the coronavirus disease 2019 (COVID-19), a large number of immunological studies related to COVID-19 have been published in various immunology journals. However, the results from these studies were discrete, and no study summarized the important immunological information about COVID-19 released by these immunology journals. This study aimed to comprehensively summarize the knowledge structure and research hotspots of COVID-19 published in major immunology journals through bibliometrics. METHODS Publications on COVID-19 in major immunology journals were obtained from the Web of Science Core Collection. CiteSpace, VOSviewer, and R-bibliometrix were comprehensively used for bibliometric and visual analysis. RESULTS 1,331 and 5,000 publications of 10 journals with high impact factors and 10 journals with the most papers were included, respectively. The USA, China, England, and Italy made the most significant contributions to these papers. University College London, National Institute of Allergy and Infectious Diseases, Harvard Medical School, University California San Diego, and University of Pennsylvania played a central role in international cooperation in the immunology research field of COVID-19. Yuen Kwok Yung was the most important author in terms of the number of publications and citations, and the H-index. CLINICAL INFECTIOUS DISEASES and FRONTIERS IN IMMUNOLOGY were the most essential immunology journals. These immunology journals mostly focused on the following topics: "Delta/Omicron variants", "cytokine storm", "neutralization/neutralizing antibody", "T cell", "BNT162b2", "mRNA vaccine", "vaccine effectiveness/safety", and "long COVID". CONCLUSION This study systematically uncovered a holistic picture of the current research on COVID-19 published in major immunology journals from the perspective of bibliometrics, which will provide a reference for future research in this field.
Collapse
Affiliation(s)
- Jiefeng Zhao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jinfeng Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaojian Zhu
- Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhengming Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qinrong Wu
- Department of General Surgery, Yingtan City People’s Hospital, Yingtan, Jiangxi, China
| | - Rongfa Yuan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|