1
|
Zhou M, Lu Y, Tang Y, Zhang T, Xiao D, Zhang M, Zhang S, Li J, Cai X, Lin Y. A DNA-based nanorobot for targeting, hitchhiking, and regulating neutrophils to enhance sepsis therapy. Biomaterials 2025; 318:123183. [PMID: 39951831 DOI: 10.1016/j.biomaterials.2025.123183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/29/2024] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
Targeted regulation of neutrophils is an effective approach for treating neutrophil-driven inflammatory diseases, but challenges remain in minimizing off-target effects and extending drug half-life. A DNA-based nanorobot was developed to target neutrophils by using an N-acetyl Pro-Gly-Pro (Ac-PGP) peptide to specifically bind to the C-X-C motif of chemokine receptor 2 (CXCR2) on neutrophil membranes. This robot (a tetrahedral framework nucleic acid modified with Ac-PGP, APT) identified and hitchhiked neutrophils to accumulate at inflammatory sites and prolong its half-lives, whilst also was internalized to influence the neutrophil cell cycle and maturation process to regulate oxidative stress, inflammation, migration, and recruitment in both in vivo and in vitro inflammation experiments. Consequently, the tissue damage caused by sepsis was greatly reduced. This novel neutrophil-based nanorobot highlights the high precision of targeting and regulating neutrophils, and presents a potential strategy for treating multiple neutrophil-driven diseases.
Collapse
Affiliation(s)
- Mi Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yifei Lu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuanlin Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shunhao Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China; National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
2
|
Shin HE, Giannakopoulos S, Park JD, Jang HJ, Park CG, Murphy SV, Park J, Verma S, Park W. Lipid nanoparticles target neutrophils to reduce SARS-CoV-2-induced lung injury and inflammation. J Control Release 2025; 382:113736. [PMID: 40254136 DOI: 10.1016/j.jconrel.2025.113736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 04/03/2025] [Accepted: 04/12/2025] [Indexed: 04/22/2025]
Abstract
The need to understand key players driving pulmonary inflammation and fibrosis in COVID-19 patients leading to effective preventive strategies is imminent. Excessive neutrophil activation, including extracellular trap (NET) formation, is associated with severe COVID-19 and long-term sequelae. However, the clinical applications of neutrophil-targeting therapies are challenging due to short bioavailability and lack of cell-type specificity. This study presents a lipid nanoparticle (LNP) platform designed to deliver two established NET inhibitors, DNase I and Sivelestat (Siv) referred to as DPNLNPs, specifically to lung neutrophils. In vitro and in vivo experiments demonstrate that DPNLNPs preferentially accumulate in the lung neutrophils and degrade NETs as efficiently as the free DNase I and Siv. Additionally, administration of DPNLNPs in K18-hACE2 mice significantly inhibited SARS-CoV-2-induced NETs at a much lower dose than the free drugs and correlated with reduced lung and systemic inflammation, lung epithelium injury, and collagen deposition. Importantly, DPNLNP treatment only during the symptomatic phase of infection improved SARS-CoV-2 outcome revealing the complex role of NETs in COVID-19 pathogenesis. Together, this study serves as a proof-of-concept for adapting the LNP platform to deliver more than one immunomodulatory drug in a cell-specific manner to manage NET-associated complications in COVID-19 and other respiratory diseases.
Collapse
Affiliation(s)
- Ha Eun Shin
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI 96813, USA
| | - Stefanos Giannakopoulos
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI 96813, USA
| | - Joo Dong Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Hye Jung Jang
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering and Department of Intelligent Precision Healthcare Convergence, Institute for Cross-disciplinary Studies (ICS), SKKU, Suwon, Gyeonggi 16419, Republic of Korea
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI 96813, USA.
| | - Saguna Verma
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu, HI 96813, USA.
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea; Department of MetaBioHealth, ICS, SKKU, Suwon, Gyeonggi 16419, Republic of Korea.
| |
Collapse
|
3
|
Monsalve DM, Acosta-Ampudia Y, Acosta NG, Celis-Andrade M, Şahin A, Yilmaz AM, Shoenfeld Y, Ramírez-Santana C. NETosis: A key player in autoimmunity, COVID-19, and long COVID. J Transl Autoimmun 2025; 10:100280. [PMID: 40071133 PMCID: PMC11894324 DOI: 10.1016/j.jtauto.2025.100280] [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: 02/04/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
NETosis, the process through which neutrophils release neutrophil extracellular traps (NETs), has emerged as a crucial mechanism in host defense and the pathogenesis of autoimmune responses. During the SARS-CoV-2 pandemic, this process received significant attention due to the central role of neutrophil recruitment and activation in infection control. However, elevated neutrophil levels and dysregulated NET formation have been linked to coagulopathy and endothelial damage, correlating with disease severity and poor prognosis in COVID-19. Moreover, it is known that SARS-CoV-2 can induce persistent low-grade systemic inflammation, known as long COVID, although the underlying causes remain unclear. It has been increasingly acknowledged that excessive NETosis and NET generation contribute to further pathophysiological abnormalities following SARS-CoV-2 infection. This review provides an updated overview of the role of NETosis in autoimmune diseases, but also the relationship between COVID-19 and long COVID with autoimmunity (e.g., latent and overt autoimmunity, molecular mimicry, epitope spreading) and NETosis (e.g., immune responses, NET markers). Finally, we discuss potential therapeutic strategies targeting dysregulated NETosis to mitigate the severe complications of COVID-19 and long COVID.
Collapse
Affiliation(s)
- Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Nicolás Guerrero Acosta
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mariana Celis-Andrade
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Ali Şahin
- Selcuk University, Faculty of Medicine, Konya, Turkiye
| | - Ahsen Morva Yilmaz
- TUBITAK Marmara Research Center (TUBITAK-MAM), Life Sciences, Medical Biotechnology Unit, Kocaeli, Turkiye
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University, Herzelia, Israel
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
4
|
Petry J, Shoykhet M, Weiser T, Griesbaum L, Bashiri Dezfouli A, Verschoor A, Wollenberg B. SARS-CoV-2 S1 protein induces IgG-mediated platelet activation and is prevented by 1.8-cineole. Biomed Pharmacother 2025; 187:118100. [PMID: 40306177 DOI: 10.1016/j.biopha.2025.118100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/12/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025] Open
Abstract
COVID-19 patients face an increased risk of thromboembolic complications, yet the exact pathophysiological role of platelets in the disease remains unclear. Considering the multifaceted nature of COVID-19 symptoms, including platelet hyperactivation and inflammation, the development of compounds that simultaneously target both represents a promising therapeutic strategy. The monoterpene 1.8-cineole (CNL-1976) is known for its anti-inflammatory and anti-aggregatory effects. Thus, understanding the mechanism behind platelet hyperactivation and the effect of 1.8-cineole during COVID-19 is crucial when aiming for a reduction of disease severity. In this study, we investigated the mechanism of platelet activation triggered by the SARS-CoV-2 S1 spike protein (S1). Utilizing S1-coupled beads, we discovered that platelet activation and aggregation were dependent on plasma components, particularly S1-specific IgG antibodies. The formation of immune complexes through IgG binding to S1 facilitated the crosslinking of the platelet expressed FcγRIIa receptor, initiating platelet activation and aggregation, as well as formation of platelet-leukocyte aggregates (PLAs). Importantly, treatment with 1.8-cineole significantly inhibited S1-bead-induced platelet activity and PLA formation. These findings strongly suggest that antibody-mediated platelet activation via FcγRIIa directly contributes to the well-recognized prothrombotic environment during COVID-19. Moreover, our data indicate that 1.8-cineole can serve as a potential therapeutic compound, alleviating platelet-driven thromboinflammatory complications associated with COVID-19 and post-acute sequelae of SARS-CoV-2 (PASC).
Collapse
Affiliation(s)
- Julie Petry
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany
| | - Maria Shoykhet
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany
| | - Tobias Weiser
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany
| | - Lena Griesbaum
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany
| | - Ali Bashiri Dezfouli
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany; Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Department of Radiation Oncology, TUM University Hospital, Germany
| | - Admar Verschoor
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany; University of Lübeck, Department of Dermatology, University Clinic Schleswig-Holstein (UKSH), Germany
| | - Barbara Wollenberg
- Technical University of Munich, School of Medicine and Health, Department of Otorhinolaryngology, Head and Neck Surgery, TUM University Hospital, Germany.
| |
Collapse
|
5
|
Chen SH, Chen CH, Lin HC, Yeh SA, Hwang TL, Chen PJ. Drug repurposing of cyclin-dependent kinase inhibitors for neutrophilic acute respiratory distress syndrome and psoriasis. J Adv Res 2025; 72:485-500. [PMID: 39089617 DOI: 10.1016/j.jare.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Neutrophilic inflammation, characterized by dysregulated neutrophil activation, triggers a variety of inflammatory responses such as chemotactic infiltration, oxidative bursts, degranulation, neutrophil extracellular traps (NETs) formation, and delayed turnover. This type of inflammation is pivotal in the pathogenesis of acute respiratory distress syndrome (ARDS) and psoriasis. Despite current treatments, managing neutrophil-associated inflammatory symptoms remains a significant challenge. AIM OF REVIEW This review emphasizes the role of cyclin-dependent kinases (CDKs) in neutrophil activation and inflammation. It aims to highlight the therapeutic potential of repurposing CDK inhibitors to manage neutrophilic inflammation, particularly in ARDS and psoriasis. Additionally, it discusses the necessary precautions for the clinical application of these inhibitors due to potential off-target effects and the need for dose optimization. KEY SCIENTIFIC CONCEPTS OF REVIEW CDKs regulate key neutrophilic functions, including chemotactic responses, degranulation, NET formation, and apoptosis. Repurposing CDK inhibitors, originally developed for cancer treatment, shows promise in controlling neutrophilic inflammation. Clinical anticancer drugs, palbociclib and ribociclib, have demonstrated efficacy in treating neutrophilic ARDS and psoriasis by targeting off-label pathways, phosphoinositide 3-kinase (PI3K) and phosphodiesterase 4 (PDE4), respectively. While CDK inhibitors offer promising therapeutic benefits, their clinical repurposing requires careful consideration of off-target effects and dose optimization. Further exploration and clinical trials are necessary to ensure their safety and efficacy in treating inflammatory conditions.
Collapse
Affiliation(s)
- Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung 831301, Taiwan.
| | - Chun-Hong Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Hsin-Chieh Lin
- Department of Chinese Medicine, E-Da Cancer Hospital, I-Shou University, Kaohsiung 824410, Taiwan; School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Shyh-An Yeh
- Medical Physics and Informatics Laboratory of Electronic Engineering and Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 80778, Taiwan; Department of Medical Imaging and Radiological Sciences, I-Shou University, Kaohsiung 824410, Taiwan; Department of Radiation Oncology, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Tsong-Long Hwang
- Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan.
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan; Graduate Institute of Medicine, College of Medicine, I-Shou University, Kaohsiung 824410, Taiwan.
| |
Collapse
|
6
|
Filippini DFL, Jiang M, Kramer L, van der Poll T, Cremer O, Hla TTW, Retter A, Bos LDJ. Plasma H3.1 nucleosomes as biomarkers of infection, inflammation and organ failure. Crit Care 2025; 29:198. [PMID: 40390092 PMCID: PMC12090586 DOI: 10.1186/s13054-025-05415-6] [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: 02/07/2025] [Accepted: 04/11/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) are a vital part of the innate immune response, while excessive NET formation can cause tissue damage. H3.1 nucleosomes, a component of NETs, have emerged as a potential biomarker. This study aimed to evaluate H3.1 nucleosomes in critical illness, assessing their relationship with sepsis, organ failure, inflammatory subphenotypes and outcomes. METHODS The MARS cohort was used, comprising of consecutive Intensive Care Unit patients, with plasma samples collected on days 0, 2 and 4. H3.1 nucleosome concentrations were measured using the Nu.Q® NETs Immunoassay. H3.1 nucleosome concentrations were compared across sepsis presence and organ failure, both at baseline and longitudinally. The relationship between H3.1 nucleosome concentrations and clinical outcomes was investigated. RESULTS 1713 critically ill patients were included, with a total of 3671 plasma samples. Baseline H3.1 nucleosome concentrations differed between sepsis confirmed by clinical adjudication (740 ng/mL), sepsis unconfirmed by clinical adjudication (416 ng/mL) and non-sepsis (463 ng/mL, P < 0.001). H3.1 concentrations were associated with SOFA score (r = 0.40) and were higher in patients with disseminated intravascular coagulation, acute kidney injury and hyperinflammatory sepsis. H3.1 concentration was highly predictive for the need of renal replacement therapy (hazard ratio 2.00 per log10 increase), correcting for mortality. CONCLUSIONS Sepsis and organ failure were closely associated with plasma H3.1 nucleosome concentrations. While individual diagnostic performance for sepsis and organ failure remained low, H3.1 levels predicted the need for renal replacement therapy and disseminated intravascular coagulation, revealing unique insights into the innate immune response. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT01905033; IRB number 10-056C, registered June 16, 2010.
Collapse
Affiliation(s)
- Daan F L Filippini
- Department of Intensive Care Medicine, Amsterdam UMC - location AMC, University of Amsterdam, 1105AZ, Amsterdam, The Netherlands.
| | - Michael Jiang
- Department of Intensive Care Medicine, Amsterdam UMC - location AMC, University of Amsterdam, 1105AZ, Amsterdam, The Netherlands
| | - Lina Kramer
- Department of Intensive Care Medicine, Amsterdam UMC - location AMC, University of Amsterdam, 1105AZ, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Infection and Molecular Medicine (C.I.M.M.), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Division of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Olaf Cremer
- Department of Intensive Care Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Teddy Tun Win Hla
- Department of Critical Care, St Thomas' Hospital, London, UK
- Volition Diagnostics UK Limited, London, UK
| | - Andrew Retter
- Department of Critical Care, St Thomas' Hospital, London, UK
- School of Immunology and Microbial Sciences, King's College, London, UK
- Volition Diagnostics UK Limited, London, UK
| | - Lieuwe D J Bos
- Department of Intensive Care Medicine, Amsterdam UMC - location AMC, University of Amsterdam, 1105AZ, Amsterdam, The Netherlands
- Department of Pulmonology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Yeung ST, Damani-Yokota P, Thannickal SA, Bartnicki E, Bernier ED, Barnett CR, Khairallah C, Duerr R, Noval MG, Segal LN, Stapleford KA, Khanna KM. Nerve- and airway-associated interstitial macrophages mitigate SARS-CoV-2 pathogenesis via type I interferon signaling. Immunity 2025; 58:1327-1342.e5. [PMID: 40286790 DOI: 10.1016/j.immuni.2025.04.001] [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: 07/03/2024] [Revised: 12/27/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
Despite vaccines, rapidly mutating viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continue to threaten human health due to an impaired immunoregulatory pathway and a hyperactive immune response. Our understanding of the local immune mechanisms used by tissue-resident macrophages to safeguard the host from excessive inflammation during SARS-CoV-2 infection remains limited. Here, we found that nerve- and airway-associated interstitial macrophages (NAMs) are required to control mouse-adapted SARS-CoV-2 (MA-10) infection. Control mice restricted lung viral distribution and survived infection, whereas NAM depletion enhanced viral spread and inflammation and led to 100% mortality. Mechanistically, type I interferon receptor (IFNAR) signaling by NAMs was critical for limiting inflammation and viral spread, and IFNAR deficiency in CD169+ macrophages mirrored NAM-depleted outcomes and abrogated their expansion. These findings highlight the essential protective role of NAMs in regulating viral spread and inflammation, offering insights into SARS-CoV-2 pathogenesis and underscoring the importance of NAMs in mediating host immunity and disease tolerance.
Collapse
Affiliation(s)
- Stephen T Yeung
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Payal Damani-Yokota
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sara A Thannickal
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Eric Bartnicki
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Eduardo D Bernier
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Clea R Barnett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Camille Khairallah
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ralf Duerr
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Medicine, Vaccine Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maria G Noval
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Leopoldo N Segal
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kenneth A Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kamal M Khanna
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
8
|
Katayama H. Neutrophil Extracellular Traps Capturing SARS-CoV-2 in the Lung Tissue (Alveoli and Parenchyma) Cause Microthrombi - A Strategy to Eliminate SARS-CoV-2 From the Circulation as Degraded Fibrin Clots. Circ Rep 2025; 7:379-382. [PMID: 40352121 PMCID: PMC12061506 DOI: 10.1253/circrep.cr-24-0157] [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: 12/06/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 05/14/2025] Open
Abstract
Background It has been thought that neutrophil extracellular traps (NETs) and thrombosis exacerbate COVID-19, but, on the other hand, NETs are an important player in innate immunity. The precise roles of NETs and thrombosis in the course of COVID-19 have not been fully elucidated. Methods and Results The roles were investigated in the literature and a new theory was formulated. When neutrophils encounter SARS-CoV-2 in the lung tissue, they undergo NETosis and capture the virus. This capture is triggered by electrostatic interaction between histones in NETs and SARS-CoV-2; histones are highly positively charged, and viruses, including SARS-CoV-2, have a net negative charge under physiological pH. NETs that capture SARS-CoV-2 fall into alveolar capillaries through the collapsed endothelium to spare the lung tissue from the toxicity of NETs. NETs in the microvessels cause microthrombosis; positively charged histones induce the aggregation of negatively charged platelets, which leads to microthrombi. Microthrombi engulfing SARS-CoV-2 are consolidated into fibrin clots, which are eventually degraded by increased fibrinolysis and eliminated from the circulation. Conclusions This novel theory suggests that NETosis and microthrombosis are phenomena inevitably elicited in COVID-19, and in combination they are a system newly termed "NETombosis". Undegraded fibrin clots remaining in the microcirculation may be the cause of the sequelae, because they cause long-lasting circulatory failure in various organs.
Collapse
|
9
|
Tsou PS, Ali RA, Lu C, Sule G, Carmona-Rivera C, Lucotti S, Ikari Y, Wu Q, Campbell PL, Gurrea-Rubio M, Maeda K, Fox SE, Brodie WD, Mattichak MN, Foster C, Tambralli A, Yalavarthi S, Amin MA, Kmetova K, Fonseca BM, Chong E, Zuo Y, Maile MD, Imberti L, Caruso A, Caccuri F, Quaresima V, Sottini A, Kuhns DB, Fink D, Castagnoli R, Delmonte OM, Kenney H, Zhang Y, Magliocco M, Su H, Notarangelo L, Zemans RL, Mao-Draayer Y, Matei IR, Salvatore M, Lyden D, Kanthi Y, Kaplan MJ, Knight JS, Fox DA. Soluble CD13 is a potential mediator of neutrophil-induced thrombogenic inflammation in SARS-CoV-2 infection. JCI Insight 2025; 10:e184975. [PMID: 40168094 DOI: 10.1172/jci.insight.184975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 03/27/2025] [Indexed: 04/03/2025] Open
Abstract
The soluble variant of the ectopeptidase CD13 (sCD13), released from the cell surface by matrix metalloproteinase 14 (MMP14), is a potent pro-inflammatory mediator, displaying chemotactic, angiogenic, and arthritogenic properties through bradykinin receptor B1 (B1R). We revealed a link between sCD13 and amplified neutrophil-mediated inflammatory responses in SARS-CoV-2 infection. sCD13 was markedly elevated in patients with COVID-19 and correlated with disease severity and variants, ethnicity, inflammation markers, and neutrophil extracellular trap formation (NETosis). Neutrophils treated with sCD13 showed heightened NETosis and chemotaxis, which were inhibited by sCD13 receptor blockade. Meanwhile sCD13 did not induce platelet aggregation. Single-cell analysis of COVID-19 lungs revealed coexpression of CD13 and MMP14 by various cell types, and higher CD13 expression compared with controls. Neutrophils with high CD13 mRNA were enriched for genes associated with immaturity, though CD13 protein expression was lower. Histological examination of COVID-19 lungs revealed CD13-positive leukocytes trapped in vessels with fibrin thrombi. Flow cytometry verified the presence of B1R and a second sCD13 receptor, protease-activated receptor 4, on monocytes and neutrophils. These findings identify sCD13 as a potential instigator of COVID-19-associated NETosis, potentiating vascular stress and thromboembolic complications. The potent pro-inflammatory effects of sCD13 may contribute to severe COVID-19, suggesting that sCD13 and its receptors might be therapeutic targets.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Ramadan A Ali
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Chenyang Lu
- Division of Rheumatology, Department of Internal Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gautam Sule
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Serena Lucotti
- Department of Pediatrics, Weill Cornell Medical College, New York, New York, USA
| | - Yuzo Ikari
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Qi Wu
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Phillip L Campbell
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Mikel Gurrea-Rubio
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Kohei Maeda
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharon E Fox
- Department of Pathology, Louisiana State University, Health Sciences Center, New Orleans, Louisiana, USA
| | - William D Brodie
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Megan N Mattichak
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Caroline Foster
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Ajay Tambralli
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - M Asif Amin
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Katarina Kmetova
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Bruna Mazetto Fonseca
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
- School of Medical Science, University of Campinas (UNICAMP), Campinas, Brazil
| | - Emily Chong
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Yu Zuo
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael D Maile
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, Brescia, Italy
| | - Arnaldo Caruso
- Section of Microbiology, University of Brescia, Brescia, Italy
| | | | - Virginia Quaresima
- Clinical Chemistry Laboratory, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alessandra Sottini
- Clinical Chemistry Laboratory, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Douglas B Kuhns
- Leidos Biomedical Research, Inc.; Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Danielle Fink
- Leidos Biomedical Research, Inc.; Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Riccardo Castagnoli
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Ottavia M Delmonte
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Heather Kenney
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Yu Zhang
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Mary Magliocco
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Helen Su
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Luigi Notarangelo
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Rachel L Zemans
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine; and Program in Cellular and Molecular Biology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yang Mao-Draayer
- Multiple Sclerosis Center of Excellence, Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Irina R Matei
- Department of Pediatrics, Weill Cornell Medical College, New York, New York, USA
| | - Mirella Salvatore
- Joan and Sanford I. Weill Department of Medicine and Department of Population Health Sciences, Weill Cornell Medical College, New York, New York, USA
| | - David Lyden
- Department of Pediatrics, Weill Cornell Medical College, New York, New York, USA
| | - Yogendra Kanthi
- Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - David A Fox
- Division of Rheumatology, Department of Internal Medicine, and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
10
|
Kaiser R, Gold C, Stark K. Recent Advances in Immunothrombosis and Thromboinflammation. Thromb Haemost 2025. [PMID: 40311639 DOI: 10.1055/a-2523-1821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Inflammation and thrombosis are traditionally considered two separate entities of acute host responses to barrier breaks. While inciting inflammatory responses is a prerequisite to fighting invading pathogens and subsequent restoration of tissue homeostasis, thrombus formation is a crucial step of the hemostatic response to prevent blood loss following vascular injury. Though originally designed to protect the host, excessive induction of either inflammatory signaling or thrombus formation and their reciprocal activation contribute to a plethora of disorders, including cardiovascular, autoimmune, and malignant diseases. In this state-of-the-art review, we summarize recent insights into the intricate interplay of inflammation and thrombosis. We focus on the protective aspects of immunothrombosis as well as evidence of detrimental sequelae of thromboinflammation, specifically regarding recent studies that elucidate its pathophysiology beyond coronavirus disease 2019 (COVID-19). We introduce recently identified molecular aspects of key cellular players like neutrophils, monocytes, and platelets that contribute to both immunothrombosis and thromboinflammation. Further, we describe the underlying mechanisms of activation involving circulating plasma proteins and immune complexes. We then illustrate how these factors skew the inflammatory state toward detrimental thromboinflammation across cardiovascular as well as septic and autoimmune inflammatory diseases. Finally, we discuss how the advent of new technologies and the integration with clinical data have been used to investigate the mechanisms and signaling cascades underlying immunothrombosis and thromboinflammation. This review highlights open questions that will need to be addressed by the field to translate our mechanistic understanding into clinically meaningful therapeutic targeting.
Collapse
Affiliation(s)
- Rainer Kaiser
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christoph Gold
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
11
|
Åkesson P, Mellhammar L, Rasmussen M, Inghammar M, Jesperson S, Månsson F, Economou Lundeberg E, Walles J, Wallberg M, Frigyesi A, Linder A. Aerosolized Dornase Alfa (DNase I) for the Treatment of Severe Respiratory Failure in COVID-19: A Randomized Controlled Trial. Open Forum Infect Dis 2025; 12:ofaf246. [PMID: 40365079 PMCID: PMC12069806 DOI: 10.1093/ofid/ofaf246] [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: 12/18/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background Lung injury in COVID-19 is characterized by neutrophil invasion and the release of neutrophil extracellular traps (NETs). An aberrant NET formation may induce local inflammation and increase sputum viscosity. Inhalation of DNase I (dornase alfa) is a treatment option that degrades NETs in the airways. Previous case series have indicated positive clinical effects of inhaled dornase alfa. Methods Patients admitted to the hospital with acute COVID-19 and hypoxia (oxygen saturation <90%) were randomly assigned to receive aerosolized dornase alfa twice daily for 5 days or a placebo in addition to standard of care. The primary outcome was discharge from the hospital or an oxygen saturation >93% without respiratory support. Results In total, 76 patients were randomized. The study was stopped when the Omicron virus variant appeared. The clinical response rate did not differ between patients receiving the active substance and placebo. Secondary outcomes were similar across groups, such as mortality, a new episode of hypoxia, length of stay in the hospital, and adverse events. A subanalysis of patients older or younger than 65 years showed no differences in primary or secondary outcomes. Conclusions Aerosolized dornase alfa failed to improve hypoxia in hospitalized patients with acute COVID-19. The study was conducted during a time of heterogeneity in viral variants and vaccination status of participants. Whether dornase alfa affects the outcomes in other respiratory infections requires further study.
Collapse
Affiliation(s)
- Per Åkesson
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Infectious Diseases, University Hospital, Lund, Sweden
| | - Lisa Mellhammar
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Infectious Diseases, University Hospital, Lund, Sweden
| | - Magnus Rasmussen
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Infectious Diseases, University Hospital, Lund, Sweden
| | - Malin Inghammar
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Infectious Diseases, University Hospital, Lund, Sweden
| | - Sara Jesperson
- Clinical Studies Sweden–Forum South, Skåne University Hospital, Lund, Sweden
| | - Fredrik Månsson
- Department of Infectious Diseases, Skåne University Hospital, Malmö, Sweden
| | | | - John Walles
- Department of Infectious Diseases, Central Hospital of Kristianstad, Kristianstad, Sweden
- Clinical Infection Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Martin Wallberg
- Department of Pulmonary Diseases, Skåne University Hospital, Lund, Sweden
| | - Attila Frigyesi
- Department of Anesthesiology and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Adam Linder
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
12
|
Petito E, Guglielmini G, De Robertis E, Becattini C, Franco L, Falcinelli E, Conti C, Gori F, Vaudo G, Cerotto V, Paliani U, Mezzasoma L, Camilloni B, Gresele P. Platelets from COVID-19 Patients Show an Altered Nitric Oxide/Reactive Oxygen Species Production Balance. Thromb Haemost 2025. [PMID: 40306665 DOI: 10.1055/a-2562-4516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Oxidative stress has been associated with COVID-19-related thrombotic complications. No investigations have explored nitric oxide (NO) and radical oxygen species (ROS) production by platelets. Indeed, activated platelets generate both NO and ROS which in turn regulate platelet function. The aim of the present study was to measure platelet NO and ROS production in COVID-19 patients, to assess whether they correlate with disease outcome and to clarify the mechanisms of platelet NO/ROS imbalance in COVID-19.Hospitalized mild and severe COVID-19 patients, age- and sex-matched healthy controls, and patients hospitalized in intensive care units for reasons different from COVID-19 were enrolled. Platelet NO and ROS production was assessed by flow cytometry. The oxidant and antioxidant capacity of COVID-19 plasma was assessed using lipid peroxidation and ORAC assays. The effect of COVID-19 plasma on platelet NO production and the impact of antioxidants on it were studied by flow cytometry.Platelets from COVID-19 patients displayed an altered NO/ROS balance, with defective NO and increased ROS production. Platelet NO production was significantly lower in patients who developed thrombotic events during hospitalization. COVID-19 patients showed significantly increased plasma lipid peroxidation and reduced antioxidant capacity compared with healthy controls. Concordantly, plasma from COVID-19 patients impaired NO production by healthy control species platelets, which was restored by the antioxidant agent Hydroxy-TEMPO.Our findings suggest that the unbalanced platelet NO/ROS production in COVID-19 plays a role in the thrombotic complications of SARS-CoV-2 infection. The restoration of platelet NO production may represent a therapeutic target for the prevention of thrombotic events in COVID-19 patients.
Collapse
Affiliation(s)
- Eleonora Petito
- Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Giuseppe Guglielmini
- Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Edoardo De Robertis
- Division of Anaesthesia, Analgesia, and Intensive Care, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cecilia Becattini
- Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Laura Franco
- Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Emanuela Falcinelli
- Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Chiara Conti
- Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Fabio Gori
- Section of Anesthesia, Intensive Care, and Pain Medicine, Azienda Ospedaliera-Universitaria Santa Maria della Misericordia, Perugia, Italy
| | - Gaetano Vaudo
- Unit of Internal Medicine, Terni University Hospital, Terni, Italy
| | - Vittorio Cerotto
- Section of Anesthesia, Intensive Care and Pain Medicine, Department of Emergency and Urgency, Città di Castello Hospital, Città di Castello, Italy
| | - Ugo Paliani
- Division of Internal Medicine, ASL 1 Umbria, Città di Castello, Italy
| | - Letizia Mezzasoma
- Department of Medicine and Surgery, Section of Biosciences and Medical Embryology, University of Perugia, Perugia, Italy
| | - Barbara Camilloni
- Department of Medicine and Surgery, Microbiology and Clinical Microbiology, University of Perugia, Perugia, Italy
| | - Paolo Gresele
- Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
13
|
Jesus Gonzalez-Contreras FD, Gutierrez-Vidal RG, Zarate X. A recombinant human SLPI variant suppresses the formation of neutrophil extracellular traps at low concentrations in vitro. Protein Expr Purif 2025; 232:106721. [PMID: 40280458 DOI: 10.1016/j.pep.2025.106721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/10/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Neutrophil extracellular traps (NETs) are web-like structures released by neutrophils to trap and kill microbes. While NETs are crucial in host defense, excessive formation can lead to autoimmune diseases. Currently, no specific drugs target NETs directly; however, some medications can indirectly modulate their formation. The secretory leukocyte protease inhibitor (SLPI) is a small protein that inhibits the activity of neutrophil elastase (NE), an enzyme essential for NETs formation. By inhibiting NE activity, SLPI prevents the chromatin from decondensing, which is necessary for NETs to form; this suggests that SLPI may protect by preventing excessive NETs development. However, evidence indicates that NE can inactivate SLPI by cleaving its N-terminus. This action can create a protease/antiprotease imbalance, potentially leading to detrimental consequences for the host. In this study, we produced a recombinant variant of SLPI (SLPI-S15G-A16G: rSLPIv) using recombinant DNA technology, expressed in Escherichia coli SHuffle T7. The protein was tagged with the small metal-binding protein (SmbP) to facilitate its expression and purification through immobilized metal-affinity chromatography. Our results demonstrated that rSLPIv exhibited immunomodulatory activity at a concentration of 10 nM in neutrophils that had been prestimulated with PMA. It reduced NETs formation by 30 % and maintained this effect for up to 6 h. Confocal microscopy confirmed these findings, revealing a rSLPIv-dependent reduction in neutrophil nuclear expansion. Thus, rSLPIv shows significant suppressive activity on NETs formation at low concentrations, making it a potential candidate as an immunotherapeutic agent.
Collapse
Affiliation(s)
- Felipe de Jesus Gonzalez-Contreras
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n, Ciudad Universitaria, San Nicolas de los Garza, NL, 66455, Mexico
| | - Roxana Guadalupe Gutierrez-Vidal
- Programa de Investigadoras e Investigadores por México CONAHCYT, Mexico; Centro de Investigacion y de Estudios Avanzados del Instituto Politecnico Nacional, Unidad Monterrey, Via del Conocimiento 201, Parque de Investigacion e Innovacion Tecnologica, Apodaca, NL, 66600, Mexico
| | - Xristo Zarate
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n, Ciudad Universitaria, San Nicolas de los Garza, NL, 66455, Mexico.
| |
Collapse
|
14
|
Cui H, Huang X. Multi-omics integration reveals YWHAE as a key mediator of ferroptosis in ARDS. Funct Integr Genomics 2025; 25:94. [PMID: 40261442 DOI: 10.1007/s10142-025-01603-3] [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: 02/25/2025] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening condition characterized by severe hypoxemia and high mortality. Ferroptosis, a form of regulated cell death driven by iron accumulation and lipid peroxidation, has emerged as a critical mechanism in ARDS pathogenesis. However, the molecular regulators of ferroptosis in ARDS remain unclear. This study integrates multi-omics analysis and experimental validation to identify ferroptosis-related targets in ARDS. Bronchoalveolar lavage fluid (BALF) samples from ARDS patients and healthy controls were subjected to proteomics and metabolomics analysis. Transcriptomic data from the GSE243066 dataset and ferroptosis-related gene databases were integrated to identify key genes. Functional enrichment analyses were performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. An LPS-induced ARDS mouse model was established for experimental validation, including Western blotting, histopathology, and ferroptosis-related biochemical assays. Multi-omics analysis identified YWHAE as a ferroptosis-associated gene significantly upregulated in ARDS. Functional enrichment revealed key pathways, including ferroptosis, hypoxia-inducible factor-1 signaling, and oxidative stress responses. Proteomic and transcriptomic integration highlighted 51 overlapping differentially expressed genes, with YWHAE emerging as a central hub in the protein-protein interaction network. Metabolomics analysis further revealed glutathione and cysteine metabolism as critical pathways linked to ferroptosis. In the ARDS mouse model, ferroptosis inhibitor ferrostatin-1 (Fer-1) attenuated LPS-induced lung injury, reduced oxidative stress markers, and downregulated YWHAE expression. This study identifies YWHAE as a novel ferroptosis-related target in ARDS through multi-omics analysis and experimental validation. These findings provide new insights into the molecular mechanisms of ferroptosis in ARDS and highlight YWHAE as a potential therapeutic target for future interventions.
Collapse
Affiliation(s)
- Honghui Cui
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Graduate School of Youjiang, Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Xia Huang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Life Science and Clinical Medicine Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| |
Collapse
|
15
|
Xiong Y, Wang J, Shang X, Chen T, Fraser DD, Fonseca GJ, Rousseau S, Ding J. Efficient and scalable construction of clinical variable networks for complex diseases with RAMEN. CELL REPORTS METHODS 2025; 5:101022. [PMID: 40215965 DOI: 10.1016/j.crmeth.2025.101022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 12/09/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Understanding the interplay among clinical variables-such as demographics, symptoms, and laboratory results-and their relationships with disease outcomes is critical for advancing diagnostics and understanding mechanisms in complex diseases. Existing methods fail to capture indirect or directional relationships, while existing Bayesian network learning methods are computationally expensive and only infer general associations without focusing on disease outcomes. Here we introduce random walk- and genetic algorithm-based network inference (RAMEN), a method for Bayesian network inference that uses absorbing random walks to prioritize outcome-relevant variables and a genetic algorithm for efficient network refinement. Applied to COVID-19 (Biobanque québécoise de la COVID-19), intensive care unit (ICU) septicemia (MIMIC-III), and COPD (CanCOLD) datasets, RAMEN reconstructs networks linking clinical markers to disease outcomes, such as elevated lactate levels in ICU patients. RAMEN demonstrates advantages in computational efficiency and scalability compared to existing methods. By modeling outcome-specific relationships, RAMEN provides a robust tool for uncovering critical disease mechanisms, advancing diagnostics, and enabling personalized treatment strategies.
Collapse
Affiliation(s)
- Yiwei Xiong
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Jingtao Wang
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Xiaoxiao Shang
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Mathematics and Statistics, McGill University, 805 Sherbrooke Street West, Montreal, QC H3A 0B9, Canada
| | - Tingting Chen
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Hematology Department, Beijing Luhe Hospital, Capital Medical University, Xinhua South Road No. 82, Tongzhou District, Beijing 101149, China
| | - Douglas D Fraser
- Children's Health Research Institute, Victoria Research Laboratories, 800 Commissioners Road East, London, ON N6C 2V5, Canada; Lawson Health Research Institute, London, ON N6C 2R5, Canada; Department of Pediatrics, Western University, London, ON N6A 5C1, Canada; Department of Physiology & Pharmacology, Western University, London, ON N6A 5C1, Canada; Department of Clinical Neurological Sciences, Western University, London, ON N6A 5C1, Canada
| | - Gregory J Fonseca
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada.
| | - Simon Rousseau
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada.
| | - Jun Ding
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; School of Computer Science, McGill University, 3480 Rue University, Montreal, QC H3A 2A7, Canada; Mila-Quebec AI Institute, 6666 Rue Saint-Urbain, Montreal, QC H2S 3H1, Canada.
| |
Collapse
|
16
|
Nie J, Zhou L, Tian W, Liu X, Yang L, Yang X, Zhang Y, Wei S, Wang DW, Wei J. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther 2025; 10:112. [PMID: 40234407 PMCID: PMC12000524 DOI: 10.1038/s41392-025-02178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 04/17/2025] Open
Abstract
Cytokine storm (CS) is a severe systemic inflammatory syndrome characterized by the excessive activation of immune cells and a significant increase in circulating levels of cytokines. This pathological process is implicated in the development of life-threatening conditions such as fulminant myocarditis (FM), acute respiratory distress syndrome (ARDS), primary or secondary hemophagocytic lymphohistiocytosis (HLH), cytokine release syndrome (CRS) associated with chimeric antigen receptor-modified T (CAR-T) therapy, and grade III to IV acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. The significant involvement of the JAK-STAT pathway, Toll-like receptors, neutrophil extracellular traps, NLRP3 inflammasome, and other signaling pathways has been recognized in the pathogenesis of CS. Therapies targeting these pathways have been developed or are currently being investigated. While novel drugs have demonstrated promising therapeutic efficacy in mitigating CS, the overall mortality rate of CS resulting from underlying diseases remains high. In the clinical setting, the management of CS typically necessitates a multidisciplinary team strategy encompassing the removal of abnormal inflammatory or immune system activation, the preservation of vital organ function, the treatment of the underlying disease, and the provision of life supportive therapy. This review provides a comprehensive overview of the key signaling pathways and associated cytokines implicated in CS, elucidates the impact of dysregulated immune cell activation, and delineates the resultant organ injury associated with CS. In addition, we offer insights and current literature on the management of CS in cases of FM, ARDS, systemic inflammatory response syndrome, treatment-induced CRS, HLH, and other related conditions.
Collapse
Grants
- 82070217, 81873427 National Natural Science Foundation of China (National Science Foundation of China)
- 82100401 National Natural Science Foundation of China (National Science Foundation of China)
- 81772477, 81201848, 82473220 National Natural Science Foundation of China (National Science Foundation of China)
- 82330010,81630010,81790624 National Natural Science Foundation of China (National Science Foundation of China)
- National High Technology Research and Development Program of China, Grant number: 2021YFA1101500.
- The Hubei Provincial Natural Science Foundation (No.2024AFB050)
- Project of Shanxi Bethune Hospital, Grant Numbber: 2023xg02); Fundamental Research Program of Shanxi Province, Grant Numbber: 202303021211224
- The Key Scientific Research Project of COVID-19 Infection Emergency Treatment of Shanxi Bethune Hospital (2023xg01), 2023 COVID-19 Research Project of Shanxi Provincial Health Commission (No.2023XG001, No. 2023XG005), Four “Batches” Innovation Project of Invigorating Medical through Science and Technology of Shanxi Province (2023XM003), Cancer special Fund research project of Shanxi Bethune Hospital (No. 2020-ZL04), and External Expert Workshop Fund Program of Shanxi Provincial Health Commission(Proteomics Shanxi studio for Huanghe professor)
- Fundamental Research Program of Shanxi Province(No.202303021221192); 2023 COVID-19 Emergency Project of Shanxi Health Commission (Nos.2023XG001,2023XG005)
Collapse
Affiliation(s)
- Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
| | - Weiwei Tian
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liping Yang
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
17
|
Hendricks AL, More KR, Devaraj A, Buzzo JR, Robledo-Avila FH, Partida-Sanchez S, Bakaletz LO, Goodman SD. Bacterial biofilm-derived H-NS protein acts as a defense against Neutrophil Extracellular Traps (NETs). NPJ Biofilms Microbiomes 2025; 11:58. [PMID: 40234459 PMCID: PMC12000423 DOI: 10.1038/s41522-025-00691-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
Extracellular DNA (eDNA) is crucial for the structural integrity of bacterial biofilms as they undergo transformation from B-DNA to Z-DNA as the biofilm matures. This transition to Z-DNA increases biofilm rigidity and prevents binding by canonical B-DNA-binding proteins, including nucleases. One of the primary defenses against bacterial infections are Neutrophil Extracellular Traps (NETs), wherein neutrophils release their own eDNA to trap and kill bacteria. Here we show that H-NS, a bacterial nucleoid associated protein (NAP) that is also released during biofilm development, is able to incapacitate NETs. Indeed, when exposed to human derived neutrophils, H-NS prevented the formation of NETs and lead to NET eDNA retraction in previously formed NETs. NETs that were exposed to H-NS also lost their ability to kill free-living bacteria which made H-NS an attractive therapeutic candidate for the control of NET-related human diseases. A model of H-NS release from biofilms and NET incapacitation is discussed.
Collapse
Affiliation(s)
- A L Hendricks
- Center for Microbe and Immunity Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - K R More
- Center for Microbe and Immunity Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - A Devaraj
- Center for Microbe and Immunity Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - J R Buzzo
- Center for Microbe and Immunity Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - F H Robledo-Avila
- Center for Microbe and Immunity Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - S Partida-Sanchez
- Center for Microbe and Immunity Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - L O Bakaletz
- Center for Microbe and Immunity Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - S D Goodman
- Center for Microbe and Immunity Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatrics, College of Medicine, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
18
|
Chen Y, Mao R, Chang Q, Yuan Y, Zhang H, Li F. A causal effects of neutrophil extracellular traps and its biomarkers on acute respiratory distress syndrome: a two-sample Mendelian randomization study. Sci Rep 2025; 15:11995. [PMID: 40199908 PMCID: PMC11978891 DOI: 10.1038/s41598-025-95676-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
Previous studies have indicated an association between neutrophil extracellular traps (NETs) and acute respiratory distress syndrome (ARDS). This study aimed to investigate the potential causal effects of NETs and NETs-related biomarkers on ARDS or vice-versa. A two-sample Mendelian randomization (MR) utilizing genome-wide association studies (GWAS) data was employed to analyze the causality. The primary analysis was conducted using inverse-variance weighted (IVW) methods; weighted median, MR-Egger, and weighted model methods were used to validate the results. Horizontal pleiotropy and outlier detection were assessed via MR-Egger and MR pleiotropy residual sum and outlier (MR-PRESSO), respectively; Cochran's Q test evaluated heterogeneity, while Leave-one-out analyses were used to evaluate the presence of predominant instrumental variables (IVs). IVW method suggested causal associations between genetically predicted IL-13 and a higher risk of ARDS [OR (95%CI) = 1.52 (1.03-2.23), P = 0.047], while there was no causal effect of other factors on ARDS (all P > 0.05). Also, ARDS had no effect on NETs and NETs-related biomarkers (all P > 0.05). Cochran's Q confirmed no significant heterogeneity. MR-Egger regression ruled out horizontal pleiotropy's influence, and MR-PRESSO analysis identified no outliers, reinforcing the study's findings. This MR study established a causal relationship between IL-13 and ARDS, suggesting its potential role as a therapeutic target and biomarker of ARDS. Future work should delve into the underlying mechanisms and clinical applications.
Collapse
Affiliation(s)
- Yuqing Chen
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Ruolin Mao
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qing Chang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yueyang Yuan
- School of Mechanical and Electrical Engineering, Hu Nan City University, Yiyang, 413099, China
| | - Hai Zhang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Feng Li
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| |
Collapse
|
19
|
Jia R, Wan L, Jin L, Tian Q, Chen Y, Zhu X, Zhang M, Zhang Y, Zong L, Wu X, Miao C, Cai Y, Ma J, Hu L, Liu WT. Fucoidan reduces NET accumulation and alleviates chemotherapy-induced peripheral neuropathy via the gut-blood-DRG axis. J Neuroinflammation 2025; 22:100. [PMID: 40186245 PMCID: PMC11969723 DOI: 10.1186/s12974-025-03431-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy (CIPN) is a serious adverse reaction to chemotherapy with limited treatment options. Research has indicated that neutrophil extracellular traps (NETs) are critical for the pathogenesis of CIPN. LPS/HMGB1 serve as important inducers of NETs. Here, we aimed to target the inhibition of NET formation (NETosis) to alleviate CIPN. METHODS Oxaliplatin (L-OHP) was used to establish a CIPN model. The mice were pretreated with fucoidan to investigate the therapeutic effect. SR-A1-/- mice were used to examine the role of scavenger receptor A1 (SR-A1) in CIPN. Bone marrow-derived macrophages (BMDMs) isolated from SR-A1-/- mice and WT mice were used to investigate the mechanism by which macrophage phagocytosis of NETs alleviates CIPN. RESULTS Clinically, we found that the contents of LPS, HMGB1 and NETs in the plasma of CIPN patients were significantly increased and positively correlated with the VAS score. Fucoidan decreased the LPS/HMGB1/NET contents and relieved CIPN in mice. Mechanistically, fucoidan upregulated SR-A1 expression and promoted the phagocytosis of LPS/HMGB1 by BMDMs. Fucoidan also facilitated the engulfment of NETs by BMDMs via the recognition and localization of SR-A1 and HMGB1. The therapeutic effects of fucoidan were abolished by SR-A1 knockout. RNA-seq analysis revealed that fucoidan increased sqstm1 (p62) gene expression. Fucoidan promoted the competitive binding of sqstm1 and Nrf2 to Keap1, increasing Nrf2 nuclear translocation and SR-A1 transcription. Additionally, the sequencing analysis (16 S) of microbial diversity revealed that fucoidan increased the gut microbiota diversity and abundance and increased the Bacteroides/Firmicutes ratio. CONCLUSIONS Altogether, fucoidan promotes the SR-A1-mediated phagocytosis of LPS/HMGB1/NETs and maintains gut microbial homeostasis, which may provide a potential therapeutic strategy for CIPN.
Collapse
Affiliation(s)
- Rumeng Jia
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Li Wan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Lai Jin
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Qingyan Tian
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yongyi Chen
- Department of Anesthesiology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, 210009, China
| | - Xia Zhu
- Department of Oncology, Lianyungang Municipal Oriental Hospital, Lianyungang, Jiangsu, 222042, China
| | - Mengyao Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yajie Zhang
- Central Laboratory, Department of Biobank, Nanjing Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210022, China
| | - Lijuan Zong
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Chen Miao
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yihang Cai
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jianxin Ma
- Department of Oncology, Lianyungang Municipal Oriental Hospital, Lianyungang, Jiangsu, 222042, China.
| | - Liang Hu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Wen-Tao Liu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
20
|
Rolling CC, Lewirt S, Beitzen-Heineke A, Beckmann L, Bokemeyer C, Alsdorf W, Voigtlaender M, Langer F. Checkpoint Inhibitors, CAR T Cells, and the Hemostatic System: What Do We Know So Far? Hamostaseologie 2025; 45:175-187. [PMID: 40334710 DOI: 10.1055/a-2528-5071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells are novel therapeutic strategies that enhance anticancer immunity by activating or engineering cancer-targeting T cells. The resulting hyperinflammation carries several side effects, ranging from autoimmune-like symptoms to cytokine release syndrome (CRS), with potentially severe consequences. Recent findings indicate that ICIs increase the risk of venous and arterial thromboembolic adverse events. Patients with prior VTE might be at higher risk of developing new events under ICI while other risk factors vary across studies. So far, data on CAR T-linked coagulopathies are limited. Hypofibrinogenemia in the presence of CRS is the most commonly observed dysregulation of hemostatic parameters. A rare but particularly severe adverse event is the development of disseminated intravascular coagulation activation, which can occur in the setting of CRS and may be linked to immune effector cell-associated hemophagocytic lymphohistiocytosis. While the increasing number of studies on thromboembolic complications and coagulation alterations under ICIs and CAR T therapies are concerning, these results might be influenced by the retrospective study design and the heterogeneous patient populations. Importantly, numerous promising new T cell-based immunotherapies are currently under investigation for various cancers and are expected to become very prominent therapy options in the near future. Therefore, coagulopathies and thrombosis under T cell-directed immuno- and anti-cancer therapies is important. Our review provides an overview of the current understanding of ICI- and CAR T-associated thromboembolism. We discuss pathogenic mechanisms of inflammation-associated coagulation activation and explore potential biomarkers for VTE.
Collapse
Affiliation(s)
- Christina C Rolling
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Lewirt
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Beitzen-Heineke
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, United States
| | - Lennart Beckmann
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Minna Voigtlaender
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Langer
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
Mattone M, Masci GM, Landini N, Zingaropoli MA, Catalano C, Ciardi MR, Panebianco V. MMP-9 metalloproteinase and its regulator are not associated with mid-term CT residual abnormalities in patients with COVID-19 pneumonia. Acta Radiol Open 2025; 14:20584601251330563. [PMID: 40291835 PMCID: PMC12033756 DOI: 10.1177/20584601251330563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
Background COVID-19 patients may have residual pulmonary alterations after the acute disease, with fibrotic-like alterations. Since metalloproteinases (MMP) and their regulators may be involved in inflammation and abnormal repair processing, we aimed to investigate the correlations between MMP-9, a tissue inhibitor of metalloproteinases (TIMP-1) and chest CT abnormalities in acute phase and mid-term follow-up. Methods COVID-19 patients with plasma analyses and CT scans performed at acute onset and 3 months after discharge (T post) were evaluated. MMP-9, TIMP-1, and MMP-9/TIMP-1 ratio were analyzed. CT extents of COVID-19 pneumonia and fibrotic-like alterations were visually scored (score range 0-25). Spearman rank correlation analysis (p-value <.05) was computed between acute and mid-term plasma analyses and CT scores. Results 39 patients were enrolled. At hospital admission, MMP-9, TIMP-1, and MMP-9/TIMP-1 had a median of 240.5 ng/mL, 258.8 ng/mL, and 0.9. The median CT global and fibrotic-like scores were 9 and 6. At T post, MMP-9 and TIMP-1 were not statistically different (p-value <.05). There was a reduction of CT global score (p-value = .00007). A significant correlation was found between MMP-9 and CT global score at hospital admission (ρ = 0.456, p-value = .003) and between MMP-9/TIMP-1 ratio and CT global score at hospital admission (ρ = 0.406, p-value = .009). No other significant correlations were found between plasma enzymes and CT alterations, both in acute and mid-term follow-up. Conclusion MMP-9 plasma levels and MMP-9/TIMP-1 ratio correlate with lung involvement during the acute phase. None of the levels of MMP-9, TIMP-1, and MMP-9/TIMP-1 ratio may be adopted as predictors of residual pulmonary alterations in mid-term follow-up.
Collapse
Affiliation(s)
- Monica Mattone
- Department of Radiological Sciences, Oncology and Pathology, Policlinico Umberto I, “Sapienza” University, Rome, Italy
| | - Giorgio Maria Masci
- Department of Radiological Sciences, Oncology and Pathology, Policlinico Umberto I, “Sapienza” University, Rome, Italy
| | - Nicholas Landini
- Department of Radiological Sciences, Oncology and Pathology, Policlinico Umberto I, “Sapienza” University, Rome, Italy
| | - Maria Antonella Zingaropoli
- Department of Public Health and Infectious Diseases, Policlinico Umberto I, “Sapienza” University, Rome, Italy
| | - Carlo Catalano
- Department of Radiological Sciences, Oncology and Pathology, Policlinico Umberto I, “Sapienza” University, Rome, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Policlinico Umberto I, “Sapienza” University, Rome, Italy
| | - Valeria Panebianco
- Department of Radiological Sciences, Oncology and Pathology, Policlinico Umberto I, “Sapienza” University, Rome, Italy
| |
Collapse
|
22
|
Marques-Ferreira G, Lourenço AA, Campi-Azevedo AC, Clarindo FA, Bernardes AFL, Lamounier LO, Guimaraes NR, Adelino TER, de Melo Iani FC, Santos DA, Magalhães VCR, da Mata CPSM, Reis EVDS, Moraes TDFS, Gomes-de-Pontes L, da Fonseca FG, Teixeira-Carvalho A, Menegueti MG, Auxiliadora-Martins M, Amaral PHR, Pérez JCG, Martins-Filho OA, Coelho-Dos-Reis JG. Unique signatures of airway and systemic immunity in severe COVID-19 patients infected with alpha to Omicron SARS-CoV-2 variants of concern. Inflamm Res 2025; 74:57. [PMID: 40153054 DOI: 10.1007/s00011-025-02018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/30/2025] Open
Abstract
In this study, systemic and localized immunity induced by SARS-CoV-2 variants of concern or interest (VOC/VOI) was investigated. For that, serum and tracheal aspirate soluble chemokines, pro-inflammatory/regulatory cytokines, and growth factors were measured in severe COVID-19 patients under mechanical ventilation upon infection with different SARS-CoV-2 variants, namely Alpha, Gamma, Zeta, Delta and Omicron. Increased levels of soluble mediators were observed in serum from severe COVID-19 patients regardless of the variant. In tracheal aspirate samples, the patients infected with the Gamma, Zeta, Delta, and Omicron variants exhibited reduced levels of inflammatory cytokines when compared to those infected with the Alpha variant. The trend of lower cytokine levels was also observed in the serum of patients across these variants, except for the Delta variant. By using network analysis and cytokine storm signatures, the data confirmed that severe COVID-19 induced by different variants have a completely divergent pattern of connectivity in serum samples as well as tracheal aspirates. Patients infected with variants at later time points in the pandemic such as Omicron exhibited networks of weak central architecture in serum samples as compared to tracheal aspirates, with lower number of neighborhood connections and clusters of pro-inflammatory and regulatory cytokines. By and large, this study points out to important systemic and local divergences and to loss of airway localized immunity in severe COVID-19 patients infected with SARS-CoV-2 variants, which brings insight into understanding host responses and viral escape vis-à-vis the virus mutations and evolution.
Collapse
Affiliation(s)
- Geovane Marques-Ferreira
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Alice Aparecida Lourenço
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | - Felipe Alves Clarindo
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - André Felipe Leal Bernardes
- Laboratório Central de Saúde Pública de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Ludmila Oliveira Lamounier
- Laboratório Central de Saúde Pública de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Natalia Rocha Guimaraes
- Laboratório Central de Saúde Pública de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Talita Emile Ribeiro Adelino
- Laboratório Central de Saúde Pública de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Felipe Campos de Melo Iani
- Laboratório Central de Saúde Pública de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Assis Santos
- Laboratório de Micologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vanessa Caroline Randi Magalhães
- Laboratório de Micologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Camila Pacheco Silveira Martins da Mata
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
- Hospital Risoleta Tolentino Neves, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Erik Vinicius de Sousa Reis
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Thaís de Fátima Silva Moraes
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Letícia Gomes-de-Pontes
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | - Mayra Gonçalves Menegueti
- Departamento de Enfermagem Geral e Especializada, Escola de Enfermagem de Ribeirão Preto da Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Maria Auxiliadora-Martins
- Divisão de Medicina Intensiva, Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Juan Carlos González Pérez
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Jordana Grazziela Coelho-Dos-Reis
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627- Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| |
Collapse
|
23
|
Weener HJ, van Haaps TF, van Helden RWJ, Albers HJ, Haverkate R, Middelkamp HHT, Ridderikhof ML, van Mens TE, van den Berg A, Mummery CL, Orlova VV, Middeldorp S, van Es N, van der Meer AD. Blood-perfused Vessels-on-Chips stimulated with patient plasma recapitulate endothelial activation and microthrombosis in COVID-19. LAB ON A CHIP 2025; 25:1787-1800. [PMID: 40034052 PMCID: PMC11877278 DOI: 10.1039/d4lc00848k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
A subset of coronavirus disease 2019 (COVID-19) patients develops severe symptoms, characterized by acute lung injury, endothelial dysfunction and microthrombosis. Viral infection and immune cell activation contribute to this phenotype. It is known that systemic inflammation, evidenced by circulating inflammatory factors in patient plasma, is also likely to be involved in the pathophysiology of severe COVID-19. Here, we evaluate whether systemic inflammatory factors can induce endothelial dysfunction and subsequent thromboinflammation. We use a microfluidic Vessel-on-Chip model lined by human induced pluripotent stem cell-derived endothelial cells (hiPSC-ECs), stimulate it with plasma from hospitalized COVID-19 patients and perfuse it with human whole blood. COVID-19 plasma exhibited elevated levels of inflammatory cytokines compared to plasma from healthy controls. Incubation of hiPSC-ECs with COVID-19 plasma showed an activated endothelial phenotype, characterized by upregulation of inflammatory markers and transcriptomic patterns of host defense against viral infection. Treatment with COVID-19 plasma induced increased platelet aggregation in the Vessel-on-Chip, which was associated partially with formation of neutrophil extracellular traps (NETosis). Our study demonstrates that factors in the plasma play a causative role in thromboinflammation in the context of COVID-19. The presented Vessel-on-Chip can enable future studies on diagnosis, prevention and treatment of severe COVID-19.
Collapse
Affiliation(s)
- Huub J Weener
- Department of Bioengineering Technologies, University of Twente, Enschede, The Netherlands.
| | - Thijs F van Haaps
- Department of Vascular Medicine, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension & Thrombosis, Amsterdam, The Netherlands
| | - Ruben W J van Helden
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hugo J Albers
- BIOS Lab-on-a-Chip Group, University of Twente, Enschede, The Netherlands
| | - Rozemarijn Haverkate
- Department of Bioengineering Technologies, University of Twente, Enschede, The Netherlands.
| | | | - Milan L Ridderikhof
- Department of Emergency Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Thijs E van Mens
- Department of Medicine-Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Christine L Mummery
- Department of Bioengineering Technologies, University of Twente, Enschede, The Netherlands.
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Saskia Middeldorp
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Nick van Es
- Department of Vascular Medicine, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension & Thrombosis, Amsterdam, The Netherlands
| | - Andries D van der Meer
- Department of Bioengineering Technologies, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
24
|
Karkar A, Nair P, Mukherjee M, Raghavan NG. Autoimmune response to COVID-19 characterised by eosinophilia, NETosis and EETosis. BMJ Case Rep 2025; 18:e260300. [PMID: 40132935 DOI: 10.1136/bcr-2024-260300] [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: 03/27/2025] Open
Abstract
A previously healthy man in his 50s was admitted with severe COVID-19 pneumonia requiring extracorporeal membrane oxygenation (ECMO) support. He was found to have persistent eosinophilia, with a peak level of 7.5×109/L. He had received multiple courses of steroids, including dexamethasone and methylprednisolone, with no improvement. On day 47, hydrocortisone was initiated for the treatment of septic shock. This resulted in the normalisation of the eosinophil count, which correlated with radiographic and clinical improvement. The patient was transitioned off ECMO and eventually weaned off mechanical ventilation. Studies conducted using the patient's serum identified circulating autoantibodies, which induced neutrophil extracellular traps (NETosis) and eosinophil extracellular traps (EETosis). A varying response of the eosinophils to different corticosteroids was observed in vivo that corresponded with the ex vivo experiments. This case highlights an unrecognised phenomenon of differential response to corticosteroids in severe COVID-19. Furthermore, this is the first reported case of EETosis in COVID-19.
Collapse
Affiliation(s)
- Aram Karkar
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Parameswaran Nair
- McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, Hamiltn, Ontario, Canada
| | - Manali Mukherjee
- McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, Hamiltn, Ontario, Canada
| | - Natya G Raghavan
- McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, Hamiltn, Ontario, Canada
| |
Collapse
|
25
|
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
|
26
|
Nakazawa D, Masuda S, Nishibata Y, Watanabe-Kusunoki K, Tomaru U, Ishizu A. Neutrophils and NETs in kidney disease. Nat Rev Nephrol 2025:10.1038/s41581-025-00944-3. [PMID: 40102634 DOI: 10.1038/s41581-025-00944-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Neutrophils, conventionally regarded as a homogeneous immune cell population, have emerged as a heterogeneous group of cells with distinct gene profiles and immune properties. Activated neutrophils release a spectrum of bioactive substances, including cytokines, chemokines, proteolytic enzymes, reactive oxygen species and neutrophil extracellular traps (NETs), which are composed of decondensed DNA and antimicrobial proteins. NETs have a pivotal role in innate immunity, including in preventing the ascent of uropathogenic bacteria into the kidneys, as they efficiently trap pathogenic microorganisms. However, although indispensable for defence against pathogens, NETs also pose risks of self-damage owing to their cytotoxicity, thrombogenicity and autoantigenicity. Accordingly, neutrophils and NETs have been implicated in the pathogenesis of various disorders that affect the kidneys, including acute kidney injury, vasculitis, systemic lupus erythematosus, thrombotic microangiopathy and in various aetiologies of chronic kidney disease. Pathological alterations in the glomerular vascular wall can promote the infiltration of neutrophils, which can cause tissue damage and inflammation through their interactions with kidney-resident cells, including mesangial cells and podocytes, leading to local cell death. Targeting neutrophil activation and NET formation might therefore represent a new therapeutic strategy for these conditions.
Collapse
Affiliation(s)
- Daigo Nakazawa
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sakiko Masuda
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yuka Nishibata
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Kanako Watanabe-Kusunoki
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
27
|
Huang X, He R, Jiang Y, Tang J, Xu X, Laoguo S, Chen G, Ma J. Neutrophil extracellular traps: potential thrombotic markers and therapeutic targets in colorectal cancer. J Leukoc Biol 2025; 117:qiae235. [PMID: 39454636 DOI: 10.1093/jleuko/qiae235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/24/2024] [Indexed: 10/28/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are promising promoters in venous thromboembolism (VTE). In the present study, we have investigated the potential thrombogenic role of NETs in colorectal cancer (CRC). A total of 583 patients with gastrointestinal malignancies who were diagnosed with or without VTE by extremities arteriovenous ultrasound and computed tomography were enrolled. The incidence of VTE in CRC was as high as 17.53%. In serological ELISA experiments, Cit-H3, myeloperoxidase, and cfDNA were significantly overexpressed in CRC patients with VTE compared with CRC patients without VTE and healthy individuals. Neutrophils from CRC patients with VTE produced appreciable amounts of NETs after stimulation with phorbol-12-myristate-13-acetate, which were lacking in CRC patients without VTE and healthy individuals. CfDNA was positively correlated with plasmin-α2-antiplasmin complex and tissue plasmin activator inhibitor-1 complex, and Cit-H3 was positively correlated with plasmin-α2-antiplasmin complex, suggesting that NETs are associated with increased fibrinolytic activity. We screened some NETs-related genes by analyzing several high-throughput sequencing datasets of VTE and NETs. FCGR1A was identified as the optimal target gene by pan-cancer expression analysis and survival analysis. FCGR1A was significantly overexpressed in the peripheral blood of CRC patients without VTE compared with healthy individuals and showed a positive correlation with cfDNA. Neutrophil-derived NETs were significantly reduced by FCGR1A inhibitor exposure. These findings indicate that NETs are actively involved in VTE in CRC. NETs are promising thrombotic marker and therapeutic target in CRC to prevent the thrombotic consequences of cancer.
Collapse
Affiliation(s)
- Xianye Huang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Rongquan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Yanfeng Jiang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Jing Tang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Xiaoyu Xu
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Shixue Laoguo
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Jie Ma
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| |
Collapse
|
28
|
Lei B, Mu J, Xu G, Yang X, Huang W, Hu L, Liu D, Cheng T, Ma Y, Xu L, Liang Q, Lin Y, Zhou L, Zhou C, Zhang W, Zheng Y. Jing-Yin-Gu-Biao formula protects mice from postinfluenza Staphylococcus aureus infection by ameliorating acute lung injury and improving hypercoagulable state via inhibiting NETosis. Front Immunol 2025; 16:1567522. [PMID: 40134435 PMCID: PMC11933027 DOI: 10.3389/fimmu.2025.1567522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Background Jing-Yin-Gu-Biao formula (JYGBF) is a Chinese medicine derived from Yupingfeng power, Huoxiangzhengqi powder and Yinqiao powder, and has been widely used to treat acute respiratory infections. This study aims to observe the effects of JYGBF against postinfluenza Staphylococcus aureus (S. aureus) infection. Purpose and study design A mouse model of secondary S. aureus infection following PR8 infection was established to evaluate the protective effects of JYGBF against postinfluenza Staphylococcus aureus (S. aureus) infection and related mechanisms were validated in vivo and in vitro. Results The administration of JYGBF significantly ameliorated acute lung injury (ALI) and inhibited overactivated inflammatory response (MIP-2, IL-6, etc.) in mice with postinfluenza S. aureus infection. Single cell RNA-sequencing (scRNA-seq) data indicated that neutrophils had the highest cytokine score in lungs and JYGBF inhibited neutrophil chemotaxis, reactive oxygen species (ROS) biosynthesis and ERK1/2 cascades in neutrophils. Meanwhile, JYGBF inhibited the formation of neutrophil extracellular traps (NETs) in lungs, which is characterized by the production of ROS, peptidyl arginine deiminase 4 (PAD4), citrullinated histone H3 (CitH3), myeloperoxidase (MPO), neutrophil elastase (NE), S100A8/A9 and MPO-CitH3 colocalization. Moreover, JYGBF decreased platelet counts and the expression of its activated markers (CD62P and αIIbβ3) accompanied by the drop of fibrinogen (FIB) and fibrin degradation product (FDP), accounting for alleviating hypercoagulable state. JYGBF inhibited ERK1/2 phosphorylation in neutrophils and in lungs of infected mice. Acacetin, a critical compound from JYGBF, inhibited NET formation via downregulating ERK/ROS axis. Conclusions These results indicated that JYGBF inhibited NET formation and overactivated inflammatory response by suppressing ERK/ROS axis in neutrophils, thereby mitigating ALI and improving the hypercoagulable state during postinfluenza S. aureus infection. JYGBF could be considered a potent therapeutic agent for the prevention and treatment of postinfluenza bacterial infection.
Collapse
Affiliation(s)
- Biao Lei
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingwen Mu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guihua Xu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaodong Yang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenbo Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Liang Hu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Liu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Cheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuhe Ma
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lirong Xu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiankun Liang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Lin
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linqiong Zhou
- Shuguang Hospital, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine Epidemic Research Center, Shanghai, China
- Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Respiratory and Critical Care Medicine, Shanghai, China
| | - Chunxian Zhou
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhang
- Shuguang Hospital, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine Epidemic Research Center, Shanghai, China
- Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Respiratory and Critical Care Medicine, Shanghai, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
29
|
Hu Y, Lu Y, Dong J, Xia D, Li J, Wang H, Rao M, Wang C, Tong W. Epidemiological and clinical characteristics of COVID-19 mortality: a retrospective study. Front Med (Lausanne) 2025; 12:1464274. [PMID: 40130249 PMCID: PMC11930819 DOI: 10.3389/fmed.2025.1464274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
Background The global impact of SARS-CoV-2 and its associated coronavirus disease (COVID-19) has necessitated urgent characterization of prognostic biomarkers. This study aimed to delineate the epidemiological and clinical predictors of mortality among hospitalized COVID-19 patients. Methods A retrospective cohort study was conducted on 123 patients with laboratory-confirmed COVID-19 admitted to Huoshenshan Hospital (Wuhan, China) from 1 February 2020 to 30 April 2020. Kaplan-Meier curve and multivariate Cox regression were used to assess the independent factors with survival time. Statistical significance was set at a p-value of <0.05. Results The cohort exhibited a mortality rate of 49.6% (61/123), with the critical clinical type (HR = 7.970, p = 0.009), leukocytosis (HR = 3.408, p = 0.006), and lymphopenia (HR = 0.817, p = 0.038) emerging as independent predictors of reduced survival. Critical-type patients demonstrated significantly elevated inflammatory markers (neutrophils: 10.41 ± 6.23 × 109/L; CRP: 104.47 ± 29.18 mg/L) and coagulopathy (D-dimer: 5.21 ± 2.34 μg/ml) compared to non-critical cases. Deceased patients exhibited pronounced metabolic derangements, including hyperglycemia (9.81 ± 2.07 mmol/L) and hepatic dysfunction (ALP: 174.03 ± 30.13 U/L). Conclusion We revealed the epidemiological and clinical features of different clinical types of SARS-CoV-2 as summarized in this paper. We found that critical type, leukocyte, and lymphocyte are risk factors that affect survival time, which could be an early and helpful marker to improve management of COVID-19 patients.
Collapse
Affiliation(s)
- Yaohua Hu
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - You Lu
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Shanghai, China
| | - Jiagui Dong
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Delin Xia
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Jin Li
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Hong Wang
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Min Rao
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Chenxing Wang
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Wanning Tong
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| |
Collapse
|
30
|
Ma W, Tang S, Yao P, Zhou T, Niu Q, Liu P, Tang S, Chen Y, Gan L, Cao Y. Advances in acute respiratory distress syndrome: focusing on heterogeneity, pathophysiology, and therapeutic strategies. Signal Transduct Target Ther 2025; 10:75. [PMID: 40050633 PMCID: PMC11885678 DOI: 10.1038/s41392-025-02127-9] [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: 01/17/2024] [Revised: 12/27/2024] [Accepted: 12/27/2024] [Indexed: 03/09/2025] Open
Abstract
In recent years, the incidence of acute respiratory distress syndrome (ARDS) has been gradually increasing. Despite advances in supportive care, ARDS remains a significant cause of morbidity and mortality in critically ill patients. ARDS is characterized by acute hypoxaemic respiratory failure with diffuse pulmonary inflammation and bilateral edema due to excessive alveolocapillary permeability in patients with non-cardiogenic pulmonary diseases. Over the past seven decades, our understanding of the pathology and clinical characteristics of ARDS has evolved significantly, yet it remains an area of active research and discovery. ARDS is highly heterogeneous, including diverse pathological causes, clinical presentations, and treatment responses, presenting a significant challenge for clinicians and researchers. In this review, we comprehensively discuss the latest advancements in ARDS research, focusing on its heterogeneity, pathophysiological mechanisms, and emerging therapeutic approaches, such as cellular therapy, immunotherapy, and targeted therapy. Moreover, we also examine the pathological characteristics of COVID-19-related ARDS and discuss the corresponding therapeutic approaches. In the face of challenges posed by ARDS heterogeneity, recent advancements offer hope for improved patient outcomes. Further research is essential to translate these findings into effective clinical interventions and personalized treatment approaches for ARDS, ultimately leading to better outcomes for patients suffering from ARDS.
Collapse
Affiliation(s)
- Wen Ma
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China
| | - Songling Tang
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Yao
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tingyuan Zhou
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China
| | - Qingsheng Niu
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Liu
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyuan Tang
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Chen
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Gan
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Yu Cao
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China.
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China.
| |
Collapse
|
31
|
Adilović M. COVID-19 related complications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:259-314. [PMID: 40246346 DOI: 10.1016/bs.pmbts.2025.02.002] [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
The COVID-19 pandemic has significantly impacted global healthcare systems, revealed vulnerabilities and prompted a re-evaluation of medical practices. Acute complications from the virus, including cardiovascular and neurological issues, have underscored the necessity for timely medical interventions. Advances in diagnostic methods and personalized therapies have been pivotal in mitigating severe outcomes. Additionally, Long COVID has emerged as a complex challenge, affecting various body systems and leading to respiratory, cardiovascular, neurological, psychological, and musculoskeletal problems. This broad spectrum of complications highlights the importance of multidisciplinary management approaches that prioritize therapy, rehabilitation, and patient-centered care. Vulnerable populations such as paediatric patients, pregnant women, and immunocompromised individuals face unique risks and complications, necessitating continuous monitoring and tailored management strategies to reduce morbidity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička cesta, Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
32
|
Šutković J. Neutrophils and COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:347-384. [PMID: 40246349 DOI: 10.1016/bs.pmbts.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Neutrophils are the first line of defense against pathogens, most effectively by forming Neutrophil Extracellular Traps (NETs). Neutrophiles are further classified into several subpopulations during their development, eliminating pathogens through various mechanisms. However, due to the chaotic and uncontrolled immune response, NETs are often severely resulting in tissue damage and lung infections. The uncontrolled and poorly acknowledged host response regarding the cytokine storm is one of the major causes of severe COVID-19 conditions. Specifically, the increased formation of low-density neutrophils (LDNs), together with neutrophil extracellular traps (NETs) is closely linked with the severity and poor prognosis in patients with COVID-19. In this review, we discuss in detail the ontogeny of neutrophils at different stages and their recruitment and activation after infections, focusing on SARS-CoV-2. In addition, this chapter summarized the research progress on potential targeted drugs (NETs and Cytokine inhibitors) for neutrophil medical therapy and hoped to provide reference for the development of related therapeutic drugs for critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Jasmin Šutković
- Department Genetics and Bioegnineering, International University of Sarajevo, Hrasnička cesta, Bosnia & Herzegovina.
| |
Collapse
|
33
|
Martins-Gonçalves R, Rozini SV, Mendes-de-Almeida DP, Palhinha L, Sacramento CQ, Pereira-Silva GC, Campos MM, de Oliveira DM, Lopes-Cardoso E Souza CA, de Jesus BDBG, de Azevedo-Quintanilha IG, Mouta Nunes de Oliveira P, Pedro RS, Kegele Lignani L, Teixeira GV, Bokel J, Cardoso SW, Hoagland B, Saraiva EM, Grinsztejn B, de Sousa Maia MDL, Amorim Filho L, Hottz ED, Bozza PT. Platelet-neutrophil aggregate formation induces NLRP3 inflammasome activation in vaccine-induced thrombotic thrombocytopenia. J Thromb Haemost 2025; 23:1034-1042. [PMID: 39706368 DOI: 10.1016/j.jtha.2024.12.012] [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: 06/21/2024] [Revised: 11/06/2024] [Accepted: 12/02/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Although rare, vaccine-induced thrombotic thrombocytopenia (VITT) following adenoviral vector COVID-19 vaccination is a concerning and often severe adverse effect of vaccination. The generation of high antiplatelet factor 4 antibody titers promotes the formation of immune complexes capable of activating platelets and neutrophils through FcγRIIa. OBJECTIVES Given that platelet-leukocyte aggregate formation and inflammasome activation are common features of thromboinflammatory diseases, we aimed to evaluate if these are also features of VITT. METHODS Samples from a cohort of 57 postvaccination thrombosis patients and 28 age- and sex-matched unvaccinated individuals were used for ex vivo investigation of platelet-leukocyte aggregate formation and inflammasome activation. RESULTS Patients with clinical features of VITT presented elevated levels of activated caspase-1, interleukin-18, and interleukin-1β in the plasma. We also found that soluble factors in the plasma of VITT patients induce the formation of platelet-neutrophil aggregates but not platelet-monocyte or platelet T-cell aggregates, which are associated with increased caspase-1 activation in neutrophils ex vivo. Platelet-neutrophil aggregate formation was prevented through blockage of FcγRIIa with the neutralizing antibody IV.3 and through blockage of P-selectin or integrin αIIbβ3, also inhibiting caspase-1 activation. Additionally, MCC950, an NLRP3 inflammasome inhibitor, blocked caspase-1 activation. CONCLUSION Taken together, these data show that VITT plasma induces platelet-neutrophil aggregate formation in a FcγRIIa-dependent manner and that platelet-neutrophil interactions may contribute to thromboinflammation in VITT patients by supporting NLRP3 inflammasome activation. These data shed light on novel immunopathological events associated with inflammation and thrombosis in VITT patients.
Collapse
Affiliation(s)
- Remy Martins-Gonçalves
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Daniela P Mendes-de-Almeida
- Department of Hematology, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Department of Medical Affairs, Clinical Studies, and Post-Registration Surveillance, Institute of Technology in Immunobiologicals/Bio-Manguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Carolina Q Sacramento
- Center for Technological Development in Health, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Departament of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Gean Carlo Pereira-Silva
- Laboratory of Innate Immunity, Paulo Góes Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana M Campos
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Carlos A Lopes-Cardoso E Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Department of Hematology, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | | | - Patricia Mouta Nunes de Oliveira
- Department of Medical Affairs, Clinical Studies, and Post-Registration Surveillance, Institute of Technology in Immunobiologicals/Bio-Manguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Renata Saraiva Pedro
- Department of Medical Affairs, Clinical Studies, and Post-Registration Surveillance, Institute of Technology in Immunobiologicals/Bio-Manguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Letícia Kegele Lignani
- Department of Medical Affairs, Clinical Studies, and Post-Registration Surveillance, Institute of Technology in Immunobiologicals/Bio-Manguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Gabriellen Vitiello Teixeira
- Department of Medical Affairs, Clinical Studies, and Post-Registration Surveillance, Institute of Technology in Immunobiologicals/Bio-Manguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Joanna Bokel
- Department of Hematology, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Onco-Hematology Unit, Clínica São Vicente, Rio de Janeiro, Brazil
| | - Sandra Wagner Cardoso
- Laboratory of Clinical Research on STD/AIDS, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Brenda Hoagland
- Laboratory of Clinical Research on STD/AIDS, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Elvira M Saraiva
- Laboratory of Innate Immunity, Paulo Góes Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Beatriz Grinsztejn
- Laboratory of Clinical Research on STD/AIDS, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Maria de Lourdes de Sousa Maia
- Department of Medical Affairs, Clinical Studies, and Post-Registration Surveillance, Institute of Technology in Immunobiologicals/Bio-Manguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Luiz Amorim Filho
- Arthur de Siqueira Cavalcanti State Institute of Hematology, Rio de Janeiro, Brazil
| | - Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| | - Patricia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
34
|
Adriana Gutiérrez-Pérez I, Pérez-Rubio G, Rafael Villafan-Bernal J, Buendía-Roldán I, Zaragoza-García O, Chávez-Galán L, Rosendo-Chalma P, Fricke-Galindo I, Falfán-Valencia R, Paola Guzmán-Guzmán I. Circulating levels of PADs and citrullinated histone H3 in SARS-CoV-2 infection: Influence of genetic polymorphisms. Clin Chim Acta 2025; 569:120180. [PMID: 39904454 DOI: 10.1016/j.cca.2025.120180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Peptidyl arginine deiminases (PADs) and citrullinated H3 histone (H3Cit) play a crucial role in the inflammatory response. These components determine various clinical situations in COVID-2019 associated pneumonia. Single nucleotide polymorphisms (SNPs) in the genes PADI2 and PADI4 may influence the outcome of poorer patient outcomes. We analyze the association of circulating levels NETs biomarkers (PAD2, PAD4, and H3Cit) and the SNPs on PADI2 (rs1005753 and rs2235926) and PADI4 (rs11203366, rs11203367, and rs874881) in hospitalized patients with severe acute respiratory distress syndrome (ARDs) by SARS-CoV-2 pneumonia. METHODS A cross-sectional study in 160 hospitalized patients with ARDs by SARS-CoV-2 pneumonia. The plasma levels of PAD2, PAD4, and H3Cit were determined by ELISA method. The SNPs were determined by qPCR using TaqMan probes. Logistic regression models and receiver operating characteristics (ROC) curve were used to assess the association and predictive value of PAD2, PAD4, and H3Cit plasma levels in outcome by ARDs by SARS-CoV-2 pneumonia. RESULTS PAD2, PAD4, and H3Cit concentrations were predictors of invasive mechanical ventilation (IMV) requirement and non-survival. PAD2 were associated with non-survival, while PAD4 and H3Cit were associated with requirement IMV. In addition, PAD2 and PAD4 concentrations were related with inflammation markers such as NLR, MLR, dNLR, SII, SIRI, AISI, and NHL. In the carriers of TT genotype of rs1005753 of PADI2 were associated with increased of H3Cit, while, the carriers of GTG/GTG haplotype of PADI4 was related to the presence of increased of PAD4 circulating levels. CONCLUSION SNPs in PADI2 and PADI4 have a significant influence on concentrations of PAD2, PAD4, and H3Cit, which are predictor markers of requirement IMV and non-survival in severe ARDS by SARS-CoV-2 pneumonia.
Collapse
Affiliation(s)
- Ilse Adriana Gutiérrez-Pérez
- Laboratory of Multidisciplinary Research and Biomedical Innovation, Faculty of Chemical-Biological Sciences. Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - José Rafael Villafan-Bernal
- Investigador por Mexico, Laboratory of Immunogenomics and Metabolic Disease, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Oscar Zaragoza-García
- Laboratory of Multidisciplinary Research and Biomedical Innovation, Faculty of Chemical-Biological Sciences. Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Pedro Rosendo-Chalma
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer of Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ingrid Fricke-Galindo
- Investigador por Mexico, Laboratory of Immunogenomics and Metabolic Disease, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico.
| | - Iris Paola Guzmán-Guzmán
- Laboratory of Multidisciplinary Research and Biomedical Innovation, Faculty of Chemical-Biological Sciences. Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico.
| |
Collapse
|
35
|
Rajput S, Malviya R, Srivastava S, Ahmad I, Rab SO, Uniyal P. Cardiovascular disease and thrombosis: Intersections with the immune system, inflammation, and the coagulation system. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:228-250. [PMID: 39159826 DOI: 10.1016/j.pharma.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
The coagulation and immune system, both essential physiological systems in the human body, are intricately interconnected and play a critical role in determining the overall health of patients. These systems collaborate via various shared regulatory pathways, such as the Tissue Factor (TF) Pathway. Immunological cells that express TF and generate pro-inflammatory cytokines have the ability to affect coagulation. Conversely, coagulation factors and processes have a reciprocal effect on immunological responses by stimulating immune cells and regulating their functions. These interconnected pathways play a role in both preserving well-being and contributing to a range of pathological disorders. The close relationship between blood clotting and inflammation in the development of vascular disease has become a central focus of clinical study. This research specifically examines the crucial elements of this interaction within the contexts of cardiovascular disease and acute coronary syndrome. Tissue factor, the primary trigger of the extrinsic coagulation pathway, has a crucial function by inducing a proinflammatory reaction through the activation of coagulation factors. This, in turn, initiates coagulation and subsequent cellular signalling pathways. Protease-activated receptors establish the molecular connection between coagulation and inflammation by interacting with activated clotting factors II, X, and VII. Thrombosis, a condition characterised by the formation of blood clots, is the most dreaded consequence of cardiovascular disorders and a leading cause of death globally. Consequently, it poses a significant challenge to healthcare systems. Antithrombotic treatments efficiently target platelets and the coagulation cascade, but they come with the inherent danger of causing bleeding. Furthermore, antithrombotics are unable to fully eliminate thrombotic events, highlighting a treatment deficiency caused by a third mechanism that has not yet been sufficiently addressed, namely inflammation. Understanding these connections may aid in the development of novel approaches to mitigate the harmful mutual exacerbation of inflammation and coagulation. Gaining a comprehensive understanding of the intricate interaction among these systems is crucial for the management of diseases and the creation of efficacious remedies. Through the examination of these prevalent regulatory systems, we can discover novel therapeutic approaches that specifically target these complex illnesses. This paper provides a thorough examination of the reciprocal relationship between the coagulation and immune systems, emphasising its importance in maintaining health and understanding disease processes. This review examines the interplay between inflammation and thrombosis and its role in the development of thrombotic disorders.
Collapse
Affiliation(s)
- Shivam Rajput
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India.
| | - Saurabh Srivastava
- School of Pharmacy, KPJ Healthcare University College (KPJUC), Nilai, Malaysia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| |
Collapse
|
36
|
Pesenti L, de Oliveira Formiga R, Tamassia N, Gardiman E, Chable de la Héronnière F, Gasperini S, Chicher J, Kuhn L, Hammann P, Le Gall M, Saraceni-Tasso G, Martin C, Hosmalin A, Breckler M, Hervé R, Decker P, Ladjemi MZ, Pène F, Burgel PR, Cassatella MA, Witko-Sarsat V. Neutrophils Display Novel Partners of Cytosolic Proliferating Cell Nuclear Antigen Involved in Interferon Response in COVID-19 Patients. J Innate Immun 2025; 17:154-175. [PMID: 40015257 PMCID: PMC11867639 DOI: 10.1159/000543633] [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: 09/12/2024] [Accepted: 01/13/2025] [Indexed: 03/01/2025] Open
Abstract
INTRODUCTION Neutrophils are key players in the hyperinflammatory response during SARS-CoV-2 infection. The cytosolic proliferating cell nuclear antigen (PCNA) is a scaffolding protein highly dependent on the microenvironment status and known to interact with numerous proteins that regulate neutrophil functions. This study aimed to examine the cytosolic protein content and PCNA interactome in neutrophils from COVID-19 patients. METHODS Proteomic analyses were performed on neutrophil cytosols from healthy donors and patients with severe or critical COVID-19. In vitro approaches were used to explore the biological significance of the COVID-19-specific PCNA interactome. RESULTS Neutrophil cytosol analysis revealed a strong interferon (IFN) protein signature, with variations according to disease severity. Interactome analysis identified associations of PCNA with proteins involved in interferon signaling, cytoskeletal organization, and neutrophil extracellular trap (NET) formation, such as protein arginine deiminase type-4 (PADI4) and histone H3, particularly in critical patients. Functional studies of interferon signaling showed that T2AA, a PCNA scaffold inhibitor, downregulated IFN-related genes, including STAT1, MX1, IFIT1, and IFIT2 in neutrophils. Additionally, T2AA specifically inhibited the secretion of CXCL10, an IFN-dependent cytokine. PCNA was also found to interact with key effector proteins implicated in NET formation, such as histone H3, especially in critical COVID-19 cases. CONCLUSION The analysis of the PCNA interactome has unveiled new protein partners that enhance the interferon pathway, thereby modulating immune responses and contributing to hyperinflammation in COVID-19. These findings provide valuable insights into interferon dysregulation in other immune-related conditions. INTRODUCTION Neutrophils are key players in the hyperinflammatory response during SARS-CoV-2 infection. The cytosolic proliferating cell nuclear antigen (PCNA) is a scaffolding protein highly dependent on the microenvironment status and known to interact with numerous proteins that regulate neutrophil functions. This study aimed to examine the cytosolic protein content and PCNA interactome in neutrophils from COVID-19 patients. METHODS Proteomic analyses were performed on neutrophil cytosols from healthy donors and patients with severe or critical COVID-19. In vitro approaches were used to explore the biological significance of the COVID-19-specific PCNA interactome. RESULTS Neutrophil cytosol analysis revealed a strong interferon (IFN) protein signature, with variations according to disease severity. Interactome analysis identified associations of PCNA with proteins involved in interferon signaling, cytoskeletal organization, and neutrophil extracellular trap (NET) formation, such as protein arginine deiminase type-4 (PADI4) and histone H3, particularly in critical patients. Functional studies of interferon signaling showed that T2AA, a PCNA scaffold inhibitor, downregulated IFN-related genes, including STAT1, MX1, IFIT1, and IFIT2 in neutrophils. Additionally, T2AA specifically inhibited the secretion of CXCL10, an IFN-dependent cytokine. PCNA was also found to interact with key effector proteins implicated in NET formation, such as histone H3, especially in critical COVID-19 cases. CONCLUSION The analysis of the PCNA interactome has unveiled new protein partners that enhance the interferon pathway, thereby modulating immune responses and contributing to hyperinflammation in COVID-19. These findings provide valuable insights into interferon dysregulation in other immune-related conditions.
Collapse
Affiliation(s)
- Lucie Pesenti
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
| | | | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | | | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Johana Chicher
- Strasbourg-Esplanade Proteomics Platform, CNRS UAR1589, Molecular and Cellular Biology Institute, University of Strasbourg, Strasbourg, France
| | - Lauriane Kuhn
- Strasbourg-Esplanade Proteomics Platform, CNRS UAR1589, Molecular and Cellular Biology Institute, University of Strasbourg, Strasbourg, France
| | - Philippe Hammann
- Strasbourg-Esplanade Proteomics Platform, CNRS UAR1589, Molecular and Cellular Biology Institute, University of Strasbourg, Strasbourg, France
| | - Morgane Le Gall
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
| | | | - Clémence Martin
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
- Department of Respiratory Medicine, AP-HP, Cochin Hospital, Paris, France
| | - Anne Hosmalin
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
| | - Magali Breckler
- INSERM UMR 1125, Bobigny, France
- UFR SMBH, Li2P, Université Sorbonne Paris Nord, Bobigny, France
| | - Roxane Hervé
- INSERM UMR 1125, Bobigny, France
- UFR SMBH, Li2P, Université Sorbonne Paris Nord, Bobigny, France
| | - Patrice Decker
- INSERM UMR 1125, Bobigny, France
- UFR SMBH, Li2P, Université Sorbonne Paris Nord, Bobigny, France
| | - Maha Zohra Ladjemi
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
| | - Frédéric Pène
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
- Department of Intensive Medicine and Reanimation, AP-HP, Cochin Hospital, Paris, France
| | - Pierre-Régis Burgel
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
- Department of Respiratory Medicine, AP-HP, Cochin Hospital, Paris, France
| | - Marco A. Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | | |
Collapse
|
37
|
Serrano-Gonzalo I, Menéndez-Jandula B, Franco-García E, Arévalo-Vargas I, Lahoz-Gil C, Latre P, Roca-Esteve S, Köhler R, López de Frutos L, Giraldo P. Neutrophil extracellular traps and macrophage activation contibute to thrombosis and post-covid syndrome in SARS-CoV-2 infection. Front Immunol 2025; 16:1507167. [PMID: 40066452 PMCID: PMC11891236 DOI: 10.3389/fimmu.2025.1507167] [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: 10/07/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025] Open
Abstract
Background SARS-CoV-2 infection activates macrophages and induces the release of neutrophil extracellular traps (NETs). Excess NETs is linked to inflammatory and thrombotic complications observed in COVID-19. Aim To explore the impact of NETs and macrophage activation on SARS-CoV-2-infected patients who developed complications. Methods We included 30 patients from the first (March 2020) and 30 from the second wave (July 2021), collecting two plasma samples at diagnosis and seven days later. Data on demographics, comorbidities, and basic analytical data were compiled. NETs markers (myeloperoxidase (MPO), neutrophil elastase (NE), p-selectin (P-SEL) and S100A8/S100A9 heterodimer (MRP)) and macrophage activation markers (Chitotriosidase activity (ChT), CCL18/PARC and YKL-40) were measured. Results The first wave had higher incidences of post-COVID syndrome, ICU admissions, and mortality. Patients of each wave showed elevated blood cells, liver enzymes, and coagulation markers at the time of diagnosis, with fibrinogen and D-Dimer differing between waves. NET and macrophage markers, NE, MPO, MRP, DNAse, ChT, and CCL18 were elevated, while P-SEL, cfDNA, and YKL-40 were decreased if compared to controls. A decrease in NE and DNAse is a link to lower levels of these two markers in complications versus without complications. Conclusions This study emonstrates alterations in NETs and macrophage activation markers in COVID-19 patients, indicating an imbalance in inflammatory response regulation.
Collapse
Affiliation(s)
- Irene Serrano-Gonzalo
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG), Zaragoza, Spain
- Grupo de Investigación Mecanismos de Enfermedad Crónica e Investigación Traslacional (MECIT), Zaragoza, Spain
- Grupo Estudio de Enfermedades de Depósito Lisosomal (GEEDL), Sociedad Española de Hematología y Hemoterapia (SEHH), Zaragoza, Spain
- Grupo de Investigación en Enfermedad de Gaucher (GIIS-012), Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain
| | | | - Esther Franco-García
- Grupo de Investigación Mecanismos de Enfermedad Crónica e Investigación Traslacional (MECIT), Zaragoza, Spain
- Grupo Estudio de Enfermedades de Depósito Lisosomal (GEEDL), Sociedad Española de Hematología y Hemoterapia (SEHH), Zaragoza, Spain
- Servicio de Hematología, Hospital Ntra Sra de Gracia, Zaragoza, Spain
| | - Isidro Arévalo-Vargas
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG), Zaragoza, Spain
- Grupo de Investigación Mecanismos de Enfermedad Crónica e Investigación Traslacional (MECIT), Zaragoza, Spain
- Grupo de Investigación en Enfermedad de Gaucher (GIIS-012), Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain
| | - Calos Lahoz-Gil
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG), Zaragoza, Spain
| | - Paz Latre
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG), Zaragoza, Spain
- Medicina de Familia, Servicio de Atención primaria., Zaragoza, Spain
| | - Sonia Roca-Esteve
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG), Zaragoza, Spain
| | - Ralf Köhler
- Grupo de Investigación Mecanismos de Enfermedad Crónica e Investigación Traslacional (MECIT), Zaragoza, Spain
- Grupo de Investigación en Enfermedad de Gaucher (GIIS-012), Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), Zaragoza, Spain
| | - Laura López de Frutos
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG), Zaragoza, Spain
- Grupo Estudio de Enfermedades de Depósito Lisosomal (GEEDL), Sociedad Española de Hematología y Hemoterapia (SEHH), Zaragoza, Spain
| | - Pilar Giraldo
- Fundación Española para el Estudio y Terapéutica de la Enfermedad de Gaucher y otras lisosomales (FEETEG), Zaragoza, Spain
- Grupo de Investigación Mecanismos de Enfermedad Crónica e Investigación Traslacional (MECIT), Zaragoza, Spain
- Grupo Estudio de Enfermedades de Depósito Lisosomal (GEEDL), Sociedad Española de Hematología y Hemoterapia (SEHH), Zaragoza, Spain
- Grupo de Investigación en Enfermedad de Gaucher (GIIS-012), Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain
- Servicio de Hematología, Hospital QuironSalud, Zaragoza, Spain
| |
Collapse
|
38
|
Jiang RD, Luo YZ, Lin HF, Zheng XS, Zeng WT, Liu MQ, Deng HH, Wang Q, Lai YN, Chen Y, Guo ZS, Zeng Y, Gong QC, Qiu C, Dong M, Wang X, Wang ZY, Ji LN, Hou PP, Li Q, Shen XR, Li B, Gao Y, Zhang AH, Jiang TT, Shi AM, Zhou P, Lin XH, Deng ZQ, Li JM, Shi ZL. Impaired inflammatory resolution with severe SARS-CoV-2 infection in leptin knock out obese hamster. iScience 2025; 28:111837. [PMID: 39981511 PMCID: PMC11841202 DOI: 10.1016/j.isci.2025.111837] [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: 05/12/2024] [Revised: 09/28/2024] [Accepted: 12/13/2024] [Indexed: 02/22/2025] Open
Abstract
Comorbidities, such as obesity, increase the risk of severe COVID-19. However, the mechanisms underlying severe illnesses in individuals with obesity are poorly understood. Here, we used gene-edited leptin knock out (Leptin -/-) obese hamsters to establish a severe infection model. This model exhibits robust viral replication, severe lung lesions, pronounced clinical symptoms, and fatal infection, mirroring severe COVID-19 in patients with obesity. Using single-cell transcriptomics on lung tissues pre- and post-infection, we found that monocyte-derived alveolar macrophages (MD-AM) play a key role in lung hyper-inflammation, including two unique MD-AM cell fate branches specific to Leptin -/- hamsters. Notably, reduced Trem2-dependent efferocytosis pathways in Leptin -/- hamsters indicated weakened inflammation resolution, consistent with the scRNA-seq data from patients with obesity. In summary, our study highlights the obesity-associated mechanisms underlying severe SARS-CoV-2 infections and establishes a reliable preclinical animal model for developing obesity-specific therapeutics for critical COVID-19.
Collapse
Affiliation(s)
- Ren-Di Jiang
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Zhe Luo
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hao-Feng Lin
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Shuang Zheng
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Wen-Tao Zeng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei-Qin Liu
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Hao-Hao Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Qi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Na Lai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ying Chen
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zi-Shuo Guo
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya Zeng
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qian-Chun Gong
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zi-Yi Wang
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Li-Na Ji
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Pan-Pan Hou
- Guangzhou National Laboratory, Guangzhou, China
| | - Qian Li
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xu-Rui Shen
- Guangzhou National Laboratory, Guangzhou, China
| | - Bei Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yun Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ai-Hua Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ting-Ting Jiang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ai-Min Shi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Peng Zhou
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Xin-Hua Lin
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Zi-Qing Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Jian-Min Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
39
|
Yao C, Dong Y, Zhou H, Zou X, Alhaskawi A, Ezzi SHA, Wang Z, Lai J, Kota VG, Abdulla MHAH, Liu Z, Abdalbary SA, Alenikova O, Lu H. COVID-19 and acute limb ischemia: latest hypotheses of pathophysiology and molecular mechanisms. J Zhejiang Univ Sci B 2025; 26:333-352. [PMID: 40274383 PMCID: PMC12021539 DOI: 10.1631/jzus.b2300512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/01/2024] [Indexed: 04/26/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is a multi-system disease that can lead to various severe complications. Acute limb ischemia (ALI) has been increasingly recognized as a COVID-19-associated complication that often predicts a poor prognosis. However, the pathophysiology and molecular mechanisms underlying COVID-19-associated ALI remain poorly understood. Hypercoagulability and thrombosis are considered important mechanisms, but we also emphasize the roles of vasospasm, hypoxia, and acidosis in the pathogenesis of the disease. The angiotensin-converting enzyme 2 (ACE2) pathway, inflammation, and platelet activation may be important molecular mechanisms underlying these pathological changes induced by COVID-19. Furthermore, we discuss the hypotheses of risk factors for COVID-19-associated ALI from genetic, age, and gender perspectives based on our analysis of molecular mechanisms. Additionally, we summarize therapeutic approaches such as use of the interleukin-6 (IL-6) blocker tocilizumab, calcium channel blockers, and angiotensin-converting enzyme inhibitors, providing insights for the future treatment of coronavirus-associated limb ischemic diseases.
Collapse
Affiliation(s)
- Chengjun Yao
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yanzhao Dong
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haiying Zhou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaodi Zou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China
| | - Ahmad Alhaskawi
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sohaib Hasan Abdullah Ezzi
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zewei Wang
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingtian Lai
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Vishnu Goutham Kota
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | | | - Zhenfeng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sahar Ahmed Abdalbary
- Department of Orthopaedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef 2711860, Egypt
| | - Olga Alenikova
- Republic Scientific Practical Center of Neurology and Neurosurgery, Ministry of Health of the Republic of Belarus, Minsk 220004, Belarus
| | - Hui Lu
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
40
|
Liao Y, Liu Y, Li D, Luo S, Huang Y, Wu J, Su J, Yang Y, Wu J, Zhu Z, Yanglan M, Deng H, Wu X, Xu J, Cao F, Cai C, Li Z, Yang R, Deng X, Wei J, Wang L. COVID-19 patient serum-derived extracellular vesicles deliver miR-20b-5p induces neutrophil extracellular traps. Cell Commun Signal 2025; 23:93. [PMID: 39962581 PMCID: PMC11834185 DOI: 10.1186/s12964-025-02095-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Severe cases of COVID-19 are characterized by an excessive presence of neutrophils. Neutrophil extracellular traps (NETs), released by activated neutrophils due to SARS-CoV-2 infection, contribute to lung epithelial cell death and are key drivers in COVID-19-associated immunothrombosis. However, the mechanism underlying NET formation in COVID-19 remain unclear. METHODS Extracellular vesicles (EVs) were isolated from the serum of COVID-19 patients and healthy volunteers, while neutrophils were isolated from blood samples of healthy volunteers. Neutrophils were treated with EVs, and the formation of NETs was observed. To identify the components responsible for the COVID-19-EVs-induced NET formation, we analyzed the expression profiles of microRNA (miRNAs) in COVID-19-EVs. We identified eight highly expressed miRNAs in COVID-19-EVs and explored their potential roles in COVID-19-EVs-mediated NET formation. Additionally, we explored the role of miR-20b-5p in COVID-19-EVs-induced NET formation. RESULTS In this study, we demonstrate that patients with COVID-19 have a higher concentration of serum EVs (COVID-19-EVs) than healthy controls (Normal-EVs). We also found that COVID-19-EVs are internalized by neutrophils to induced NET formation. Through comprehensive miRNA profiling of COVID-19-EVs versus Normal-EVs, we identified 78 differentially expressed miRNAs, with 27 of these being upregulated and 51 being downregulated. Subsequently, we discovered that COVID-19-EVs that were highly abundant with certain miRNAs promote NET formation. Specifically, miR-20b-5p was found to be the strongest inducer of NET formation of the identified miRNAs. Inhibition of miR-20b-5p resulted in a significant decrease in COVID-19-EVs-mediated induction of NET formation. CONCLUSION Herein, we reveal a previously unknown role of COVID-19-EVs in NET formation, which contributes to COVID-19 progression. This study suggests that miR-20b-5p may serve as a potential therapeutic target for COVID-19 treatment.
Collapse
Affiliation(s)
- Yao Liao
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuheng Liu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Dinghao Li
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shiqi Luo
- Institute of Virology, Helmholtz Centre Munich - German Research Centre for Environmental Health, 85764, Neuherberg, Germany
- Chair for Preventions of Infectious Microbial Diseases, School of Life Sciences, Central Institute of Disease Prevention, Technical University of Munich, 85354, Freising, Germany
| | - Yun Huang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Junwei Wu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jin Su
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi Yang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ji Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zifeng Zhu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mengxi Yanglan
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haiyi Deng
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xinyi Wu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Junhao Xu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Feiyang Cao
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Chunmei Cai
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhen Li
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ruibing Yang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xiaoyan Deng
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jie Wei
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Lifu Wang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
41
|
El Azhary K, Ghazi B, Kouhen F, El Bakkouri J, Chamlal H, El Ghanmi A, Badou A. Clinical Impact of Neutrophil Variation on COVID-19 Complications. Diagnostics (Basel) 2025; 15:457. [PMID: 40002608 PMCID: PMC11854688 DOI: 10.3390/diagnostics15040457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Corona virus disease 2019 (COVID-19) poses a threat to global public health. The early identification of critical cases is crucial to providing timely treatment to patients. Here, we investigated whether the neutrophil levels could predict COVID-19 complications. Methods: We performed a retrospective study of patients with COVID-19, admitted to the Cheikh Khalifa International University Hospital, Casablanca, Morocco. Laboratory test results collected upon admission and during hospitalization were analyzed based on clinical information. Results: Our study revealed that a rise in neutrophil "PNN" levels was associated with respiratory deterioration and intubation. They were positively correlated with the procalcitonin and C-reactive protein levels. Interestingly, PNN (polynuclear neutrophil) levels on day 5 proved to be a better predictor of intubation, acute respiratory distress syndrome (ARDS), and mortality than the initial PNN counts, C-reactive protein, or procalcitonin. Moreover, binary logistic regression with stratified PNN-day 5 data revealed that a PNN level on day 5 > 7.7 (109/L) was an independent risk factor for mortality and ARDS. Finally, the PNN levels on day 5 and proinflammatory cytokine IL-6 were positively correlated. Conclusions: Our data showed that neutrophilia proved to be an excellent predictor of complications and mortality during hospitalization and could be used to improve the management of patients with COVID-19.
Collapse
Affiliation(s)
- Khadija El Azhary
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| | - Bouchra Ghazi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura 27182, Morocco
| | - Fadila Kouhen
- Laboratory of Neurosciences and Oncogenetics, Neurooncology and Oncogenetic Team, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco;
- Department of Radiotherapy, International University Hospital Cheikh Khalifa, Casablanca 82403, Morocco
| | - Jalila El Bakkouri
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Cheikh Khalifa International University Hospital, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco
| | - Hasna Chamlal
- Computer Science and Systems Laboratory (LIS), Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| | - Adil El Ghanmi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura 27182, Morocco
| | - Abdallah Badou
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| |
Collapse
|
42
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [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: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
43
|
Sud'ina GF. Neutrophils, Fast and Strong 2.0: Heterogeneity of Neutrophil Parameters in Health and in Disease. Biomedicines 2025; 13:436. [PMID: 40002849 PMCID: PMC11853638 DOI: 10.3390/biomedicines13020436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Neutrophils are very important cells of the immune system, and every year, new nuances of their functional activity in the body and participation in various pathological processes are discovered [...].
Collapse
Affiliation(s)
- Galina F Sud'ina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| |
Collapse
|
44
|
Starikova EA, Mammedova JT, Rubinstein AA, Sokolov AV, Kudryavtsev IV. Activation of the Coagulation Cascade as a Universal Danger Sign. Curr Issues Mol Biol 2025; 47:108. [PMID: 39996829 PMCID: PMC11854423 DOI: 10.3390/cimb47020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Hemostasis is a mechanism that stops bleeding from an injured vessel, involves multiple interlinked steps, culminating in the formation of a "clot" sealing the damaged area. Moreover, it has long been recognized that inflammation also provokes the activation of the coagulation system. However, there has been an increasing amount of evidence revealing the immune function of the hemostasis system. This review collects and analyzes the results of the experimental studies and data from clinical observations confirming the inflammatory function of hemostasis. Here, we summarize the latest knowledge of the pathways in immune system activation under the influence of coagulation factors. The data analyzed allow us to consider the components of hemostasis as receptors recognizing «foreign» or damaged «self» or/and as «self» damage signals that initiate and reinforce inflammation and affect the direction of the adaptive immune response. To sum up, the findings collected in the review allow us to classify the coagulation factors, such as Damage-Associated Molecular Patterns that break down the conventional concepts of the coagulation system.
Collapse
Affiliation(s)
- Eleonora A. Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Department of Microbiology and Virology, Institute of Medical Education Almazov National Medical Research Centre, 2 Akkuratova Street, 197341 Saint Petersburg, Russia
| | - Jennet T. Mammedova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Department of Molecular Biotechnology, Chemical and Biotechnology Faculty, Saint Petersburg State Institute of Technology, Moskovski Ave., 26, 190013 Saint Petersburg, Russia
| | - Artem A. Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
| | - Alexey V. Sokolov
- Laboratory of Systemic Virology, Department of Molecular Biology of Viruses, Smorodintsev Research Institute of Influenza, 15/17, Prof. Popova Str., 197376 Saint Petersburg, Russia;
| | - Igor V. Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| |
Collapse
|
45
|
Zhu W, Zheng Y, Yu M, Witman N, Zhou L, Wei J, Zhang Y, Topchyan P, Nguyen C, Wang D, Janecke R, Padmanabhan A, Baumann Kreuziger L, White GC, Hari P, Gu T, Fields AT, Kornblith LZ, Aster R, Zhu J, Cui W, Jobe S, Graham MB, Wang D, Wen R. Prothrombotic antibodies targeting the spike protein's receptor-binding domain in severe COVID-19. Blood 2025; 145:635-647. [PMID: 39576992 PMCID: PMC11811936 DOI: 10.1182/blood.2024025010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/21/2024] [Accepted: 11/06/2024] [Indexed: 11/24/2024] Open
Abstract
ABSTRACT Thromboembolic complication is common in severe coronavirus disease 2019 (COVID-19), leading to an investigation into the presence of prothrombotic antibodies akin to those found in heparin-induced thrombocytopenia (HIT). In a study of samples from 130 hospitalized patients, collected 3.6 days after COVID-19 diagnosis, 80% had immunoglobulin G (IgG) antibodies recognizing complexes of heparin and platelet factor 4 (PF4; PF4/H), and 41% had antibodies inducing PF4-dependent P-selectin expression in CpG oligodeoxynucleotide-treated normal platelets. Unlike HIT, both PF4/H-reactive and platelet-activating antibodies were found in patients with COVID-19 regardless of recent heparin exposure. Notably, PF4/H-reactive IgG antibodies correlated with those targeting the receptor-binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 spike protein. Moreover, introducing exogenous RBD to or removing RBD-reactive IgG from COVID-19 plasma or IgG purified from COVID-19 plasma significantly reduced their ability to activate platelets. RBD-specific antibodies capable of platelet activation were cloned from peripheral blood B cells of patients with COVID-19. These antibodies possessed sequence motifs in the heavy-chain complementarity-determining region 3 (HCDR3), resembling those identified in pathogenic HIT antibodies. Furthermore, IgG+ B cells having these HCDR3 signatures were markedly expanded in patients with severe COVID-19. Importantly, platelet-activating antibodies present in patients with COVID-19 were associated with a specific elevation of platelet α-granule proteins in the plasma and showed a positive correlation with markers for inflammation and tissue damage, suggesting a functionality of these antibodies in patients. The demonstration of functional and structural similarities between certain RBD-specific antibodies in patients with COVID-19 and pathogenic antibodies typical of HIT suggests a novel mechanism by which RBD-specific antibodies might contribute to thrombosis in COVID-19.
Collapse
Affiliation(s)
- Wen Zhu
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | | | - Mei Yu
- Versiti Blood Research Institute, Milwaukee, WI
| | - Nathan Witman
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Lu Zhou
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Jianhui Wei
- Versiti Blood Research Institute, Milwaukee, WI
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yongguang Zhang
- Versiti Blood Research Institute, Milwaukee, WI
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Paytsar Topchyan
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Christine Nguyen
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - David Wang
- School of Art and Science Undergraduate Program, Washington University in St. Louis, St. Louis, MO
| | - Rae Janecke
- Versiti Blood Research Institute, Milwaukee, WI
| | - Anand Padmanabhan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Lisa Baumann Kreuziger
- Versiti Blood Research Institute, Milwaukee, WI
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | | | - Parameswaran Hari
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Tongjun Gu
- Versiti Blood Research Institute, Milwaukee, WI
| | - Alexander T. Fields
- Department of Surgery, University of California San Francisco, San Francisco, CA
| | - Lucy Z. Kornblith
- Department of Surgery, University of California San Francisco, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Richard Aster
- Versiti Blood Research Institute, Milwaukee, WI
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Weiguo Cui
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shawn Jobe
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Mary Beth Graham
- Division of Infectious Disease, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Demin Wang
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Renren Wen
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
46
|
Retter A, Singer M, Annane D. "The NET effect": Neutrophil extracellular traps-a potential key component of the dysregulated host immune response in sepsis. Crit Care 2025; 29:59. [PMID: 39905519 PMCID: PMC11796136 DOI: 10.1186/s13054-025-05283-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Neutrophils release neutrophil extracellular traps (NETs) as part of a healthy host immune response. NETs physically trap and kill pathogens as well as activating and facilitating crosstalk between immune cells and complement. Excessive or inadequately resolved NETs are implicated in the underlying pathophysiology of sepsis and other inflammatory diseases, including amplification of the inflammatory response and inducing thrombotic complications. Here, we review the growing evidence implicating neutrophils and NETs as central players in the dysregulated host immune response. We discuss potential strategies for modifying NETs to improve patient outcomes and the need for careful patient selection.
Collapse
Affiliation(s)
- Andrew Retter
- Critical Care, Guy's and St Thomas' NHS Foundation Trust, London, UK.
- School of Immunology and Microbial Sciences, King's College, London, UK.
- Volition, London, UK.
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Djillali Annane
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, INSERM, Garches, France
- IHU PROMETHEUS, Comprehensive Sepsis Center, Garches, France
- University Versailles Saint Quentin-University Paris Saclay, INSERM, Garches, France
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), Garches, France
| |
Collapse
|
47
|
Kim J, Seo D, Yoo SY, Lee HJ, Kim J, Yeom JE, Lee JY, Park W, Hong KS, Lee W. Lung-homing nanoliposomes for early intervention in NETosis and inflammation during acute lung injury. NANO CONVERGENCE 2025; 12:8. [PMID: 39894864 PMCID: PMC11788270 DOI: 10.1186/s40580-025-00475-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025]
Abstract
Acute lung injury (ALI) is characterized by severe inflammation in lung tissue, excessive immune response and impaired lung function. In hospitalized high-risk patients and cases of secondary infection due to surgical contamination, it can lead to higher mortality rates and require immediate intervention. Currently, clinical treatments are limited in symptomatic therapy as mechanical ventilation and corticosteroids, having insufficient efficacy in mitigating the cause of progression to severe illness. Here we report a pulmonary targeting lung-homing nanoliposome (LHN) designed to attenuate excessive Neutrophil Extracellular Trap formation (NETosis) through sivelestat and DNase-1, coupled with an anti-inflammatory effect mediated by 25-hydroxycholesterol (25-HC), offering a promising intervention for the acute phase of ALI. Through intratracheal delivery, we intend prompt and constant action within the lungs to effectively prevent excessive NETosis. Isolated neutrophils from blood samples of severe ARDS patients demonstrated significant anti-NETosis effects, as well as reduced proinflammatory cytokine secretion. Furthermore, in a murine model of LPS-induced ALI, we confirmed improvements in lung histopathology, and early respiratory function. Also, attenuation of systemic inflammatory response syndrome (SIRS), with notable reductions in NETosis and neutrophil trafficking was investigated. This presents a targeted therapeutic approach that can be applied in early stages of high-risk patients to prevent severe pulmonary disease progression.
Collapse
Affiliation(s)
- Jungbum Kim
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Donghyuk Seo
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - So-Yeol Yoo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Jin Lee
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jisun Kim
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ji Eun Yeom
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae-Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
- Department of MetaBioHealth, Institute for Cross-disciplinary Studies, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
| | - Kyung Soo Hong
- Division of Pulmonology and Allergy, Department of Internal Medicine, College of Medicine, Regional Center for Respiratory Diseases, Yeungnam University, Yeungnam University Medical Center, Daegu, 42415, Republic of Korea.
| | - Wonhwa Lee
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Department of MetaBioHealth, Institute for Cross-disciplinary Studies, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
48
|
Oliveira JD, Vieira-Damiani G, da Silva LQ, Leonardi GR, Vaz CO, Jacintho-Robison BC, Mazetto BM, de Paula EV, Monica FZ, Orsi FA. Impact of antiplatelets, anticoagulants and cyclic nucleotide stimulators on neutrophil extracellular traps (NETs) and inflammatory markers during COVID-19. J Thromb Thrombolysis 2025; 58:199-209. [PMID: 39546241 DOI: 10.1007/s11239-024-03057-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
While the association between coronavirus disease-19 (COVID-19) and neutrophils extracellular traps (NETs) is recognized, uncertainties remain regarding its precise onset, timing of resolution and target therapy. To assess changes in inflammatory and NET markers during the first week of COVID-19 hospitalization, and the association with disease severity. "In vitro" experiments investigated the effect of antiplatelets, anticoagulants, and cyclic nucleotide stimulators on NETs release. Prospective cohort study, changes in interleukin (IL)-6, IL-8, IL-17, TNF-α, RANTES, PF4, and citrullinated-H3 (citH3) levels within each outcome group was evaluated using ANOVA. Differences between moderately ill, critically ill, and non-survivors were determined using Kruskal-Wallis and logistic regression. Healthy neutrophils were stimulated with phorbol-12-myristate-13-acetate (PMA) or COVID-19 sera and treated with unfractionated heparin (UFH), low molecular weight heparin (LMWH), aspirin (ASA), ticagrelor, cinaciguat, sildenafil, and milrinone. The proportion of NETosis was assessed using IncuCyte Cell Imager. Of the 125 patients, 40.8% had moderate COVID-19, 40.8% had critical COVID-19 but recovered, and 18.4% died. From admission to hospitalization day 8, IL-6 levels decreased in moderately and critically ill, but not in non-survivors, while citH3 levels increased in critically ill and non-survivors. IL-6, IL-8, and TNF-α levels were associated with critical and fatal COVID-19. The release of NETs by neutrophils stimulated with PMA or COVID-19 sera was decreased in the presence of ASA, UFH, LMWH and cyclic nucleotide stimulators in a dose-dependent manner. In the first week of hospitalization, NET markers rose later than inflammatory markers in severe COVID-19 cases. Cyclic nucleotide stimulators, ASA and heparin may emerge as treatment approaches as they may modulate NETosis.
Collapse
Affiliation(s)
- José D Oliveira
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- School of Medical Sciences, Department of Clinical Pathology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Gislaine Vieira-Damiani
- Department of Biology, Federal Institute of Education Science and Technology of São Paulo, Capivari, Brazil
| | - Letícia Q da Silva
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Guilherme R Leonardi
- Department of Translational Medicine (Pharmacology), Faculty of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Camila O Vaz
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- School of Medical Sciences, Department of Clinical Pathology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Bruna C Jacintho-Robison
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- School of Medical Sciences, Department of Clinical Pathology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Bruna M Mazetto
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Erich V de Paula
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Fabíola Z Monica
- Department of Translational Medicine (Pharmacology), Faculty of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Fernanda A Orsi
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil.
- School of Medical Sciences, Department of Clinical Pathology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil.
- Department of Pathology, School of Medical Sciences, University of Campinas, Campinas, Brazil.
| |
Collapse
|
49
|
He W, Yan L, Hu D, Hao J, Liou Y, Luo G. Neutrophil heterogeneity and plasticity: unveiling the multifaceted roles in health and disease. MedComm (Beijing) 2025; 6:e70063. [PMID: 39845896 PMCID: PMC11751288 DOI: 10.1002/mco2.70063] [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: 09/02/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Neutrophils, the most abundant circulating leukocytes, have long been recognized as key players in innate immunity and inflammation. However, recent discoveries unveil their remarkable heterogeneity and plasticity, challenging the traditional view of neutrophils as a homogeneous population with a limited functional repertoire. Advances in single-cell technologies and functional assays have revealed distinct neutrophil subsets with diverse phenotypes and functions and their ability to adapt to microenvironmental cues. This review provides a comprehensive overview of the multidimensional landscape of neutrophil heterogeneity, discussing the various axes along which diversity manifests, including maturation state, density, surface marker expression, and functional polarization. We highlight the molecular mechanisms underpinning neutrophil plasticity, focusing on the complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications that shape neutrophil responses. Furthermore, we explore the implications of neutrophil heterogeneity and plasticity in physiological processes and pathological conditions, including host defense, inflammation, tissue repair, and cancer. By integrating insights from cutting-edge research, this review aims to provide a framework for understanding the multifaceted roles of neutrophils and their potential as therapeutic targets in a wide range of diseases.
Collapse
Affiliation(s)
- Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Dongxue Hu
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| |
Collapse
|
50
|
Ueki H, Wang IH, Kiso M, Horie K, Iida S, Mine S, Ujie M, Hsu HW, Wu CHH, Imai M, Suzuki T, Kamitani W, Kawakami E, Kawaoka Y. Neutrophil adhesion to vessel walls impairs pulmonary circulation in COVID-19 pathology. Nat Commun 2025; 16:455. [PMID: 39805823 PMCID: PMC11730596 DOI: 10.1038/s41467-024-55272-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025] Open
Abstract
Microthrombus formation is associated with COVID-19 severity; however, the detailed mechanism remains unclear. In this study, we investigated mouse models with severe pneumonia caused by SARS-CoV-2 infection by using our in vivo two-photon imaging system. In the lungs of SARS-CoV-2-infected mice, increased expression of adhesion molecules in intravascular neutrophils prolonged adhesion time to the vessel wall, resulting in platelet aggregation and impaired lung perfusion. Re-analysis of scRNA-seq data from peripheral blood mononuclear cells from COVID-19 cases revealed increased expression levels of CD44 and SELL in neutrophils in severe COVID-19 cases compared to a healthy group, consistent with our observations in the mouse model. These findings suggest that pulmonary perfusion defects caused by neutrophil adhesion to pulmonary vessels contribute to COVID-19 severity.
Collapse
Affiliation(s)
- Hiroshi Ueki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
| | - I-Hsuan Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Maki Kiso
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kenta Horie
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research (IAAR), Chiba University, Chiba, Japan
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Sohtaro Mine
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Michiko Ujie
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hung-Wei Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chen-Hui Henry Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Masaki Imai
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Wataru Kamitani
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Eiryo Kawakami
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research (IAAR), Chiba University, Chiba, Japan
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Kanagawa, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- The University of Tokyo, Pandemic Preparedness, Infection and Advanced Research Center, Tokyo, Japan.
| |
Collapse
|