1
|
Tang X, Zhang C, Geng Q, Chen D, Ma W. Antibody-dependent enhancement of ORFV uptake into host cells. Virulence 2025; 16:2466503. [PMID: 39954287 PMCID: PMC11834454 DOI: 10.1080/21505594.2025.2466503] [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: 02/02/2024] [Revised: 12/28/2024] [Accepted: 02/09/2025] [Indexed: 02/17/2025] Open
Abstract
Orf virus (ORFV) has been demonstrated to infect both goat non-immune cells, specifically goat epithelial cells, and goat blood immune cells. Our previous studies have indicated that ORFV gains entry into goat epithelial cells via clathrin-mediated endocytosis and macropinocytosis pathways. However, the pathway by which ORFV enters goat blood immune cells has not yet been elucidated. Our findings revealed a differential viral internalization pathway in ORFV-infects goat immune cells contrasting the internalization pathways in goat epithelial cells, potentially involving an antibody-related mechanism. Therefore, our hypothesis posits that ORFV gains entry into goat immune cells via the antibody-dependent enhancement (ADE) pathway. Our experimental findings confirm the presence of the ADE effect in ORFV-infected goat immune cells, mediated by Fc receptors (FcRs) as demonstrated in antibody-blocking experiments. Furthermore, the ADE effect was also observed in goat epithelial cells. Nevertheless, the ADE effect observed in goat epithelial cells was not found to be dependent on the interaction between the virus-antibody complex and Fc receptors, as demonstrated by antibody-blocking experiments. Instead, it is suggested that an alternative mechanism involving the complement factor and complement receptors (CRs) may be responsible. Overall, this research offers insights into the unique ADE pathway of ORFV infection in different cell types, offering a novel perspective on the infection and pathogenic mechanisms of ORFV.
Collapse
Affiliation(s)
- Xidian Tang
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, China
- Academy of Science and Technology, Chuxiong Normal University, Chuxiong, Yunnan Province, China
| | - Chenyibo Zhang
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, China
| | - Qingru Geng
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, China
| | - Dekun Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, China
| | - Wentao Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province, China
| |
Collapse
|
2
|
Dai Z, Wang K, Bai C, Li Y, Yu Q, Chen Z, Liao J, Ding J, Wang Y. Discovery of a novel Thiazole amide inhibitor of Inflammasome and Pyroptosis pathways. Bioorg Chem 2025; 160:108477. [PMID: 40252370 DOI: 10.1016/j.bioorg.2025.108477] [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: 01/13/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Upon the activation of inflammasomes, inflammatory caspases cleave and activate gasdermin D (GSDMD), leading to pore formation that causes cell membrane rupture and amplifies downstream inflammatory responses. Dysregulated inflammasome activation and pyroptosis signaling pathways are implicated in numerous inflammatory diseases. In our work, a set of novel thiazole amide compounds with inhibitory activity against NLRP3 inflammasome-induced pyroptosis was identified. Of all the compounds tested, compound 21 demonstrated the most potent anti-pyroptotic effects. It suppressed GSDMD cleavage and decreased IL-1β and lactate dehydrogenase (LDH) release in a concentration-dependent manner. Compound 21 bound to NLRP3 protein and increased the thermal stability of NLRP3 concentration-dependently. The molecular docking and dynamics simulations revealed that compound 21 binds to the NLRP3 protein's active site, suppressing inflammasome activation. Further investigations showed that compound 21 also partially blocked upstream NF-κB signaling and downstream GSDMD N-terminal domain (GSDMD-NT) oligomerization, which explains its broad inhibitory effects on pyroptosis driven by multiple inflammasomes. Overall, this study presents a promising thiazole amide compound with inhibitory activity against inflammasome activation and subsequent pyroptosis, warranting further exploration.
Collapse
Affiliation(s)
- Zhen Dai
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, Sichuan, China.
| | - Ke Wang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, Sichuan, China
| | - Chenli Bai
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, Sichuan, China
| | - Yong Li
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing 402160, China.
| | - Quanwei Yu
- Targeted Tracer Research and development laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhiping Chen
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, Sichuan, China
| | - Jihong Liao
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, Sichuan, China
| | - Jianjun Ding
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Nanjing, China
| | - Yuxi Wang
- Targeted Tracer Research and development laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
3
|
Tonutti A, Motta F, Isailovic N, Selmi C, Timilsina S, Eric Gershwin M, De Santis M. Mechanistic considerations linking SARS-CoV-2 infection, inflammation, and the loss of immune tolerance. Curr Opin Immunol 2025; 95:102567. [PMID: 40412200 DOI: 10.1016/j.coi.2025.102567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/27/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025]
Abstract
The immune response to SARS-CoV-2 has been implicated in the onset of multiple, seemingly unrelated, autoimmune diseases. The immune response to SARS-CoV-2 has also been implicated in the unmasking and/or production of multiple autoantibodies, even in the absence of clinical disease. Despite such data, it remains unclear whether antibodies targeting antiviral signaling proteins and mitochondrial antigens reflect bystander activation or alternatively contribute to de novo viral immune escape mechanisms. With these comments in mind, a variety of professional antibody presenting cells and including lung resident macrophages of COVID-19 infected patients are impacted and dependent on the uptake of antibody-opsonized virus by Fcγ receptors; yet infection is aborted via antibody-dependent effector mechanisms or pyroptosis, possibly leading to autoantibody production, and autoinflammatory manifestations, respectively. TRIM21/Ro52, a cytosolic E3-ubiquitin ligase with an Fc-gamma receptor domain, functions as an intracytoplasmic antibody receptor, directs immune complexes binding virions but also autoantigens to autophagy. During autophagy, Ig-virions-TRIM21/Ro52-autoantigens complexes bind directly to class II human leukocyte antigen in lysosomal compartment, leading to subsequent presentation on the cell surface. This process favors the development of a specific humoral immune response but has the potential to lead to loss of tolerance. Interestingly, TRIM21/Ro52 can also contribute to pyroptosis. We propose that TRIM21/Ro52 is well-placed at the crossroad between the inflammatory response and clinical autoimmunity.
Collapse
Affiliation(s)
- Antonio Tonutti
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy; Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Francesca Motta
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy; Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Natasa Isailovic
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy; Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy.
| | - Suraj Timilsina
- Division of Rheumatology, Allergy and Clinical Immunology, University of California School of Medicine, Davis, CA, USA
| | - Merrill Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California School of Medicine, Davis, CA, USA
| | - Maria De Santis
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy; Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| |
Collapse
|
4
|
Wang Y, Cheng Y, Wang S, Liu D, Gao Y, Li J, Jiang Y, Cui W, Qiao X, Li Y, Wang L. Unraveling the cross-talk between a highly virulent PEDV strain and the host via single-cell transcriptomic analysis. J Virol 2025:e0055525. [PMID: 40396761 DOI: 10.1128/jvi.00555-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 04/28/2025] [Indexed: 05/22/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes severe intestinal damage and high mortality in neonatal piglets. The continuous emergence of new strains has brought new challenges to prevention and control. In this study, we isolated and characterized a prevalent PEDV virulent strain and analyzed 19,612 jejunal cells from PEDV-infected and control piglets using single-cell sequencing, revealing significant changes in cellular composition, gene expression, and intercellular communication. In response to PEDV infection, epithelial repair was enhanced through increased proliferation and differentiation of stem cells, transit-amplifying (TA) cells, and intestinal progenitor cells into enterocytes. Additionally, PEDV disrupted intercellular communication, compromising epithelial functionality while triggering immune responses, with IFN-γ and IL-10 signaling activation acting as critical regulators of immune balance and tissue homeostasis. Beyond enterocytes, viral genes were detected in various other cell types. Further experiments confirmed that PEDV could initiate replication in B and T lymphocytes but was unable to produce infectious progeny, with T cells additionally undergoing virus-induced apoptosis. These findings provide new insights into PEDV tropism, immune evasion, and epithelial repair, revealing complex host-pathogen interactions that shape disease progression and tissue regeneration, thereby contributing to a better understanding of enteric coronavirus pathogenesis.IMPORTANCEThe persistent circulation of porcine epidemic diarrhea virus (PEDV) poses a major threat to the swine industry, with emerging strains complicating prevention and control efforts. Currently, no effective measures completely prevent virus transmission, highlighting the need to understand PEDV-host interactions. In this study, we isolated a prevalent virulent strain and used single-cell sequencing to identify new PEDV-infected cell types and explore the complex interplay between the host and PEDV. These findings provide essential insights into viral pathogenesis and facilitate the design of targeted antiviral interventions.
Collapse
Affiliation(s)
- Yanan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yu Cheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Shuai Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Dan Liu
- China Institute of Veterinary Drug Control, Beijing, China
| | - Yueyi Gao
- China Institute of Veterinary Drug Control, Beijing, China
| | - Jiaxuan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Wen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xinyuan Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Li Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
5
|
Ghimire R, Shrestha R, Amaradhi R, Liu L, More S, Ganesh T, Ford AK, Channappanavar R. Toll-like receptor 7 (TLR7)-mediated antiviral response protects mice from lethal SARS-CoV-2 infection. J Virol 2025; 99:e0166824. [PMID: 40162785 PMCID: PMC12090760 DOI: 10.1128/jvi.01668-24] [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: 09/23/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced impaired antiviral immunity and excessive inflammatory responses cause lethal pneumonia. However, the in vivo roles of key pattern recognition receptors that elicit protective antiviral and fatal inflammatory responses, specifically in the lungs, are not well described. Coronaviruses possess single-stranded RNA genome that activates TLR7/8 to induce an antiviral interferon (IFN) and robust inflammatory cytokine response. Here, using wild-type and TLR7-deficient (TLR7-/-) mice infected with mouse-adapted SARS-CoV-2 (MA-CoV-2), we examined the role of TLR7 in the lung antiviral and inflammatory response and severe pneumonia. We showed that TLR7 deficiency significantly increased lung virus loads and morbidity/mortality, which correlated with reduced levels of type I IFNs (Ifna/b), type III IFNs (Ifnl), and IFN-stimulated genes (ISGs) in the lungs. A detailed evaluation of MA-CoV-2-infected lungs revealed increased neutrophil accumulation and lung pathology in TLR7-/- mice. We further showed that blocking type I IFN receptor (IFNAR) signaling enhanced SARS-CoV-2 replication in the lungs and caused severe lung pathology, leading to 100% mortality compared to infected control mice. Moreover, immunohistochemical assessment of the lungs revealed increased numbers of SARS-CoV-2 antigen-positive macrophages, pneumocytes, and bronchial epithelial cells in TLR7-/- and IFNAR-deficient mice compared to control mice. In summary, we conclusively demonstrated that despite TLR7-induced robust lung inflammation, TLR7-induced IFN/ISG responses suppress lung virus replication and pathology and provide protection against SARS-CoV-2-induced fatal pneumonia. Additionally, given the similar disease outcomes in control, TLR7-/-, and IFNAR-deficient MA-CoV-2-infected mice and coronavirus disease 2019 (COVID-19) patients, we propose that MA-CoV-2-infected mice constitute an excellent model for studying COVID-19.IMPORTANCESevere coronavirus disease 2019 (COVID-19) is caused by a delicate balance between a strong antiviral and an exuberant inflammatory response. A robust antiviral immunity and regulated inflammation are protective, while a weak antiviral response and excessive inflammation are detrimental. However, the key host immune sensors that elicit protective antiviral and inflammatory responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge are poorly defined. Here, we examined the role of viral RNA-mediated TLR7 activation in the lung antiviral and inflammatory responses in SARS-CoV-2-infected mice. We demonstrate that TLR7 deficiency led to a high rate of morbidity and mortality, which correlated with an impaired antiviral interferon (IFN)-I/III response, enhanced lung virus replication, and severe lung pathology. Furthermore, we show that blocking IFN-I signaling using anti-IFN receptor antibody promoted SARS-CoV-2 replication in the lungs and caused severe disease. These results provide conclusive evidence that TLR7 and IFN-I receptor deficiencies lead to severe disease in mice, replicating clinical features observed in COVID-19 patients.
Collapse
Affiliation(s)
- Roshan Ghimire
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Rakshya Shrestha
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lin Liu
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alexandra K. Ford
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Rudragouda Channappanavar
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
6
|
Ghosh S, Das Sarma J. The age-dependent neuroglial interaction with peripheral immune cells in coronavirus-induced neuroinflammation with a special emphasis on COVID-19. Biogerontology 2025; 26:111. [PMID: 40380990 DOI: 10.1007/s10522-025-10252-9] [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: 02/28/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
Neurodegenerative diseases are chronic progressive disorders that impair memory, cognition, and motor functions, leading to conditions such as dementia, muscle weakness, and speech difficulties. Aging disrupts the stringent balance between pro- and anti-inflammatory cytokines, increasing neuroinflammation, which contributes to neurodegenerative diseases. The aging brain is particularly vulnerable to infections due to a weakened and compromised immune response and impaired integrity of the blood-brain barrier, allowing pathogens like viruses to trigger neurodegeneration. Coronaviruses have been linked to both acute and long-term neurological complications, including cognitive impairments, psychiatric disorders, and neuroinflammation. The virus can induce a cytokine storm, damaging the central nervous system (CNS) and worsening existing neurological conditions. Though its exact mechanism of neuroinvasion remains elusive, evidence suggests it disrupts the blood-brain barrier and triggers immune dysregulation, leading to persistent neurological sequelae in elderly individuals. This review aims to understand the interaction between the peripheral immune system and CNS glial cells in aged individuals, which is imperative in addressing coronavirus-induced neuroinflammation and concomitant neurodegeneration.
Collapse
Affiliation(s)
- Satavisha Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata, 741246, India.
- Department of Ophthalmology, University of Pennsylvania, 19104, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Liu S, Xu W, Tu B, Xiao Z, Li X, Huang L, Yuan X, Zhou J, Yang X, Yang J, Chang D, Chen W, Wang FS. Early Immunological and Inflammation Proteomic Changes in Elderly COVID-19 Patients Predict Severe Disease Progression. Biomedicines 2025; 13:1162. [PMID: 40426989 PMCID: PMC12108611 DOI: 10.3390/biomedicines13051162] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Elderly patients infected with SARS-CoV-2 are at higher risk of developing cytokine storm and severe outcomes; however, specific immunological and proteomic biomarkers for early prediction remain unclear in this vulnerable group. Methods: We enrolled 182 elderly COVID-19 patients from the Chinese PLA General Hospital between November 2022 and April 2023, categorizing them based on progression to respiratory failure requiring mechanical ventilation (defined as severe progression). Olink proteomic analysis was performed on admission serum from 40 propensity score-matched samples, with differentially expressed proteins (DEPs) validated by cytometric bead array (CBA) in 178 patients. To predict severe progression, a model was developed using a 70% training set and validated on a 30% validation set. LASSO regression screened features followed by logistic regression and receiver operating characteristic (ROC) analysis to optimize the model by incrementally incorporating features ranked by random forest importance. Results: Elderly patients progressing to severe COVID-19 exhibited early immune dysregulation, including neutrophilia, lymphopenia, monocytopenia, elevated procalcitonin (PCT), C-reactive protein (CRP), interleukin-6 (IL-6), neutrophil-to-lymphocyte ratio (NLR), and systemic immune-inflammation index (SII), as well as coagulation dysfunction and multi-organ injury. Proteomics identified a set of biomarkers, including tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and revealed disruptions in signaling pathways, including the mTOR and VEGF signaling pathways. The optimal predictive model, which incorporated PCT, IL-6, monocyte percentage, lymphocyte count, and TRAIL, achieved an area under curve (AUC) of 0.870 (0.729-1.000) during validation. TRAIL levels negatively correlated with fibrinogen (p < 0.05). Conclusions: Elderly COVID-19 patients with severe progression demonstrate early immune dysregulation, hyperinflammation, coagulation dysfunction, and multi-organ injury. The model we proposed effectively predicts disease progression in elderly COVID-19 patients, providing potential biomarkers for early clinical risk stratification in this vulnerable population.
Collapse
Affiliation(s)
- Shiyang Liu
- Medical School of Chinese PLA, Beijing 100853, China;
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Wen Xu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Bo Tu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Zhiqing Xiao
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100007, China; (Z.X.); (D.C.)
| | - Xue Li
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
- Yu-Yue Pathology Scientific Research Center, Chongqing 401329, China
| | - Lei Huang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Xin Yuan
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Juanjuan Zhou
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Xinxin Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Junlian Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - De Chang
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100007, China; (Z.X.); (D.C.)
| | - Weiwei Chen
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| |
Collapse
|
8
|
Chen B, Farzan M, Choe H. SARS-CoV-2 spike protein: structure, viral entry and variants. Nat Rev Microbiol 2025:10.1038/s41579-025-01185-8. [PMID: 40328900 DOI: 10.1038/s41579-025-01185-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been a devastating global pandemic for 4 years and is now an endemic disease. With the emergence of new viral variants, COVID-19 is a continuing threat to public health despite the wide availability of vaccines. The virus-encoded trimeric spike protein (S protein) mediates SARS-CoV-2 entry into host cells and also induces strong immune responses, making it an important target for development of therapeutics and vaccines. In this Review, we summarize our latest understanding of the structure and function of the SARS-CoV-2 S protein, the molecular mechanism of viral entry and the emergence of new variants, and we discuss their implications for development of S protein-related intervention strategies.
Collapse
Affiliation(s)
- Bing Chen
- Division of Molecular Medicine, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
| | - Michael Farzan
- Division of Infectious Diseases, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
- Center for Integrated Solutions for Infectious Diseases (CISID), The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Hyeryun Choe
- Division of Infectious Diseases, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Mezouar S, Mege J. Monitoring Macrophage Polarization in Infectious Disease, Lesson From SARS-CoV-2 Infection. Rev Med Virol 2025; 35:e70034. [PMID: 40148134 PMCID: PMC11976041 DOI: 10.1002/ird3.70006] [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: 03/11/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The concept of macrophage polarization has been largely used in human diseases to define a typology of activation of myeloid cells reminiscent of lymphocyte functional subsets. In COVID-19, several studies have investigated myeloid compartment dysregulation and macrophage polarization as an indicator of disease prognosis and monitoring. SARS-CoV-2 induces an in vitro activation state in monocytes and macrophages that does not match the polarization categories in most studies. In COVID-19 patients, monocytes and macrophages are activated but they do not show a polarization profile. Therefore, the investigation of polarization under basic conditions was not relevant to assess monocyte and macrophage activation. The analysis of monocytes and macrophages with high-throughput methods has allowed the identification of new functional subsets in the context of COVID-19. This approach proposes an innovative stratification of myeloid cell activation. These new functional subsets of myeloid cells would be better biomarkers to assess the risk of complications in COVID-19, reserving the concept of polarization for pharmacological programme evaluation. This review reappraises the polarization of monocytes and macrophages in viral infections, particularly in COVID-19.
Collapse
Affiliation(s)
- Soraya Mezouar
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Faculty of Medical and Paramedical SciencesAix‐Marseille UniversityHIPE Human LabMarseilleFrance
| | - Jean‐Louis Mege
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Department of ImmunologyLa Timone HospitalMarseilleFrance
| |
Collapse
|
10
|
Min R, Bai Y, Wang NR, Liu X. Gasdermins in pyroptosis, inflammation, and cancer. Trends Mol Med 2025:S1471-4914(25)00090-5. [PMID: 40307076 DOI: 10.1016/j.molmed.2025.04.003] [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: 11/23/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Pyroptosis is a type of programmed inflammatory cell death characterized by balloon-like swelling, membrane rupture, and the release of inflammatory cytokines and danger signals. Pyroptosis is directly triggered by activated gasdermins (GSDMs) which bind to membrane phospholipids, oligomerize, and form pores in cell membranes. GSDM activation is mediated by various effector proteases via cleavage of the linker region or post-translational modification to release the active N-terminal fragment in response to a variety of pathogenic or intrinsic danger signals. GSDM-mediated pyroptosis is involved in the pathogenesis of an array of infectious and inflammatory diseases and cancers. This review discusses recent advances related to the physiological and pathological functions of GSDM-mediated pyroptosis, as well as therapeutic strategies targeting pyroptosis.
Collapse
Affiliation(s)
- Rui Min
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Bai
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ning-Rui Wang
- School of Laboratory Medicine, Nanchang Medical College, Nanchang, Jiangxi 330052, China
| | - Xing Liu
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China.
| |
Collapse
|
11
|
Zhang J, Wang D, Kwok C, Xu L, Famulok M. Aptamer-engaged nanotherapeutics against SARS-CoV-2. DISCOVER NANO 2025; 20:71. [PMID: 40289185 PMCID: PMC12034613 DOI: 10.1186/s11671-025-04245-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/26/2025] [Indexed: 04/30/2025]
Abstract
The COVID-19 pandemic, caused by the virus SARS-CoV-2 infection, has underscored the critical importance of rapid and accurate therapeutics. The neutralization of SARS-CoV-2 is paramount in controlling the spread and impact of COVID-19. In this context, the integration of aptamers and aptamer-related nanotherapeutics presents a valuable and scientifically significant approach. Despite the potential, current reviews in this area are often not comprehensive and specific enough to encapsulate the full scope of therapeutic principles, strategies, advancements, and challenges. This review aims to fill that gap by providing an in-depth examination of the role of aptamers and their related molecular medicine in COVID-19 therapeutics. We first introduce the unique properties, selection, and recognition mechanism of aptamers to bind with high affinity to various targets. Next, we delve into the therapeutic potential of aptamers, focusing on their ability to inhibit viral entry and replication, as well as modulate the host immune response. The integration of aptamers with nucleic acid nanomedicine is explored. Finally, we address the challenges and future perspectives of aptamer and nucleic acid nanomedicine in COVID-19 therapeutics, including issues of stability, delivery, and manufacturing scalability. We conclude by underscoring the importance of continued research and development in this field to meet the ongoing challenges posed by COVID-19 and potential future pandemics. Our review will be a valuable resource for researchers and clinicians interested in the latest developments at the intersection of molecular biology, nanotechnology, and infectious disease management.
Collapse
Affiliation(s)
- Jing Zhang
- Life Science and Chemistry College, Hunan University of Technology, Zhuzhou, 412007, China
| | - Dan Wang
- Life & Medical Sciences Institute (LIMES), Pharmaceutical Institute, Universität Bonn, 53121, Bonn, Germany.
| | - Chiu Kwok
- Life & Medical Sciences Institute (LIMES), Pharmaceutical Institute, Universität Bonn, 53121, Bonn, Germany
| | - Liujun Xu
- Department of Respiratory and Critical Care, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, 324000, China.
| | - Michalina Famulok
- Life & Medical Sciences Institute (LIMES), Pharmaceutical Institute, Universität Bonn, 53121, Bonn, Germany
| |
Collapse
|
12
|
Davanzo GG, Castelucci BG, de Souza GF, Muraro SP, Menezes Dos Reis L, de Oliveira IB, Fachi JL, Virgilio-da-Silva JV, Berçot MR, Fernandes MF, de Oliveira S, Araujo NVP, Ribeiro G, de Castro G, Costa WLG, Santoro AL, Rodrigues-Luiz GF, do Carmo HRP, Breder I, Mori MA, Farias AS, Martins-de-Souza D, Guarnieri JW, Wallace DC, Vinolo MAR, Proença-Módena JL, Beheshti A, Sposito AC, Moraes-Vieira PM. Obesity-Induced Metabolic Priming Exacerbates SARS-CoV-2 Inflammation. Immunology 2025. [PMID: 40265287 DOI: 10.1111/imm.13934] [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: 01/07/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
Despite the early recognition that individuals living with obesity are more prone to develop adverse outcomes during COVID-19, the mechanisms underlying these conditions are still unclear. During obesity, an accumulation of free fatty acids (FFAs) in the circulation promotes low-grade inflammation. Here, we show that FFAs induce epigenetic reprogramming of monocytes, exacerbating their inflammatory profile after SARS-CoV-2 infection, a mechanism named metabolic-primed immunity. Monocytes from people with obesity or primed with palmitate, a central component of circulating FFAs, presented elevated viral load and higher gene expression of IL-6. Palmitate-primed monocytes upregulate fatty acid oxidation and FFAs entry into the mitochondria. FFA-derived acetyl-CoA is then converted into citrate, exiting the mitochondria and is used to support H3K18 histone acetylation, which regulates IL-6 accessibility. Ingestion of palm oil by lean and healthy individuals increased circulating FFAs levels and was sufficient to exacerbate the inflammatory profile of monocytes upon SARS-CoV-2 infection. Our findings demonstrate that obesity-derived FFAs induce the metabolic priming of monocytes, which exacerbates the inflammatory response observed in people with severe COVID-19.
Collapse
Affiliation(s)
- Gustavo Gastão Davanzo
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Bianca Gazieri Castelucci
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Gabriela Fabiano de Souza
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Stéfanie Primon Muraro
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Larissa Menezes Dos Reis
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | | | - José Luís Fachi
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - João Victor Virgilio-da-Silva
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Marcelo Rodrigues Berçot
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Mariane Font Fernandes
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Nathalia Vitoria Pereira Araujo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Guilherme Ribeiro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Gisele de Castro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Webster Leonardo Guimarães Costa
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Adriana Leandra Santoro
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Gabriela Flavia Rodrigues-Luiz
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Helison Rafael P do Carmo
- Department of Clinical Medicine, School of Medical Sciences, University of Campinas, São Paulo, Brazil
| | - Ikaro Breder
- Department of Clinical Medicine, School of Medical Sciences, University of Campinas, São Paulo, Brazil
| | - Marcelo A Mori
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Alessandro S Farias
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
- Autoimmune Research Laboratory, Department of Genetics, Microbiology, and Immunology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Joseph W Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
| | - José Luiz Proença-Módena
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
| | - Afshin Beheshti
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Space Biomedicine, McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrei C Sposito
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Pedro M Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
| |
Collapse
|
13
|
Peng X, Han Y, Xue S, Zhou Y, Jiang W, Xia A, Wu W, Gao Y, Wu F, Wang Q. Low Antibody-Dependent Enhancement of Viral Entry Activity Supports the Safety of Inactivated SARS-CoV-2 Vaccines. Vaccines (Basel) 2025; 13:425. [PMID: 40333308 PMCID: PMC12031465 DOI: 10.3390/vaccines13040425] [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: 02/22/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND/OBJECTIVES The antibody-dependent enhancement (ADE) of viral entry has been documented for SARS-CoV-2 infection both in vitro and in vivo. However, the potential for the SARS-CoV-2 vaccination to elicit similar ADE effects remains unclear. METHODS In this study, we assessed the in vitro ADE potential of monoclonal antibodies (mAbs) derived from individuals vaccinated with the inactivated SARS-CoV-2 vaccine and compared them to those from one convalescent donor. RESULTS Our analysis revealed no significant difference in binding affinity or neutralizing capacity between the vaccinated and convalescent mAbs. However, the inactivated SARS-CoV-2 vaccination induced fewer ADE-inducing mAbs, particularly those targeting the Class III epitope on the receptor-binding domain (RBD) compared to those from the convalescent individual. Moreover, no significant in vitro ADE was detected in either vaccinated or convalescent sera, indicating low levels of ADE-inducing antibodies in the sera. CONCLUSIONS An inactivated SARS-CoV-2 vaccination induces fewer ADE-inducing antibodies compared to natural infection, further emphasizing the safety of inactivated SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Xiaofang Peng
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| | - Yuru Han
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| | - Song Xue
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| | - Yunjiao Zhou
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai 201619, China;
| | - Weiyu Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| | - Anqi Xia
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| | - Wei Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| | - Yidan Gao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| | - Fan Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Fudan University, Shanghai 200040, China; (X.P.); (Y.H.); (S.X.); (W.J.); (A.X.); (W.W.); (Y.G.)
| |
Collapse
|
14
|
Lage SL, Bricker-Holt K, Rocco JM, Rupert A, Donovan FX, Abramzon YA, Chandrasekharappa SC, McNinch C, Cook L, Amaral EP, Rosenfeld G, Dalhuisen T, Eun A, Hoh R, Fehrman E, Martin JN, Deeks SG, Henrich TJ, Peluso MJ, Sereti I. Persistent immune dysregulation and metabolic alterations following SARS-CoV-2 infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.16.25325949. [PMID: 40321289 PMCID: PMC12047922 DOI: 10.1101/2025.04.16.25325949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
SARS-CoV-2 can cause a variety of post-acute sequelae including Long COVID19 (LC), a complex, multisystem disease characterized by a broad range of symptoms including fatigue, cognitive impairment, and post-exertional malaise. The pathogenesis of LC is incompletely understood. In this study, we performed comprehensive cellular and transcriptional immunometabolic profiling within a cohort that included SARS-CoV-2-naïve controls (NC, N=30) and individuals with prior COVID-19 (~4-months) who fully recovered (RC, N=38) or went on to experience Long COVID symptoms (N=58). Compared to the naïve controls, those with prior COVID-19 demonstrated profound metabolic and immune alterations at the proteomic, cellular, and epigenetic level. Specifically, there was an enrichment in immature monocytes with sustained inflammasome activation and oxidative stress, elevated arachidonic acid levels, decreased tryptophan, and variation in the frequency and phenotype of peripheral T-cells. Those with LC had increased CD8 T-cell senescence and a distinct transcriptional profile within CD4 and CD8 T-cells and monocytes by single cell RNA sequencing. Our findings support a profound and persistent immunometabolic dysfunction that follows SARS-CoV-2 which may form the pathophysiologic substrate for LC. Our findings suggest that trials of therapeutics that help restore immune and metabolic homeostasis may be warranted to prevent, reduce, or resolve LC symptoms.
Collapse
Affiliation(s)
- Silvia Lucena Lage
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Katherine Bricker-Holt
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Joseph M. Rocco
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Adam Rupert
- AIDS Monitoring Laboratory, Frederick National Laboratory for Cancer Research; Frederick, USA
| | - Frank X. Donovan
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute; Bethesda, USA
| | - Yevgeniya A. Abramzon
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute; Bethesda, USA
| | | | - Colton McNinch
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Logan Cook
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Eduardo Pinheiro Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| | - Gabriel Rosenfeld
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Thomas Dalhuisen
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Avery Eun
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Emily Fehrman
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco; San Francisco, USA
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Timothy J. Henrich
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Michael J. Peluso
- Department of Medicine, University of California, San Francisco; San Francisco, USA
| | - Irini Sereti
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, USA
| |
Collapse
|
15
|
Wu PX, Yang WP, Feng T, Zhang J, Zhu GQ, Du XG, Ru Y, Zhao YF, Wu S, Li D, Zheng HX. African swine fever virus I177L induces host inflammatory responses by facilitating the TRAF6-TAK1 axis and NLRP3 inflammasome assembly. J Virol 2025; 99:e0208024. [PMID: 40135893 PMCID: PMC11998506 DOI: 10.1128/jvi.02080-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
African swine fever virus (ASFV) is the pathogen of African swine fever (ASF), and its infection causes a lethal disease in pigs, with severe pathological lesions. These changes indicate excessive inflammatory responses in infected pigs, which is the main cause of death, but the ASFV proteins worked in this physiological process and the mechanisms underlying ASFV-induced inflammation remain unclear. Here, we identify that viral I177L works in these inflammatory responses. Mechanistically, I177L facilitates TRAF6 ubiquitination that enhances its binding to TAK1, which promotes TAK1 ubiquitination and phosphorylation. These processes depend on the E3 ubiquitin ligase activity of TRAF6. The upregulation of I177L to TRAF6-TAK1 interaction and TAK1 activation is responsible for I177L's activated effect on the NF-κB signaling pathway. Additionally, I177L promotes assembly of the NLRP3 inflammasome and ASC oligomerization, thus leading to the activation of the NLRP3 inflammasome and the production and secretion of mature IL-1β. TAK1 inhibition efficiently reverses ASFV-activated NF-κB signaling and inflammatory responses and suppresses ASFV replication. Furthermore, I177L-deficient ASFV induces milder inflammatory responses in pigs compared with parental ASFV, which still protects pigs against ASFV challenge. The finding confirms ASFV I177L as an important proinflammatory protein in vitro and in vivo and reveals a key mechanism underlying ASFV-mediated inflammatory responses for the first time, which enriches our knowledge of the complex ASFV, thus benefiting our understanding of the interplay between ASFV infection and the host's inflammatory responses.IMPORTANCEAfrican swine fever (ASF) is a devastating viral disease in pigs, and excessive inflammatory responses induced by ASFV mainly cause death. Thus, the study of the proinflammatory virulent proteins and the detailed mechanisms are important to ASF control. Here, I177L was demonstrated to be an essential protein in ASFV-mediated inflammation, which performs by simultaneously activating the NF-κB signaling and the NLRP3 inflammasome. The finding elucidates the molecular mechanism underlying ASFV-activated inflammatory responses for the first time. It provides a theoretical foundation for reducing the high mortality caused by excessive inflammation and opens new avenues for small-molecule drug development and vaccine design targeting ASFV.
Collapse
Affiliation(s)
- Pan-Xue Wu
- State Key Laboratory of Animal Biotech Breeding College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wen-Ping Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Tao Feng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Guo-Qiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xu-Guang Du
- State Key Laboratory of Animal Biotech Breeding College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
| | - Yi Ru
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Yao-Feng Zhao
- State Key Laboratory of Animal Biotech Breeding College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
| | - Sen Wu
- State Key Laboratory of Animal Biotech Breeding College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
| | - Dan Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Hai-Xue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
16
|
Brady C, Tipton T, Carnell O, Longet S, Gooch K, Hall Y, Salguero J, Tomic A, Carroll M. A systems biology approach to define SARS-CoV-2 correlates of protection. NPJ Vaccines 2025; 10:69. [PMID: 40229322 PMCID: PMC11997207 DOI: 10.1038/s41541-025-01103-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
Correlates of protection (CoPs) for SARS-CoV-2 have yet to be sufficiently defined. This study uses the machine learning platform, SIMON, to accurately predict the immunological parameters that reduced clinical pathology or viral load following SARS-CoV-2 challenge in a cohort of 90 non-human primates. We found that anti-SARS-CoV-2 spike antibody and neutralising antibody titres were the best predictors of clinical protection and low viral load in the lung. Since antibodies to SARS-CoV-2 spike showed the greatest association with clinical protection and reduced viral load, we next used SIMON to investigate the immunological features that predict high antibody titres. It was found that a pre-immunisation response to seasonal beta-HCoVs and a high frequency of peripheral intermediate and non-classical monocytes predicted low SARS-CoV-2 spike IgG titres. In contrast, an elevated T cell response as measured by IFNγ ELISpot predicted high IgG titres. Additional predictors of clinical protection and low SARS-CoV-2 burden included a high abundance of peripheral T cells. In contrast, increased numbers of intermediate monocytes predicted clinical pathology and high viral burden in the throat. We also conclude that an immunisation strategy that minimises pathology post-challenge did not necessarily mediate viral control. This would be an important finding to take forward into the development of future vaccines aimed at limiting the transmission of SARS-CoV-2. These results contribute to SARS-CoV-2 CoP definition and shed light on the factors influencing the success of SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Caolann Brady
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom.
| | - Tom Tipton
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
| | - Oliver Carnell
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Stephanie Longet
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- International Center for Infectiology Research (CIRI), Team GIMAP, Claude Bernard Lyon 1 University, Saint-Etienne, France
| | - Karen Gooch
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Yper Hall
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Javier Salguero
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Adriana Tomic
- National Emerging Infectious Diseases Laboratories, Boston, MA, USA
- Department of Virology, Immunology & Microbiology, Boston University Medical School, Boston, MA, USA
- Biomedical Engineering, Boston University, College of Engineering, Boston, MA, USA
| | - Miles Carroll
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
17
|
Mukherjee S, Bayry J. The Yin and Yang of TLR4 in COVID-19. Cytokine Growth Factor Rev 2025; 82:70-85. [PMID: 39490235 DOI: 10.1016/j.cytogfr.2024.10.001] [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: 07/28/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 11/05/2024]
Abstract
Various pattern recognition receptors (PRRs), including toll-like receptors (TLRs), play a crucial role in recognizing invading pathogens as well as damage-associated molecular patterns (DAMPs) released in response to infection. The resulting signaling cascades initiate appropriate immune responses to eliminate these pathogens. Current evidence suggests that SARS-CoV-2-driven activation of TLR4, whether through direct recognition of the spike glycoprotein (alone or in combination with endotoxin) or by sensing various TLR4-activating DAMPs or alarmins released during viral infection, acts as a critical mediator of antiviral immunity. However, TLR4 exerts a dual role in COVID-19, demonstrating both beneficial and deleterious effects. Dysregulated TLR4 signaling is implicated in the proinflammatory consequences linked to the immunopathogenesis of COVID-19. Additionally, TLR4 polymorphisms contribute to severity of the disease. Given its significant immunoregulatory impact on COVID-19 immunopathology and host immunity, TLR4 has emerged as a key target for developing inhibitors and immunotherapeutic strategies to mitigate the adverse effects associated with SARS-CoV-2 and related infections. Furthermore, TLR4 agonists are also being explored as adjuvants to enhance immune responses to SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory (IBIL), Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal 713 340, India.
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris 75006, France; Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678 623, India.
| |
Collapse
|
18
|
Bai Y, Pan Y, Liu X. Mechanistic insights into gasdermin-mediated pyroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00837-0. [PMID: 40128620 DOI: 10.1038/s41580-025-00837-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2025] [Indexed: 03/26/2025]
Abstract
Pyroptosis, a novel mode of inflammatory cell death, is executed by membrane pore-forming gasdermin (GSDM) family members in response to extracellular or intracellular injury cues and is characterized by a ballooning cell morphology, plasma membrane rupture and the release of inflammatory mediators such as interleukin-1β (IL-1β), IL-18 and high mobility group protein B1 (HMGB1). It is a key effector mechanism for host immune defence and surveillance against invading pathogens and aberrant cancerous cells, and contributes to the onset and pathogenesis of inflammatory and autoimmune diseases. Manipulating the pore-forming activity of GSDMs and pyroptosis could lead to novel therapeutic strategies. In this Review, we discuss the current knowledge regarding how GSDM-mediated pyroptosis is initiated, executed and regulated, its roles in physiological and pathological processes, and the crosstalk between different modes of programmed cell death. We also highlight the development of drugs that target pyroptotic pathways for disease treatment.
Collapse
Affiliation(s)
- Yang Bai
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Youdong Pan
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xing Liu
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Academy of Natural Sciences (SANS), Shanghai, China.
| |
Collapse
|
19
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [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: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
20
|
Weiss S, Lin HM, Acosta E, Komarova NL, Chen P, Wodarz D, Baine I, Duerr R, Wajnberg A, Gervais A, Bastard P, Casanova JL, Arinsburg SA, Swartz TH, Aberg JA, Bouvier NM, Liu ST, Alvarez RA, Chen BK. Post-transfusion activation of coagulation pathways during severe COVID-19 correlates with COVID-19 convalescent plasma antibody profiles. J Clin Invest 2025; 135:e181136. [PMID: 40091845 PMCID: PMC11910229 DOI: 10.1172/jci181136] [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: 04/05/2024] [Accepted: 01/24/2025] [Indexed: 03/19/2025] Open
Abstract
Early antibody therapy can prevent severe SARS-CoV-2 infection (COVID-19). However, the effectiveness of COVID-19 convalescent plasma (CCP) therapy in treating severe COVID-19 remains inconclusive. To test a hypothesis that some CCP units are associated with a coagulopathy hazard in severe disease that offsets its benefits, we tracked 304 CCP units administered to 414 hospitalized COVID-19 patients to assess their association with the onset of unfavorable post-transfusion D-dimer trends. CCP recipients with increasing or persistently elevated D-dimer trajectories after transfusion experienced higher mortality than those whose D-dimer levels were persistently low or decreasing after transfusion. Within the CCP donor-recipient network, recipients with increasing or persistently high D-dimer trajectories were skewed toward association with a minority of CCP units. In in vitro assays, CCP from "higher-risk" units had higher cross-reactivity with the spike protein of human seasonal betacoronavirus OC43. "Higher-risk" CCP units also mediated greater Fcγ receptor IIa signaling against cells expressing SARS-CoV-2 spike compared with "lower-risk" units. This study finds that post-transfusion activation of coagulation pathways during severe COVID-19 is associated with specific CCP antibody profiles and supports a potential mechanism of immune complex-activated coagulopathy.
Collapse
Affiliation(s)
| | - Hung-Mo Lin
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | - Dominik Wodarz
- Department of Ecology, Behavior and Evolution, UCSD, La Jolla, California, USA
| | - Ian Baine
- Department of Transfusion Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ralf Duerr
- Department of Medicine
- Department of Microbiology, and
- Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Ania Wajnberg
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adrian Gervais
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | | | | | | | - Nicole M. Bouvier
- Division of Infectious Diseases and
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sean T.H. Liu
- Division of Infectious Diseases and
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | |
Collapse
|
21
|
Cui J, Li H, Ye D, Zhang G, Zhang Y, Yang L, Sim MMS, Wood JP, Wei Y, Li Z, Wu C. Inhibiting NINJ1-dependent plasma membrane rupture protects against inflammasome-induced blood coagulation and inflammation. eLife 2025; 12:RP91329. [PMID: 40094828 PMCID: PMC11913443 DOI: 10.7554/elife.91329] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
Systemic blood coagulation accompanies inflammation during severe infections like sepsis and COVID. We previously established a link between coagulopathy and pyroptosis, a vital defense mechanism against infection. During pyroptosis, the formation of gasdermin-D (GSDMD) pores on the plasma membrane leads to the release of tissue factor (TF)-positive microvesicles (MVs) that are procoagulant. Mice lacking GSDMD release fewer of these procoagulant MVs. However, the specific mechanisms coupling the activation of GSDMD to MV release remain unclear. Plasma membrane rupture (PMR) in pyroptosis was recently reported to be actively mediated by the transmembrane protein Ninjurin-1 (NINJ1). Here, we show that NINJ1 promotes procoagulant MV release during pyroptosis. Haploinsufficiency or glycine inhibition of NINJ1 limited the release of procoagulant MVs and inflammatory cytokines, and partially protected against blood coagulation and lethality triggered by bacterial flagellin. Our findings suggest a crucial role for NINJ1-dependent PMR in inflammasome-induced blood coagulation and inflammation.
Collapse
Affiliation(s)
- Jian Cui
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
| | - Hua Li
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
| | - Dien Ye
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
| | - Guoying Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Yan Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Ling Yang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Martha MS Sim
- Department of Molecular and Cellular Biochemistry, University of KentuckyLexingtonUnited States
| | - Jeremy P Wood
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
- Department of Molecular and Cellular Biochemistry, University of KentuckyLexingtonUnited States
- The Gill Heart and Vascular Institute, College of Medicine, University of KentuckyLexingtonUnited States
| | - Yinan Wei
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Zhenyu Li
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Congqing Wu
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of KentuckyLexingtonUnited States
- Department of Surgery, College of Medicine, University of KentuckyLexingtonUnited States
| |
Collapse
|
22
|
Ebenig A, Lange MV, Gellhorn Serra M, Kupke A, Plesker R, Qu B, Brown RJP, Maier TJ, Mühlebach MD. Differential efficacy of first licensed western vaccines protecting without immunopathogenesis Wuhan-1-challenged hamsters from severe COVID-19. NPJ Vaccines 2025; 10:51. [PMID: 40097436 PMCID: PMC11914482 DOI: 10.1038/s41541-025-01100-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
Four COVID-19 vaccines were developed, tested, and authorized early in Europe and the US. Comirnaty and Spikevax are mRNA-based, whereas Jcovden and Vaxzevria utilize adenoviral vectors (AdV). We described a hamster model of COVID-19 utilizing Wuhan-1 strain SARS-CoV-2, in which vaccine-associated immunopathogenesis can be induced by Alum-adjuvanted Spike protein (Alum+S). Such animals were vaccinated with the authorized vaccines or Alum+S, challenged, and examined. All vaccinated hamsters produced antibodies targeting S. Neutralizing antibodies (nAb) were induced only by authorized vaccines. While nAbs were present after one vaccination with AdV-vaccines, mRNA vaccines needed a boost immunization. Upon challenge, all authorized vaccines protected from severe disease. Less tissue damage and no live virus (one exception) were detectable in the lungs. In contrast, Alum+S immunized hamsters developed VAERD. Our data reveal the absence of induction of VAERD by early commercial vaccines in hamsters, while animals´ immune responses and protection seem to match the clinical vaccine efficacy.
Collapse
Affiliation(s)
- Aileen Ebenig
- Division Veterinary Medicine, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Mona V Lange
- Division Veterinary Medicine, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | | | - Alexandra Kupke
- Institute for Virology, Philipps University, 35043, Marburg, Germany
- German Center for Infection Research, Gießen-Marburg-Langen, Germany
| | - Roland Plesker
- Division Veterinary Medicine, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Bingqian Qu
- Division Veterinary Medicine, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Richard J P Brown
- Division Veterinary Medicine, Paul-Ehrlich-Institut, 63225, Langen, Germany
- Department of Translational and Computational Infection Research, Ruhr University Bochum, 44801, Bochum, Germany
| | - Thorsten J Maier
- Division Safety of Biomedicines and Diagnostics, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Michael D Mühlebach
- Division Veterinary Medicine, Paul-Ehrlich-Institut, 63225, Langen, Germany.
- German Center for Infection Research, Gießen-Marburg-Langen, Germany.
| |
Collapse
|
23
|
Rodrigues TS, Zamboni DS. Inflammasome Activation by RNA Respiratory Viruses: Mechanisms, Viral Manipulation, and Therapeutic Insights. Immunol Rev 2025; 330:e70003. [PMID: 39891396 DOI: 10.1111/imr.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 02/03/2025]
Abstract
Respiratory viruses, including SARS-CoV-2, influenza, parainfluenza, rhinovirus, and respiratory syncytial virus (RSV), are pathogens responsible for lower respiratory tract infections, particularly in vulnerable populations such as children and the elderly. Upon infection, these viruses are recognized by pattern recognition receptors, leading to the activation of inflammasomes, which are essential for mediating inflammatory responses. This review discusses the mechanisms by which these RNA respiratory viruses activate inflammasomes, emphasizing the roles of various signaling pathways and components involved in this process. Additionally, we highlight the specific interactions between viral proteins and inflammasome sensors, elucidating how these viruses manipulate the host immune response to facilitate infection. Understanding the dynamics of inflammasome activation in response to respiratory viruses provides critical insights for developing immunomodulatory therapeutic strategies aimed at mitigating inflammation and improving outcomes in respiratory tract infections.
Collapse
Affiliation(s)
- Tamara S Rodrigues
- Department of Cell Biology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Dario S Zamboni
- Department of Cell Biology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
24
|
Centofanti E, Oyler-Yaniv A, Oyler-Yaniv J. Deep learning-based image classification reveals heterogeneous execution of cell death fates during viral infection. Mol Biol Cell 2025; 36:ar29. [PMID: 39841552 PMCID: PMC11974948 DOI: 10.1091/mbc.e24-10-0438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Cell fate decisions, such as proliferation, differentiation, and death, are driven by complex molecular interactions and signaling cascades. While significant progress has been made in understanding the molecular determinants of these processes, historically, cell fate transitions were identified through light microscopy that focused on changes in cell morphology and function. Modern techniques have shifted toward probing molecular effectors to quantify these transitions, offering more precise quantification and mechanistic understanding. However, challenges remain in cases where the molecular signals are ambiguous, complicating the assignment of cell fate. During viral infection, programmed cell death (PCD) pathways, including apoptosis, necroptosis, and pyroptosis, exhibit complex signaling and molecular cross-talk. This can lead to simultaneous activation of multiple PCD pathways, which confounds assignment of cell fate based on molecular information alone. To address this challenge, we employed deep learning-based image classification of dying cells to analyze PCD in single herpes simplex virus-1 (HSV-1)-infected cells. Our approach reveals that despite heterogeneous activation of signaling, individual cells adopt predominantly prototypical death morphologies. Nevertheless, PCD is executed heterogeneously within a uniform population of virus-infected cells and varies over time. These findings demonstrate that image-based phenotyping can provide valuable insights into cell fate decisions, complementing molecular assays.
Collapse
Affiliation(s)
- Edoardo Centofanti
- The Department of Systems Biology at Harvard Medical School, Boston, MA 02115
| | - Alon Oyler-Yaniv
- The Department of Systems Biology at Harvard Medical School, Boston, MA 02115
| | | |
Collapse
|
25
|
Rubio-Casillas A, Redwan EM, Uversky VN. More antibodies are not always better: Fc effector functions play a critical role in SARS-CoV-2 infection and protection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:413-447. [PMID: 40246351 DOI: 10.1016/bs.pmbts.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Traditional vaccinology has primarily focused on neutralizing antibody titers as the main correlate of vaccine efficacy, often overlooking the multifaceted roles of antibody Fc effector functions in orchestrating protective immune responses. Fc-mediated immune responses play a pivotal role in immune modulation and pathogen clearance. Emerging evidence from natural infections and vaccine studies highlights the critical contribution of Fc effector functions in determining the quality and durability of immunity. This work explores the limitations of current vaccine evaluation paradigms that prioritize neutralization over Fc effector mechanisms. It also describes findings from a study showing an unexpected role for SARS-CoV-2 anti-spike antibodies: both convalescent plasma and patient-derived monoclonal antibodies (mAbs) lead to maximum phagocytic capacity by monocytes at low concentrations, whereas at higher concentrations the phagocytic capacity was reduced. Given that the severity of COVID-19 disease and antibody titers are strongly positively correlated, this work challenges the paradigm that high antibodies offer better protection against severe disease. It is proposed that humoral and cellular responses elicited by vaccination should never be higher than those produced by natural infection. By integrating antibody Fc effector functions into vaccine development, a paradigm shift is proposed that emphasizes synergic antibody responses. Such an approach could transform vaccine efficacy assessment, enhance protection against dangerous pathogens, and drive innovation in vaccine design.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
26
|
Fratta Pasini AM, Stranieri C, Di Leo EG, Bertolone L, Aparo A, Busti F, Castagna A, Vianello A, Chesini F, Friso S, Girelli D, Cominacini L. Identification of Early Biomarkers of Mortality in COVID-19 Hospitalized Patients: A LASSO-Based Cox and Logistic Approach. Viruses 2025; 17:359. [PMID: 40143288 PMCID: PMC11946718 DOI: 10.3390/v17030359] [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: 12/05/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to identify possible early biomarkers of mortality among clinical and biochemical parameters, iron metabolism parameters, and cytokines detected within 24 h from admission in hospitalized COVID-19 patients. We enrolled 80 hospitalized patients (40 survivors and 40 non-survivors) with COVID-19 pneumonia and acute respiratory failure. The median time from the onset of COVID-19 symptoms to hospital admission was lower in non-survivors than survivors (p < 0.05). Respiratory failure, expressed as the ratio of arterial oxygen partial pressure to the fraction of inspired oxygen (P/F), was more severe in non-survivors than survivors (p < 0.0001). Comorbidities were similar in both groups. Among biochemical parameters and cytokines, eGFR and interleukin (IL)-1β were found to be significantly lower (p < 0.05), while LDH, IL-10, and IL-8 were significantly higher in non-survivors than in survivors (p < 0.0005, p < 0.05 and p < 0.005, respectively). Among other parameters, LDH values distribution showed the most significant difference between study groups (p < 0.0001). LASSO feature selection combined with Cox proportional hazards and logistic regression models was applied to identify features distinguishing between survivors and non-survivors. Both approaches highlighted LDH as the strongest predictor, with IL-22 and creatinine emerging in the Cox model, while IL-10, eGFR, and creatinine were influential in the logistic model (AUC = 0.744 for Cox, 0.723 for logistic regression). In a similar manner, we applied linear regression for predicting LDH levels, identifying the P/F ratio as the top predictor, followed by IL-10 and eGFR (NRMSE = 0.128). Collectively, these findings underscore LDH's critical role in mortality prediction, with P/F and IL-10 as key determinants of LDH increases in this Italian COVID-19 cohort.
Collapse
Affiliation(s)
- Anna Maria Fratta Pasini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Chiara Stranieri
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Edoardo Giuseppe Di Leo
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Lorenzo Bertolone
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Antonino Aparo
- Interdepartmental Laboratory of Medical Research, Research Center LURM, University of Verona, 37134 Verona, Italy;
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy (S.F.)
| | - Alice Vianello
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Fabio Chesini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Simonetta Friso
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy (S.F.)
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Luciano Cominacini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| |
Collapse
|
27
|
Li Y, Guo B. GSDMD-mediated pyroptosis: molecular mechanisms, diseases and therapeutic targets. MOLECULAR BIOMEDICINE 2025; 6:11. [PMID: 39994107 PMCID: PMC11850691 DOI: 10.1186/s43556-025-00249-8] [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: 08/22/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Pyroptosis is a regulated form of inflammatory cell death in which Gasdermin D (GSDMD) plays a central role as the key effector molecule. GSDMD-mediated pyroptosis is characterized by complex biological features and considerable heterogeneity in its expression, mechanisms, and functional outcomes across various tissues, cell types, and pathological microenvironments. This heterogeneity is particularly pronounced in inflammation-related diseases and tumors. In the context of inflammatory diseases, GSDMD expression is typically upregulated, and its activation in macrophages, neutrophils, T cells, epithelial cells, and mitochondria triggers both pyroptotic and non-pyroptotic pathways, leading to the release of pro-inflammatory cytokines and exacerbation of tissue damage. However, under certain conditions, GSDMD-mediated pyroptosis may also serve a protective immune function. The expression of GSDMD in tumors is regulated in a more complex manner, where it can either promote immune evasion or, in some instances, induce tumor cell death. As our understanding of GSDMD's role continues to progress, there have been advancements in the development of inhibitors targeting GSDMD-mediated pyroptosis; however, these therapeutic interventions remain in the preclinical phase. This review systematically examines the cellular and molecular complexities of GSDMD-mediated pyroptosis, with a particular emphasis on its roles in inflammation-related diseases and cancer. Furthermore, it underscores the substantial therapeutic potential of GSDMD as a target for precision medicine, highlighting its promising clinical applications.
Collapse
Affiliation(s)
- Yujuan Li
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China.
| | - Bin Guo
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| |
Collapse
|
28
|
Sepúlveda-Delgado J, Llorente L, Hernández-Doño S. A Comprehensive Review of Fc Gamma Receptors and Their Role in Systemic Lupus Erythematosus. Int J Mol Sci 2025; 26:1851. [PMID: 40076476 PMCID: PMC11899777 DOI: 10.3390/ijms26051851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/27/2025] [Accepted: 02/02/2025] [Indexed: 03/14/2025] Open
Abstract
Receptors for the immunoglobulin G constant fraction (FcγRs) are widely expressed in cells of the immune system. Complement-independent phagocytosis prompted FcγR research to show that the engagement of IgG immune complexes with FcγRs triggers a variety of cell host immune responses, such as phagocytosis, antibody-dependent cell cytotoxicity, and NETosis, among others. However, variants of these receptors have been implicated in the development of and susceptibility to autoimmune diseases such as systemic lupus erythematosus. Currently, the knowledge of FcγR variants is a required field of antibody therapeutics, which includes the engineering of recombinant soluble human Fc gamma receptors, enhancing the inhibitory and blocking the activating FcγRs function, vaccines, and organ transplantation. Importantly, recent interest in FcγRs is the antibody-dependent enhancement (ADE), a mechanism by which the pathogenesis of certain viral infections is enhanced. ADEs may be responsible for the severity of the SARS-CoV-2 infection. Therefore, FcγRs have become a current research topic. Therefore, this review briefly describes some of the historical knowledge about the FcγR type I family in humans, including the structure, affinity, and mechanism of ligand binding, FcγRs in diseases such as systemic lupus erythematosus (SLE), and the potential therapeutic approaches related to these receptors in SLE.
Collapse
Affiliation(s)
- Jesús Sepúlveda-Delgado
- Research Division, Servicios de Salud IMSS BIENESTAR, Hospital Regional de Alta Especialidad Ciudad Salud, Tapachula 30700, Mexico;
| | - Luis Llorente
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14000, Mexico
| | - Susana Hernández-Doño
- Physiology and Pharmacology Department, Chemistry and Pharmacy Faculty, Universidad de El Salvador, San Salvador 01101, El Salvador
| |
Collapse
|
29
|
Tian C, Ao Z, Cerneckis J, Cai H, Chen L, Niu H, Takayama K, Kim J, Shi Y, Gu M, Kanekiyo T, Guo F. Understanding monocyte-driven neuroinflammation in Alzheimer's disease using human brain organoid microphysiological systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.16.638539. [PMID: 40027735 PMCID: PMC11870548 DOI: 10.1101/2025.02.16.638539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) pathogenesis strongly correlates with neuroinflammation. Peripheral monocytes are crucial components of the human immune system that may play a role in neuroinflammation, but their contribution to AD pathogenesis is largely understudied partially due to the lack of appropriate human models. Here, we present human cortical organoid microphysiological systems (hCO-MPSs) for modeling dynamic AD neuroinflammation mediated by monocytes. By incorporating 3D printed devices into an existing cortical organoid protocol, 96 hCO-MPSs can be established with significantly reduced necrosis and hypoxia as well as enhanced viability within a commonly used 96 well plate, and each hCO-MPS consists of a doughnut-shaped hCO and a 3D printed device per well. Using this approach, monocytes from AD patients exhibit higher infiltration, decreased amyloid-beta (Aβ) clearance, and stronger inflammatory responses compared to monocytes from age-matched control donors. Moreover, pro-inflammatory effects such as elevated astrocyte activation and neuronal apoptosis were observed to be induced by AD monocytes. Furthermore, the significant increase in the expression of IL1B and CCL3, both at the transcriptional and protein levels, indicated the pivotal role of these cytokine and chemokine in monocyte-mediated AD neuroinflammation. Our findings provide insight for understanding monocytes' role in AD pathogenesis, and the user-friendly MPS models we present are compatible with existing laboratory settings, highlighting their potential for modeling neuroinflammation and developing new therapeutics for various neuroinflammatory diseases.
Collapse
|
30
|
Tajti G, Gebetsberger L, Pamlitschka G, Aigner-Radakovics K, Leitner J, Steinberger P, Stockinger H, Ohradanova-Repic A. Cyclophilin-CD147 interaction enables SARS-CoV-2 infection of human monocytes and their activation via Toll-like receptors 7 and 8. Front Immunol 2025; 16:1460089. [PMID: 39963132 PMCID: PMC11830813 DOI: 10.3389/fimmu.2025.1460089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Monocytes and macrophages, as important constituents of the innate immune system, are equipped with multiple Toll-like-receptors (TLRs) to recognize invading pathogens, such as SARS-CoV-2, and mount an antiviral response. Nevertheless, their uncontrolled activation can lead to hyperinflammation seen in severe COVID-19. Surprisingly, we observed that recombinant SARS-CoV-2 Spike (S) and Nucleocapsid (N) proteins triggered only a weak proinflammatory response in human peripheral blood monocytes. By employing THP-1 and Jurkat NF-κB::eGFP reporter cell lines expressing specific TLRs, various TLR ligands and blocking antibodies, we determined that surface TLRs, including TLR2/1, TLR2/6 and TLR4 do not play a major role in SARS-CoV-2 sensing. However, monocytes are potently activated by the replication-competent SARS-CoV-2, and the response correlates with the viral uptake that is observed only in monocytes, but not in lymphocytes. We show that monocyte activation involves two distinct steps. Firstly, SARS-CoV-2 infects monocytes in a process independent of the S protein and the prime SARS-CoV-2 receptor angiotensin-converting enzyme 2. Instead, the alternative SARS-CoV-2 receptor CD147, which is highly expressed on monocytes, recognizes its well-known interaction partners cyclophilins A and B that are incorporated into SARS-CoV-2 virions. Secondly, upon viral uptake via the cyclophilin-CD147 interaction, that can be inhibited by specific CD147 blocking antibodies or competition with recombinant human cyclophilin A and B, SARS-CoV-2 RNA is recognized by TLR7/8 in endosomes, leading to upregulation of tumor necrosis factor (TNF), interleukin (IL)-1β and IL-6, comprising the core hyperinflammatory signature. Taken together, our data reveal a novel mechanism how human monocytes sense SARS-CoV-2 and suggest that targeting the cyclophilin-CD147 axis might be beneficial to alleviate overt myeloid-driven inflammation triggered by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Gabor Tajti
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Laura Gebetsberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Gregor Pamlitschka
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Katharina Aigner-Radakovics
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Judith Leitner
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Peter Steinberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Hannes Stockinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Anna Ohradanova-Repic
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| |
Collapse
|
31
|
Mbow M, Hoving D, Cisse M, Diallo I, Honkpehedji YJ, Huisman W, Pothast CR, Jongsma MLM, König MH, de Kroon AC, Linh LTK, Azimi S, Tak T, Kruize YCM, Kurniawan F, Dia YA, Zhang JLH, Prins C, Roukens AHE, de Vries JJC, Wammes LJ, Smits HH, Adegnika AA, Zlei M, Kuijpers TW, Wieske L, Dieye A, Mboup S, Kremsner PG, Eftimov F, Velavan TP, Berlin I, Heemskerk MHM, Yazdanbakhsh M, Jochems SP. Immune responses to SARS-CoV-2 in sub-Saharan Africa and western Europe: a retrospective, population-based, cross-sectional study. THE LANCET. MICROBE 2025; 6:100942. [PMID: 39708825 DOI: 10.1016/j.lanmic.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/17/2024] [Accepted: 07/03/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND SARS-CoV-2 has been associated with a higher proportion of asymptomatic infections and lower mortality in sub-Saharan Africa than high-income countries. However, there is currently a lack of data on cellular immune responses to SARS-CoV-2 in people living in Africa compared with people in high-income regions of the world. We aimed to assess geographical variation in peripheral and mucosal immune responses. METHODS In this retrospective, population-based, cross-sectional study, we analysed peripheral blood and nasal curettage samples from seven clinical studies involving individuals from Senegal (Senegalese cohort), the Netherlands, and Germany (European cohort). Samples were collected between Nov 1, 2018, and Dec 20, 2021. We included samples from individuals with no, mild, or severe COVID-19. A validation cohort of individuals from Senegal and Gabon (n=64) was used to validate key findings from the main cohort. Matching of individuals between geographical regions by age, sex, viral load, and infection severity and duration was used to address confounding factors. We examined the cellular, humoral, and cytokine immune responses using cytometry by time of flight, spectral flow cytometry, ELISA, and Luminex. FINDINGS We included 133 individuals (59 from the Senegalese cohort and 74 from the European cohort). In contrast to the European cohort, mild COVID-19 in the Senegalese cohort was not associated with any statistically significant perturbations in blood or nasal immune cell profiles, nor with increased pro-inflammatory cytokines, although SARS-CoV-2-specific adaptive immunity was readily induced, as seen in Europeans. In severe COVID-19, both the Senegalese and European cohorts showed lymphopenia (Senegal: 2·9-times decrease, p=0·0010 vs Europe: 1·6-times decrease, p=0·0046) and increased neutrophil frequencies in blood (Senegal: 2·0-times increase, p=0·0044 vs Europe: 1·3-times increase, p=0·026) and the nasal mucosa CD66b+CD16low neutrophils (Senegal: 9·9-times increase, p=0·045 vs Europe: 392-times increase, p<0·0001). However, in contrast to Europeans, the Senegalese cohort had no significant expansion of immature immune populations, inflammasome activation, or monocyte recruitment to the nasal mucosa. INTERPRETATION The observed divergent immunological trajectories during SARS-CoV-2 infection offer a potential explanation for the reported attenuated disease course in sub-Saharan Africa and highlight the need to further investigate immune responses to SARS-CoV-2 in understudied populations. FUNDING European and Developing Countries Clinical Trials Partnership 2 programme (AIDCO), LUMC Gisela Thier Fellowship, Dutch Research Council (NWO), European Research Council, and Leids Universitair Fonds.
Collapse
Affiliation(s)
- Moustapha Mbow
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal; Institute of Health Research, Epidemiological Surveillance and Training, Dakar, Senegal
| | - Dennis Hoving
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Marouba Cisse
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal; Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Ibrahima Diallo
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal; Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Yabo J Honkpehedji
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Wesley Huisman
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Cilia R Pothast
- Department of Hematology Leiden University Medical Centre, Leiden, Netherlands
| | - Marlieke L M Jongsma
- Department of Cell and Chemical Biology, ONCODE Institute Leiden University Medical Centre, Leiden, Netherlands
| | - Marion H König
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Alicia C de Kroon
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Le Thi Kieu Linh
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany; Vietnamese-German Center for Medical Research, VG-CARE, Hanoi, Viet Nam
| | - Shohreh Azimi
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Tamar Tak
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Yvonne C M Kruize
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Farid Kurniawan
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands; Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Yacine Amet Dia
- Institute of Health Research, Epidemiological Surveillance and Training, Dakar, Senegal
| | - Jaimie L H Zhang
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Corine Prins
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Anna H E Roukens
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Jutte J C de Vries
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Linda J Wammes
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Hermelijn H Smits
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Ayola A Adegnika
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon; Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany; The German Center for Infection Research (DZIF), Tübingen, Germany
| | - Mihaela Zlei
- Department of Immunology Leiden University Medical Centre, Leiden, Netherlands; Department of Flow Cytometry, Regional Institute of Oncology, Iasi, Romania
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Disease Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Luuk Wieske
- Department of Neurology and Neurophysiology Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Alioune Dieye
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Souleymane Mboup
- Institute of Health Research, Epidemiological Surveillance and Training, Dakar, Senegal
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon; Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany; The German Center for Infection Research (DZIF), Tübingen, Germany
| | - Filip Eftimov
- Department of Neurology and Neurophysiology Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Thirumalaisamy P Velavan
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany; The German Center for Infection Research (DZIF), Tübingen, Germany; Vietnamese-German Center for Medical Research, VG-CARE, Hanoi, Viet Nam; Faculty of Medicine, Duy Tan University, Da Nang, Viet Nam
| | - Ilana Berlin
- Department of Cell and Chemical Biology, ONCODE Institute Leiden University Medical Centre, Leiden, Netherlands
| | | | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Simon P Jochems
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands.
| |
Collapse
|
32
|
Zodda E, Pons M, DeMoya-Valenzuela N, Calvo-González C, Benítez-Rodríguez C, López-Ayllón BD, Hibot A, Zuin A, Crosas B, Cascante M, Montoya M, Pujol MD, Rubio-Martínez J, Thomson TM. Induction of the Inflammasome by the SARS-CoV-2 Accessory Protein ORF9b, Abrogated by Small-Molecule ORF9b Homodimerization Inhibitors. J Med Virol 2025; 97:e70145. [PMID: 39902616 DOI: 10.1002/jmv.70145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/07/2024] [Accepted: 11/07/2024] [Indexed: 02/05/2025]
Abstract
Viral accessory proteins play critical roles in viral escape from host innate immune responses and in viral inflammatory pathogenesis. Here we show that the SARS-CoV-2 accessory protein, ORF9b, but not other SARS-CoV-2 accessory proteins (ORF3a, ORF3b, ORF6, ORF7, ORF8, ORF9c, and ORF10), strongly activates inflammasome-dependent caspase-1 in A549 lung carcinoma cells and THP-1 monocyte-macrophage cells. Exposure to lipopolysaccharide (LPS) and ATP additively enhanced the activation of caspase-1 by ORF9b, suggesting that ORF9b and LPS follow parallel pathways in the activation of the inflammasome and caspase-1. Following rational in silico approaches, we have designed small molecules capable of inhibiting the homodimerization of ORF9b, which experimentally inhibited ORF9b-ORF9b homotypic interactions, caused mitochondrial eviction of ORF9b, inhibited ORF9b-induced activation of caspase-1 in A549 and THP-1 cells, cytokine release in THP-1 cells, and restored type I interferon (IFN-I) signaling suppressed by ORF9b in both cell models. These small molecules are first-in-class compounds targeting a viral accessory protein critical for viral-induced exacerbated inflammation and escape from innate immune responses, with the potential of mitigating the severe immunopathogenic damage induced by highly pathogenic coronaviruses and restoring antiviral innate immune responses curtailed by viral infection.
Collapse
Grants
- This work was funded by the Spanish National Research Council (CSIC, project numbers CSIC-COV19-006, CSIC-COV-19-201, CSIC-COV-19-117, SGL2103019, SGL2103015, 202020E079 and 202320E187 and LINCGLOBAL INCGL20009), the Catalan Agency for Management of University and Research Grants (AGAUR, 2020PANDE00048, 2021SGR1490, 2021SGR00350), the Spanish Ministry of Science (PID2021-123399OB-I00), the CSIC's Global Health Platform (PTI Salud Global), The Networked Center for Biomedical Research in Liver and Digestive Diseases (CIBER-EHD), the Spanish Structures and Excellence María de Maeztu program (CEX2021-001202-M), the European Commission-Next Generation EU (Regulation EU 2020/2094), and INDICASAT-AIP.
Collapse
Affiliation(s)
- Erika Zodda
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Mònica Pons
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Natàlia DeMoya-Valenzuela
- Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona, Spain
- Theoretical and Computational Chemistry Research Institute (IQTCUB), Barcelona, Spain
| | - Cristina Calvo-González
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Cristina Benítez-Rodríguez
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Blanca D López-Ayllón
- Viral immunology Lab, Molecular Biomedicine Department, Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| | - Achraf Hibot
- Laboratory of Pharmaceutical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Alice Zuin
- Regulation of the Proteasome Laboratory, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Bernat Crosas
- Regulation of the Proteasome Laboratory, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBER-EHD), Madrid, Spain
| | - María Montoya
- Viral immunology Lab, Molecular Biomedicine Department, Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| | - María D Pujol
- Laboratory of Pharmaceutical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Jaime Rubio-Martínez
- Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona, Spain
- Theoretical and Computational Chemistry Research Institute (IQTCUB), Barcelona, Spain
| | - Timothy M Thomson
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBER-EHD), Madrid, Spain
- High-Altitude Research Institute (IIA), Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Investigaciones Científicas y Servicio de Alta Tecnología (INDICASAT AIP), Panama City, Panama
| |
Collapse
|
33
|
Benamar M, Lai PS, Huang CY, Chen Q, Oktelik FB, Contini P, Wang M, Okin D, Crestani E, Fong J, Fion TMC, Gokbak MN, Harb H, Phipatanakul W, Marri L, Vassallo C, Guastalla A, Kim M, Sui HY, Berra L, Goldberg MB, Angelini C, De Palma R, Chatila TA. Notch4 regulatory T cells and SARS-CoV-2 viremia shape COVID19 survival outcome. Allergy 2025; 80:557-569. [PMID: 39361431 PMCID: PMC11805648 DOI: 10.1111/all.16333] [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: 03/28/2024] [Revised: 08/05/2024] [Accepted: 09/07/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Immune dysregulation and SARS-CoV-2 plasma viremia have been implicated in fatal COVID-19 disease. However, how these two factors interact to shape disease outcomes is unclear. METHODS We carried out viral and immunological phenotyping on a prospective cohort of 280 patients with COVID-19 presenting to acute care hospitals in Boston, Massachusetts and Genoa, Italy between June 1, 2020 and February 8, 2022. Disease severity, mortality, plasma viremia, and immune dysregulation were assessed. A mouse model of lethal H1N1 influenza infection was used to analyze the therapeutic potential of Notch4 and pyroptosis inhibition in disease outcome. RESULTS Stratifying patients based on %Notch4+ Treg cells and/or the presence of plasma viremia identified four subgroups with different clinical trajectories and immune phenotypes. Patients with both high %Notch4+ Treg cells and viremia suffered the most disease severity and 90-day mortality compared to the other groups even after adjusting for baseline comorbidities. Increased Notch4 and plasma viremia impacted different arms of the immune response in SARS-CoV-2 infection. Increased Notch4 was associated with decreased Treg cell amphiregulin expression and suppressive function whereas plasma viremia was associated with increased monocyte cell pyroptosis. Combinatorial therapies using Notch4 blockade and pyroptosis inhibition induced stepwise protection against mortality in a mouse model of lethal H1N1 influenza infection. CONCLUSIONS The clinical trajectory and survival outcome in hospitalized patients with COVID-19 is predicated on two cardinal factors in disease pathogenesis: viremia and Notch4+ Treg cells. Intervention strategies aimed at resetting the immune dysregulation in COVID-19 by antagonizing Notch4 and pyroptosis may be effective in severe cases of viral lung infection.
Collapse
Affiliation(s)
- Mehdi Benamar
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Peggy S. Lai
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Ching-Ying Huang
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Qian Chen
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Fatma Betul Oktelik
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Paola Contini
- Department of Internal Medicine, Unit of Clinical Immunology and Translational Medicine, University of Genova, Italy
- Clinical Immunology Division, IRCCS-San Martino Hospital-Genova
| | - Muyun Wang
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Okin
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Elena Crestani
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason Fong
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Tsz Man Chan Fion
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Merve Nida Gokbak
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Hani Harb
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Institute for Medical Microbiology and Virology, Technical University Dresden, Germany
| | - Wanda Phipatanakul
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Luca Marri
- Department of Internal Medicine, Unit of Clinical Immunology and Translational Medicine, University of Genova, Italy
- Clinical Immunology Division, IRCCS-San Martino Hospital-Genova
| | - Chiara Vassallo
- Clinical Immunology Division, IRCCS-San Martino Hospital-Genova
| | | | - Minsik Kim
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hui-Yu Sui
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lorenzo Berra
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marcia B. Goldberg
- Infectious Diseases Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Claudia Angelini
- Istituto per le Applicazioni del Calcolo “M. Picone”, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Raffaele De Palma
- Department of Internal Medicine, Unit of Clinical Immunology and Translational Medicine, University of Genova, Italy
- CNR-Institute of Biomolecular Chemistry (IBC), Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | - Talal A. Chatila
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Li J, Yang Y, Zhao C, Zhao J, Wang X, Ye S, Wang D, Zhou C, Li J, Wang S, Li K, Liu C, He X, Qin J. Microglial C/EBPβ-Fcgr1 regulatory axis blocking inhibits microglial pyroptosis and improves neurological recovery. J Neuroinflammation 2025; 22:29. [PMID: 39891259 PMCID: PMC11786472 DOI: 10.1186/s12974-025-03362-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
CAAT/Enhancer Binding Protein β (C/EBPβ) is associated with inflammatory responses in neurodegenerative pathologies, particularly in the brain. However, the regulatory role of C/EBPβ in spinal cord injury and its impact on neurological recovery remain unknown. In this study, we observed significant upregulation of C/EBPβ in microglia after spinal cord injury in mice and was associated with neuroinflammation. Knocking down C/EBPβ in the spinal cord attenuated microglia pyroptosis, reduced the production of proinflammatory cytokines, and inhibited neuronal apoptosis. Mechanistically, C/EBPβ promoted the transcription of Fcgr1, which was involved in activating microglia pyroptosis. In both in-vivo and in-vitro experiments, knocking down Cebpb or Fcgr1, or the pyroptosis inhibitor VX765 inhibited neuronal apoptosis and improved neurological recovery in mice. These findings indicate that C/EBPβ functions as a key regulator that participates in the microglia pyroptosis-mediated neuroinflammation by activating Fcgr1 transcription.
Collapse
Affiliation(s)
- Jing Li
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yubing Yang
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chenguang Zhao
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jinghao Zhao
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaohui Wang
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shengshou Ye
- Department of Neurology, Qinghai Cardiocerebrovascular Disease Specialised Hospital, Xining, Qinghai, China
| | - Dong Wang
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chengdong Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jie Li
- Institute of Photonics and Photon-technology, Northwest University, Xi'an, Shaanxi, China
| | - Shuang Wang
- Institute of Photonics and Photon-technology, Northwest University, Xi'an, Shaanxi, China
| | - Ke Li
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chunmiao Liu
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xijing He
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Orthopedics, Xi'an International Rehabilitation Medical Center, Xi'an, Shaanxi, China
| | - Jie Qin
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
35
|
Stegeman SK, Kourko O, Amsden H, Pellizzari Delano IE, Mamatis JE, Roth M, Colpitts CC, Gee K. RNA Viruses, Toll-Like Receptors, and Cytokines: The Perfect Storm? J Innate Immun 2025; 17:126-153. [PMID: 39820070 PMCID: PMC11845175 DOI: 10.1159/000543608] [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: 07/26/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs. BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs.
Collapse
Affiliation(s)
- Sophia K Stegeman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Olena Kourko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Heather Amsden
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - John E Mamatis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Madison Roth
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
36
|
Saller BS, Wöhrle S, Fischer L, Dufossez C, Ingerl IL, Kessler S, Mateo-Tortola M, Gorka O, Lange F, Cheng Y, Neuwirt E, Marada A, Koentges C, Urban C, Aktories P, Reuther P, Giese S, Kirschnek S, Mayer C, Pilic J, Falquez-Medina H, Oelgeklaus A, Deepagan VG, Shojaee F, Zimmermann JA, Weber D, Tai YH, Crois A, Ciminski K, Peyronnet R, Brandenburg KS, Wu G, Baumeister R, Heimbucher T, Rizzi M, Riedel D, Helmstädter M, Buescher J, Neumann K, Misgeld T, Kerschensteiner M, Walentek P, Kreutz C, Maurer U, Rambold AS, Vince JE, Edlich F, Malli R, Häcker G, Kierdorf K, Meisinger C, Köttgen A, Jakobs S, Weber ANR, Schwemmle M, Groß CJ, Groß O. Acute suppression of mitochondrial ATP production prevents apoptosis and provides an essential signal for NLRP3 inflammasome activation. Immunity 2025; 58:90-107.e11. [PMID: 39571574 DOI: 10.1016/j.immuni.2024.10.012] [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: 02/13/2024] [Revised: 08/15/2024] [Accepted: 10/24/2024] [Indexed: 01/18/2025]
Abstract
How mitochondria reconcile roles in functionally divergent cell death pathways of apoptosis and NLRP3 inflammasome-mediated pyroptosis remains elusive, as is their precise role in NLRP3 activation and the evolutionarily conserved physiological function of NLRP3. Here, we have shown that when cells were challenged simultaneously, apoptosis was inhibited and NLRP3 activation prevailed. Apoptosis inhibition by structurally diverse NLRP3 activators, including nigericin, imiquimod, extracellular ATP, particles, and viruses, was not a consequence of inflammasome activation but rather of their effects on mitochondria. NLRP3 activators turned out as oxidative phosphorylation (OXPHOS) inhibitors, which we found to disrupt mitochondrial cristae architecture, leading to trapping of cytochrome c. Although this effect was alone not sufficient for NLRP3 activation, OXPHOS inhibitors became triggers of NLRP3 when combined with resiquimod or Yoda-1, suggesting that NLRP3 activation requires two simultaneous cellular signals, one of mitochondrial origin. Therefore, OXPHOS and apoptosis inhibition by NLRP3 activators provide stringency in cell death decisions.
Collapse
Affiliation(s)
- Benedikt S Saller
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Svenja Wöhrle
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Larissa Fischer
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Clara Dufossez
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Isabella L Ingerl
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susanne Kessler
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Maria Mateo-Tortola
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Felix Lange
- Research Group Mitochondrial Structure and Dynamics, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Clinic for Neurology, University Medical Center of Göttingen, Göttingen, Germany
| | - Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Emilia Neuwirt
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Adinarayana Marada
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Koentges
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Chiara Urban
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Philipp Aktories
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Peter Reuther
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sebastian Giese
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susanne Kirschnek
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Carolin Mayer
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Johannes Pilic
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Hugo Falquez-Medina
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Aline Oelgeklaus
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Veerasikku Gopal Deepagan
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Farzaneh Shojaee
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Julia A Zimmermann
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Damian Weber
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Yi-Heng Tai
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians Universität München, Munich, Germany; Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany; Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Anna Crois
- Faculty of Biology, University of Freiburg, Freiburg, Germany; Institute for Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kevin Ciminski
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Remi Peyronnet
- Institute for Experimental Cardiovascular Medicine, Faculty of Medicine, University Heart Center Freiburg - Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Katharina S Brandenburg
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Gang Wu
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ralf Baumeister
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Thomas Heimbucher
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marta Rizzi
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Department of Rheumatology and Clinical Immunology and Center for Chronic Immunodeficiency, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Riedel
- Laboratory for Electron Microscopy, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Martin Helmstädter
- EMcore, Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Joerg Buescher
- Metabolomics and FACS Core Facilities, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Konstantin Neumann
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians Universität München, Munich, Germany; Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Peter Walentek
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Clemens Kreutz
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Medical Biometry and Statistics, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Ulrich Maurer
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute for Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angelika S Rambold
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Frank Edlich
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Chris Meisinger
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna Köttgen
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Genetic Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Stefan Jakobs
- Research Group Mitochondrial Structure and Dynamics, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Clinic for Neurology, University Medical Center of Göttingen, Göttingen, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy TNM, Göttingen, Germany
| | - Alexander N R Weber
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany; Clusters of Excellence EXC-2180 (iFIT) and -2124 (CMFI), University of Tübingen, Tübingen, Germany
| | - Martin Schwemmle
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Christina J Groß
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
37
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [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: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
38
|
Wells TJ, Esposito T, Henderson IR, Labzin LI. Mechanisms of antibody-dependent enhancement of infectious disease. Nat Rev Immunol 2025; 25:6-21. [PMID: 39122820 DOI: 10.1038/s41577-024-01067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 08/12/2024]
Abstract
Antibody-dependent enhancement (ADE) of infectious disease is a phenomenon whereby host antibodies increase the severity of an infection. It is well established in viral infections but ADE also has an underappreciated role during bacterial, fungal and parasitic infections. ADE can occur during both primary infections and re-infections with the same or a related pathogen; therefore, understanding the underlying mechanisms of ADE is critical for understanding the pathogenesis and progression of many infectious diseases. Here, we review the four distinct mechanisms by which antibodies increase disease severity during an infection. We discuss the most established mechanistic explanation for ADE, where cross-reactive, disease-enhancing antibodies bound to pathogens interact with Fc receptors, thereby enhancing pathogen entry or replication, ultimately increasing the total pathogen load. Additionally, we explore how some pathogenic antibodies can shield bacteria from complement-dependent killing, thereby enhancing bacterial survival. We interrogate the molecular mechanisms by which antibodies can amplify inflammation to drive severe disease, even in the absence of increased pathogen replication. We also examine emerging roles for autoantibodies in enhancing the pathogenesis of infectious diseases. Finally, we discuss how we can leverage these insights to improve vaccine design and future treatments for infectious diseases.
Collapse
Affiliation(s)
- Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Tyron Esposito
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ian R Henderson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Larisa I Labzin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
39
|
Gómez-Carballa A, Pischedda S, Pardo-Seco J, Gómez-Rial J, Martinón-Torres F, Salas A. Interferon gene expression declines over time post-COVID infection and in long COVID patients. Infect Dis (Lond) 2025; 57:35-48. [PMID: 39163143 DOI: 10.1080/23744235.2024.2389481] [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: 02/08/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Interferons (IFNs) represent a first-line defense against viruses and other pathogens. It has been shown that an impaired and uncontrolled release of these glycoproteins can result in tissue damage and explain severe progression of coronavirus disease 2019 (COVID-19). However, their potential role in Long-COVID syndrome (LC) remains debateable. OBJECTIVES The objective of the present study is to shed further light on the possible role of IFNs (and related genes) gene expression patterns in the progression of COVID-19 and LC patients. METHODS We carried out a multi-cohort study by analyzing the IFN gene expression patterns (using different IFN gene signatures) in five cohorts of acute COVID-19 (n = 541 samples) and LC patients (n = 188), and compared them to patterns observed in three autoimmune diseases (systemic lupus erythematous [n = 242], systemic sclerosis [n = 91], and Sjögren's syndrome [n = 282]). RESULTS The data show that, while the interferon signatures are strongly upregulated in severe COVID-19 patients and autoimmune diseases, it decays with the time from symptoms onset and in LC patients. Differential pathway analysis of IFN-related terms indicates an over activation in autoimmune diseases (IFN-I/II) and severe COVID-19 (IFN-I/II/III), while these pathways are mostly inactivated or downregulated in LC (IFN-I/III). By analyzing six proteomic LC datasets, we did not find evidence of a role of IFNs in this condition. CONCLUSION Our findings suggest a potential role of cytokine exhaustion mediated by IFN gene expression inactivation as a possible driver of LC.
Collapse
Affiliation(s)
- A Gómez-Carballa
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Galicia, Spain
| | - S Pischedda
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Galicia, Spain
| | - J Pardo-Seco
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Galicia, Spain
| | - J Gómez-Rial
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- TranslationalPediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Galicia, Spain
- Servicio de Inmunología, Hospital Clínico Universitario Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - F Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- TranslationalPediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Galicia, Spain
| | - A Salas
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Galicia, Spain
| |
Collapse
|
40
|
Coll RC, Schroder K. Inflammasome components as new therapeutic targets in inflammatory disease. Nat Rev Immunol 2025; 25:22-41. [PMID: 39251813 DOI: 10.1038/s41577-024-01075-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 09/11/2024]
Abstract
Inflammation drives pathology in many human diseases for which there are no disease-modifying drugs. Inflammasomes are signalling platforms that can induce pathological inflammation and tissue damage, having potential as an exciting new class of drug targets. Small-molecule inhibitors of the NLRP3 inflammasome that are now in clinical trials have demonstrated proof of concept that inflammasomes are druggable, and so drug development programmes are now focusing on other key inflammasome molecules. In this Review, we describe the potential of inflammasome components as candidate drug targets and the novel inflammasome inhibitors that are being developed. We discuss how the signalling biology of inflammasomes offers mechanistic insights for therapeutic targeting. We also discuss the major scientific and technical challenges associated with drugging these molecules during preclinical development and clinical trials.
Collapse
Affiliation(s)
- Rebecca C Coll
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK.
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
41
|
Pickering S, Wilson H, Bravo E, Perera MR, Seow J, Graham C, Almeida N, Fotopoulos L, Williams T, Moitra A, Winstone H, Nissen TAD, Galão RP, Snell LB, Doores KJ, Malim MH, Neil SJD. Antibodies to the RBD of SARS-CoV-2 spike mediate productive infection of primary human macrophages. Nat Commun 2024; 15:10764. [PMID: 39737903 PMCID: PMC11686093 DOI: 10.1038/s41467-024-54458-w] [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: 11/07/2023] [Accepted: 11/07/2024] [Indexed: 01/01/2025] Open
Abstract
The role of myeloid cells in the pathogenesis of SARS-CoV-2 is well established, in particular as drivers of cytokine production and systemic inflammation characteristic of severe COVID-19. However, the potential for myeloid cells to act as bona fide targets of productive SARS-CoV-2 infection, and the specifics of entry, remain unclear. Using a panel of anti-SARS-CoV-2 monoclonal antibodies (mAbs) we performed a detailed assessment of antibody-mediated infection of monocytes/macrophages. mAbs with the most consistent potential to mediate infection were those targeting a conserved region of the receptor binding domain (RBD; group 1/class 4). Infection was closely related to the neutralising concentration of the mAbs, with peak infection occurring below the IC50, while pre-treating cells with remdesivir or FcγRI-blocking antibodies inhibited infection. Studies performed in primary macrophages demonstrated high-level and productive infection, with infected macrophages appearing multinucleated and syncytial. Infection was not seen in the absence of antibody with the same quantity of virus. Addition of ruxolitinib significantly increased infection, indicating restraint of infection through innate immune mechanisms rather than entry. High-level production of pro-inflammatory cytokines directly correlated with macrophage infection levels. We hypothesise that infection via antibody-FcR interactions could contribute to pathogenesis in primary infection, systemic virus spread or persistent infection.
Collapse
MESH Headings
- Humans
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Macrophages/immunology
- Macrophages/virology
- Macrophages/metabolism
- SARS-CoV-2/immunology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- COVID-19/immunology
- COVID-19/virology
- Antibodies, Viral/immunology
- Nitriles/pharmacology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/pharmacology
- Pyrimidines/pharmacology
- Pyrazoles/pharmacology
- Alanine/analogs & derivatives
- Alanine/pharmacology
- Receptors, IgG/metabolism
- Receptors, IgG/immunology
- Adenosine Monophosphate/analogs & derivatives
- Adenosine Monophosphate/pharmacology
- Protein Domains
- Cells, Cultured
- Virus Internalization/drug effects
Collapse
Affiliation(s)
- Suzanne Pickering
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK.
| | - Harry Wilson
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Enrico Bravo
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Marianne R Perera
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Jeffrey Seow
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Carl Graham
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Nathalia Almeida
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Lazaros Fotopoulos
- The Stem Cell Hotel, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London, SE1 9RT, UK
| | - Thomas Williams
- The Stem Cell Hotel, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London, SE1 9RT, UK
| | - Atlanta Moitra
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Helena Winstone
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Tinne A D Nissen
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Rui Pedro Galão
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Luke B Snell
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH, UK
| | - Katie J Doores
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Michael H Malim
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Stuart J D Neil
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| |
Collapse
|
42
|
Tiniakou E, Casciola‐Rosen L, Thomas MA, Manabe Y, Antar AAR, Damarla M, Hassoun PM, Gao L, Wang Z, Zeger S, Rosen A. Autoantibodies in hospitalised patients with COVID-19. Clin Transl Immunology 2024; 13:e70019. [PMID: 39734590 PMCID: PMC11671454 DOI: 10.1002/cti2.70019] [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: 03/25/2024] [Revised: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 12/31/2024] Open
Abstract
Objectives CD209L and its homologous protein CD209 act as alternative entry receptors for the SARS-CoV-2 virus and are highly expressed in the virally targeted tissues. We tested for the presence and clinical features of autoantibodies targeting these receptors and compared these with autoantibodies known to be associated with COVID-19. Methods Using banked samples (n = 118) from Johns Hopkins patients hospitalised with COVID-19, we defined autoantibodies against CD209 and CD209L by enzyme-linked immunosorbent assay (ELISA). Clinical associations of these antibodies were compared with those of patients with anti-interferon (IFN) and anti-angiotensin-converting enzyme-2 (ACE2) autoantibodies. Results Amongst patients hospitalised with COVID-19, 19.5% (23/118) had IgM autoantibodies against CD209L and were more likely to have coronary artery disease (44% vs 19%, P = 0.03). Antibodies against CD209 were present in 5.9% (7/118); interestingly, all 7 were male (P = 0.02). In our study, the presence of either antibody was positively associated with disease severity [OR 95% confidence interval (95% CI): 1.80 (0.69-5.03)], but the association did not reach statistical significance. In contrast, 10/118 (8.5%) had IgG autoantibodies against IFNα, and 21 (17.8%) had IgM antibodies against ACE2. These patients had significantly worse prognosis (intubation or death) and prolonged hospital stays. However, when adjusting for patient characteristics on admission, only the presence of anti-ACE2 IgM remained significant [pooled common OR (95% CI), 4.14 (1.37, 12.54)]. Conclusion We describe IgM autoantibodies against CD209 and CD209L amongst patients hospitalised with COVID-19. These were not associated with disease severity. Conversely, patients with either anti-ACE2 IgM or anti-IFNα IgG antibodies had worse outcomes. Due to the small size of the study cohort, conclusions drawn should be considered cautiously.
Collapse
Affiliation(s)
- Eleni Tiniakou
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Livia Casciola‐Rosen
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mekha A Thomas
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Yuka Manabe
- Division of Infectious Diseases, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Annukka AR Antar
- Division of Infectious Diseases, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Li Gao
- Division of Allergy and Immunology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreMDUSA
| | - Zitong Wang
- Department of BiostatisticsBloomberg School of Public HealthBaltimoreMDUSA
| | - Scott Zeger
- Department of BiostatisticsBloomberg School of Public HealthBaltimoreMDUSA
| | - Antony Rosen
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
43
|
Grin PM, Baid K, de Jesus HCR, Kozarac N, Bell PA, Jiang SZ, Kappelhoff R, Butler GS, Leborgne NGF, Pan C, Pablos I, Machado Y, Vederas JC, Kim H, Benarafa C, Banerjee A, Overall CM. SARS-CoV-2 3CL pro (main protease) regulates caspase activation of gasdermin-D/E pores leading to secretion and extracellular activity of 3CL pro. Cell Rep 2024; 43:115080. [PMID: 39673710 DOI: 10.1016/j.celrep.2024.115080] [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: 06/13/2024] [Revised: 10/27/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024] Open
Abstract
SARS-CoV-2 3C-like protease (3CLpro or Mpro) cleaves the SARS-CoV-2 polyprotein and >300 intracellular host proteins to enhance viral replication. By lytic cell death following gasdermin (GSDM) pore formation in cell membranes, antiviral pyroptosis decreases 3CLpro expression and viral replication. Unexpectedly, 3CLpro and nucleocapsid proteins undergo unconventional secretion from infected cells via caspase-activated GSDMD/E pores in the absence of cell lysis. Bronchoalveolar lavage fluid of wild-type SARS-CoV-2-infected mice contains 3CLpro, which decreases in Gsdmd-/-Gsdme-/- mice. We identify new 3CLpro cut-sites in GSDMD at LQ29↓30SS, which blocks pore formation by 3CLpro cleavage at LH270↓N lying adjacent to the caspase activation site (NFLTD275↓G). Cleavage inactivation of GSDMD prevents excessive pore formation, thus countering antiviral pyroptosis and increasing 3CLpro secretion. Extracellular 3CLpro retains activity in serum, dampens platelet activation and aggregation, and inactivates antiviral interferon-λ1. Thus, in countering gasdermin pore formation and pyroptosis in SARS-CoV-2 infection, 3CLpro is secreted with extracellular pathological sequelae.
Collapse
Affiliation(s)
- Peter M Grin
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Kaushal Baid
- Vaccine and Infectious Diseases Organization, Department of Veterinary Microbiology, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada
| | - Hugo C R de Jesus
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Nedim Kozarac
- Institute for Virology and Immunology IVI, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathology, Vetsuisse Faculty, University of Bern, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, 3012 Bern, Switzerland
| | - Peter A Bell
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Steven Z Jiang
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Reinhild Kappelhoff
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Georgina S Butler
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Nathan G F Leborgne
- Institute for Virology and Immunology IVI, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathology, Vetsuisse Faculty, University of Bern, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland
| | - Christina Pan
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Isabel Pablos
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Yoan Machado
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - John C Vederas
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr. NW, Edmonton, AB T6G 2N4, Canada
| | - Hugh Kim
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Charaf Benarafa
- Department of Infectious Diseases and Pathology, Vetsuisse Faculty, University of Bern, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, 3012 Bern, Switzerland; Multidisciplinary Center for Infectious Diseases, University of Bern, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland.
| | - Arinjay Banerjee
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Vaccine and Infectious Diseases Organization, Department of Veterinary Microbiology, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada; Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Simcoe Hall, 1 King's College Cir., Toronto, ON M5S 1A8, Canada.
| | - Christopher M Overall
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Yonsei Frontier Lab, Yonsei University, 50 Yonsei-ro, Sudaemoon-ku, Seoul 03722, Republic of Korea.
| |
Collapse
|
44
|
Kenney D, O’Connell AK, Tseng AE, Turcinovic J, Sheehan ML, Nitido AD, Montanaro P, Gertje HP, Ericsson M, Connor JH, Vrbanac V, Crossland NA, Harly C, Balazs AB, Douam F. Immune Signatures of SARS-CoV-2 Infection Resolution in Human Lung Tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.583965. [PMID: 38496468 PMCID: PMC10942442 DOI: 10.1101/2024.03.08.583965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
While human autopsy samples have provided insights into pulmonary immune mechanisms associated with severe viral respiratory diseases, the mechanisms that contribute to a clinically favorable resolution of viral respiratory infections remain unclear due to the lack of proper experimental systems. Using mice co-engrafted with a genetically matched human immune system and fetal lung xenograft (fLX), we mapped the immunological events defining successful resolution of SARS-CoV-2 infection in human lung tissues. Viral infection is rapidly cleared from fLX following a peak of viral replication, histopathological manifestations of lung disease and loss of AT2 program, as reported in human COVID-19 patients. Infection resolution is associated with the activation of a limited number of hematopoietic subsets, including inflammatory monocytes and non-canonical double-negative T-cells with cytotoxic functions, which are highly enriched in viral RNA and dissipate upon infection resolution. Activation of specific human fibroblast and endothelial subsets also elicit robust antiviral and monocyte chemotaxis signatures, respectively. Notably, systemic depletion of human CD4+ cells, but not CD3+ cells, abrogates infection resolution in fLX and induces persistent infection, supporting evidence that peripheral CD4+ monocytes are important contributors to SARS-CoV-2 infection resolution in lung tissues. Collectively, our findings unravel a comprehensive picture of the immunological events defining effective resolution of SARS-CoV-2 infection in human lung tissues, revealing markedly divergent immunological trajectories between resolving and fatal COVID-19 cases.
Collapse
Affiliation(s)
- Devin Kenney
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Aoife K. O’Connell
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Anna E. Tseng
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jacquelyn Turcinovic
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Maegan L. Sheehan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Adam D. Nitido
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Paige Montanaro
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Hans P. Gertje
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Maria Ericsson
- Electron Microscopy Core Facility, Harvard Medical School, Boston, MA, USA
| | - John H. Connor
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | | | - Nicholas A. Crossland
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Christelle Harly
- Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France
- LabEx IGO ‘Immunotherapy, Graft, Oncology’, Nantes, France
- These authors contributed equally to the work
| | - Alejandro B. Balazs
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Florian Douam
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- These authors contributed equally to the work
- Lead contact
| |
Collapse
|
45
|
Nakayama EE, Shioda T. Detrimental Effects of Anti-Nucleocapsid Antibodies in SARS-CoV-2 Infection, Reinfection, and the Post-Acute Sequelae of COVID-19. Pathogens 2024; 13:1109. [PMID: 39770368 PMCID: PMC11728538 DOI: 10.3390/pathogens13121109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Antibody-dependent enhancement (ADE) is a phenomenon in which antibodies enhance subsequent viral infections rather than preventing them. Sub-optimal levels of neutralizing antibodies in individuals infected with dengue virus are known to be associated with severe disease upon reinfection with a different dengue virus serotype. For Severe Acute Respiratory Syndrome Coronavirus type-2 infection, three types of ADE have been proposed: (1) Fc receptor-dependent ADE of infection in cells expressing Fc receptors, such as macrophages by anti-spike antibodies, (2) Fc receptor-independent ADE of infection in epithelial cells by anti-spike antibodies, and (3) Fc receptor-dependent ADE of cytokine production in cells expressing Fc receptors, such as macrophages by anti-nucleocapsid antibodies. This review focuses on the Fc receptor-dependent ADE of cytokine production induced by anti-nucleocapsid antibodies, examining its potential role in severe COVID-19 during reinfection and its contribution to the post-acute sequelae of COVID-19, i.e., prolonged symptoms lasting at least three months after the acute phase of the disease. We also discuss the protective effects of recently identified anti-spike antibodies that neutralize Omicron variants.
Collapse
Affiliation(s)
| | - Tatsuo Shioda
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan;
| |
Collapse
|
46
|
Cui J, Li H, Ye D, Zhang G, Zhang Y, Yang L, Sim MM, Wood JP, Wei Y, Li Z, Wu C. Inhibiting NINJ1-dependent plasma membrane rupture protects against inflammasome-induced blood coagulation and inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.30.555561. [PMID: 37693519 PMCID: PMC10491273 DOI: 10.1101/2023.08.30.555561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Systemic blood coagulation accompanies inflammation during severe infection like sepsis and COVID. We've previously established a link between pyroptosis, a vital defense mechanism against infection, and coagulopathy. During pyroptosis, the formation of gasdermin-D (GSDMD) pores on the plasma membrane leads to the release of tissue factor (TF)-positive microvesicles (MVs) that are procoagulant. Mice lacking GSDMD release fewer TF MVs. However, the specific mechanisms leading from activation of GSDMD to MV release remain unclear. Plasma membrane rupture (PMR) in pyroptosis was recently reported to be actively mediated by the transmembrane protein Ninjurin-1 (NINJ1). Here we show that NINJ1 promotes procoagulant MV release during pyroptosis. Haploinsuffciency or glycine inhibition of NINJ1 limited the release of procoagulant MVs and inflammatory cytokines and partially protected against blood coagulation and lethality triggered by bacterial flagellin. Our findings suggest a crucial role for NINJ1-dependent PMR in inflammasome-induced blood coagulation and inflammation.
Collapse
Affiliation(s)
- Jian Cui
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Hua Li
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Dien Ye
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Guoying Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Yan Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Ling Yang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Martha M.S. Sim
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Jeremy P. Wood
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
- Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY
| | - Yinan Wei
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Zhenyu Li
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Congqing Wu
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY
| |
Collapse
|
47
|
Radovani B, Nimmerjahn F. IgG Glycosylation: Biomarker, Functional Modulator, and Structural Component. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1573-1584. [PMID: 39556784 DOI: 10.4049/jimmunol.2400447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/27/2024] [Indexed: 11/20/2024]
Abstract
The family of IgG Abs is a crucial component of adaptive immunity. Glycosylation of IgG maintains its structural integrity and modulates its effector functions. In this review, we discuss IgG glycosylation covering cell biological as well as therapeutic and disease-related aspects, focusing on the glycan structures in distinct IgG regions (Fab versus Fc). We also cover the impact of IgG glycosylation on disease modulation and therapeutic outcomes, alongside the potential for development of vaccines designed to induce Ag-specific IgG with glycoforms for optimal immune responses. Overall, we emphasize the significance of studying glycosylation to enhance our understanding of the dynamics and functional impacts of IgG glycosylation. These insights could be beneficial for advancing future research and clinical applications.
Collapse
Affiliation(s)
- Barbara Radovani
- Faculty of Biotechnology and Drug Development, University of Rijeka, Rijeka, Croatia
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Profile Center Immunomedicine, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
48
|
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
|
49
|
Allahverdiyeva S, Geyer CE, Veth J, de Vries LM, de Taeye SW, van Gils MJ, den Dunnen J, Chen H. Testosterone and estradiol reduce inflammation of human macrophages induced by anti-SARS-CoV-2 IgG. Eur J Immunol 2024; 54:e2451226. [PMID: 39246165 PMCID: PMC11628899 DOI: 10.1002/eji.202451226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
COVID-19, the disease caused by SARS-CoV-2, particularly causes severe inflammatory disease in elderly, obese, and male patients. Since both aging and obesity are associated with decreased testosterone and estradiol expression, we hypothesized that decreased hormone levels contribute to excessive inflammation in the context of COVID-19. Previously, we and others have shown that hyperinflammation in severe COVID-19 patients is induced by the production of pathogenic anti-spike IgG antibodies that activate alveolar macrophages. Therefore, we developed an in vitro assay in which we stimulated human macrophages with viral stimuli, anti-spike IgG immune complexes, and different sex hormones. Treatment with levels of testosterone reflecting young adults led to a significant reduction in TNF and IFN-γ production by human macrophages. In addition, estradiol significantly attenuated the production of a very broad panel of cytokines, including TNF, IL-1β, IL-6, IL-10, and IFN-γ. Both testosterone and estradiol reduced the expression of Fc gamma receptors IIa and III, the two main receptors responsible for anti-spike IgG-induced inflammation. Combined, these findings indicate that sex hormones reduce the inflammatory response of human alveolar macrophages to specific COVID-19-associated stimuli, thereby providing a potential immunological mechanism for the development of severe COVID-19 in both older male and female patients.
Collapse
Affiliation(s)
- Sona Allahverdiyeva
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
- Medical Microbiology and Infection PreventionAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Chiara E. Geyer
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Jennifer Veth
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Laura M. de Vries
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Steven W. de Taeye
- Medical Microbiology and Infection PreventionAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Marit J. van Gils
- Medical Microbiology and Infection PreventionAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Jeroen den Dunnen
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Hung‐Jen Chen
- Center for Experimental and Molecular MedicineAmsterdam Institute for Infection and ImmunityAmsterdam University Medical CenterAmsterdamthe Netherlands
| |
Collapse
|
50
|
Bulterys PL, Xu G, Pinsky BA, Troxell ML, Menke JR, Berry GJ, Fernandez-Pol S, Hazard FK. The Histopathologic Features of Early COVID Pneumonia in a Pediatric Patient: New Insight into the Role of Macrophages. Int J Surg Pathol 2024; 32:1595-1601. [PMID: 39435671 DOI: 10.1177/10668969241236704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
A life-threatening complication of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is acute respiratory distress syndrome. Our understanding of the pathologic changes in coronavirus disease 2019 (COVID-19) is based almost exclusively on post-mortem analyses of adults. These studies established several hallmarks of SARS-CoV-2 lung infection, including diffuse alveolar damage, microvascular thrombi, and acute bronchopneumonia. We describe a fatal example of COVID pneumonia in a 9-year-old girl who presented with fever 10 months following the diagnosis of ALK-positive anaplastic large cell lymphoma (ALCL). A chest computed tomography scan revealed left upper lobe lung consolidation and nodular airspace disease, and an initial SARS-CoV-2 nasopharyngeal swab (RT-PCR) was negative. A subsequent lung biopsy performed due to concern for relapsed ALCL demonstrated sheets of intra-alveolar and interstitial macrophages, and macrophage-rich fibrinous exudates. Immunohistochemical and in-situ hybridization stains confirmed these macrophages as the predominant SARS-CoV-2-infected cell type. Subsequent RT-PCR testing of upper and lower respiratory tract samples was positive for SARS-CoV-2 infection. Whole genome sequencing confirmed the presence of the B.1.617.2 (Delta) variant. This biopsy illustrates the histopathologic features of early COVID pneumonia in antemortem lung tissue from a pediatric patient, and establishes macrophages as a potential source of SARS-CoV-2 amplification.
Collapse
Affiliation(s)
- Philip L Bulterys
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Guangwu Xu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin A Pinsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Megan L Troxell
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua R Menke
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Florette K Hazard
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|