1
|
Savvari P, Skiadas I, Mavrokefalou E, Kakkos S, Antoniou I, Pitoulias GA, Dima E, Ferdoutsis E, Ntaios G, Giannoukas A, Kotsiou O, Zagouri F, Tsoukalas G, Kostikas K, Staramos D, Milionis H, Filis K, Savopoulos C, Kakisis I, Tzilalis V, Koulouris N, Papas T, Skrapari I, Menegas D, VICTORIA Study Group. Α 6-month, multicenter, observational study investigating the treatment of venous thromboembolism in Greece (VICTORIA study). Thromb J 2025; 23:71. [PMID: 40551211 PMCID: PMC12183886 DOI: 10.1186/s12959-025-00749-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 06/02/2025] [Indexed: 06/28/2025] Open
Abstract
BACKGROUND Real-world data are needed to inform clinical practice with regards to anticoagulation treatment for persons with venous thromboembolism (VTE). OBJECTIVES To identify the type and duration of antithrombotic treatment in persons with VTE. Anticoagulation dosage and persistence/adherence were among the secondary objectives. METHODS A multicenter, observational, prospective study conducted in Greek adults with VTE with two on-site visits -baseline and at three months- and a telephone follow-up at 6 months. RESULTS A total of 600 eligible persons were enrolled. The index event was 'PE only' in 50%, 'DVT only' in 40%, and 'DVT+PE' in 10%. Risk factors were categorized as temporary major (21%), temporary minor (37%), and persistent (43%), with active cancer present in 18% of patients. All VTE patients received anticoagulants: 73% received oral anticoagulants (72% DOACs, 1% VKAs) and 70% received parenteral anticoagulants. Treatment was oral only in 30%, parenteral only in 27%, and both in 43%. The most common DOAC was apixaban (47%). Extended anticoagulation (>6 months) was administered to 41% with only 9% (18/198) of those on DOACs receiving a reduced dose. Persistent risk factors predicted extended anticoagulation, while diabetes, COVID-19, and temporary minor risk factors did not. Adherence/persistence rates were similar between DOAC and non-DOAC-treated patients. CONCLUSION VTE was mainly treated with a combination of parenteral and oral anticoagulants. DOACs, primarily apixaban, were the most common oral treatments. Forty percent of patients received extended anticoagulation, mostly at standard dosages. Adherence and persistence rates were high for both DOAC and non-DOAC treatments.
Collapse
Affiliation(s)
- Paraskevi Savvari
- Medical Affairs, Pfizer Hellas S.A, 243 Messoghion Ave, Athens, N. Psychiko, 154 51, Greece
| | - Ioannis Skiadas
- Medical Affairs, Pfizer Hellas S.A, 243 Messoghion Ave, Athens, N. Psychiko, 154 51, Greece
| | | | - Stavros Kakkos
- Department of Vascular Surgery, University of Patras Medical School, Patras, Greece
| | - Ioulia Antoniou
- Medical Affairs, Pfizer Hellas S.A, 243 Messoghion Ave, Athens, N. Psychiko, 154 51, Greece.
| | - Georgios A Pitoulias
- Division of Vascular Surgery, 2nd Surgery Clinic, School of Health Sciences, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Effrosyni Dima
- 1st Department of Critical Care and Pulmonary Services, Evangelismos Hospital, Athens, Greece
| | - Emmanouil Ferdoutsis
- Pulmonary Medicine Department, General Hospital of Heraklion "VENIZELEIO", Heraklion, Greece
| | - Georgios Ntaios
- Department of Internal Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Athanasios Giannoukas
- Vascular Surgery Department, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Ourania Kotsiou
- Respiratory Medicine Department, School of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Flora Zagouri
- Oncology Department of Clinical Therapeutics, Alexandra Hospital, National Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Georgios Tsoukalas
- 4th Respiratory Medicine Department, General Hospital for Chest Diseases of Athens "SOTIRIA", Athens, Greece
| | | | - Dimitrios Staramos
- Vascular Surgery Department, "Konstantopouleion" General Hospital of Nea Ionia, Athens, Greece
| | - Haralampos Milionis
- 1st Department of Internal Medicine, School of Medicine, University Hospital of Ioannina, Ioannina, Greece
| | - Konstantinos Filis
- Vascular Surgery Unit, 1st Department of Propaedeutic Surgery, National and Kapodistrian University of Athens, Hippocration Hospital, Athens, Greece
| | - Christos Savopoulos
- 1st Propaedeutic Internal Medicine Department, AHEPA University General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Kakisis
- Vascular Surgery Department, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasileios Tzilalis
- Vascular Surgery Department, 401 General Military Hospital of Athens, Athens, Greece
| | - Nikolaos Koulouris
- Respiratory Medicine Department, Rehabilitation Unit, 1st Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Theophanis Papas
- Vascular Surgery Department, "Red Cross" General Hospital of Athens, Athens, Greece
| | - Ioanna Skrapari
- 1st Department of Internal Medicine, Evangelismos General Hospital, Athens, Greece
| | - Damianos Menegas
- Medical Affairs, Pfizer Hellas S.A, 243 Messoghion Ave, Athens, N. Psychiko, 154 51, Greece
| | | |
Collapse
Collaborators
Ioannis Kalomenidis, Foteini Malli, Georgios Meletis, Konstantinos Nikolakopoulos, Dimitrios A Chatzelas, Dimitrios Sagris, Christos Karathanos, Maria Kaparelou, Ilektra Voulgareli, Christos Kyriakopoulos, Panagiotis Theodoridis, Sebastian Filippas Ntekouan,
Collapse
|
2
|
Franchini M, Focosi D, Mannucci PM. Coagulation Abnormalities Associated with COVID-19: A Narrative Review. Semin Thromb Hemost 2025. [PMID: 40409294 DOI: 10.1055/a-2619-2485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Coronavirus disease 2019 (COVID-19), a viral respiratory illness caused by severe acute respiratory disease coronavirus 2 (SARS-COV-2), has caused in the last 5 years a global pandemic of unprecedented scale in the modern era. Other than the typical respiratory symptoms, patients suffering from moderate to severe COVID-19 are at risk of developing a peculiar systemic coagulopathy, known as COVID-19-associated coagulopathy. In addition to a predominantly hypercoagulable state, COVID-19 patients may experience hemorrhagic complications triggered by the viral infection. The current knowledge on the underlying molecular mechanisms, the laboratory and clinical characteristics of coagulation abnormalities associated with COVID-19, along with their management, will be summarized in this narrative review.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantua, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Pier Mannuccio Mannucci
- Fondazione IRCCS Ca'Granda-Ospedale Maggiore Policlinico and University of Milan, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| |
Collapse
|
3
|
Wu MCL, Italiano E, Jarvis-Child R, Alwis I, Smythe R, Albornoz EA, Noonan J, Portelli M, Baptista M, Maclean J, Noori P, Yang J, Lee JD, McFadyen JD, Sharland AF, Woodruff TM, Samson AL, Rapkiewicz A, Barrett TJ, Pham A, Schoenwaelder SM, Yuan Y, Jackson SP. Ischaemic endothelial necroptosis induces haemolysis and COVID-19 angiopathy. Nature 2025:10.1038/s41586-025-09076-x. [PMID: 40468079 DOI: 10.1038/s41586-025-09076-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/28/2025] [Indexed: 06/29/2025]
Abstract
Microangiopathy is a major complication of SARS-CoV-2 infection and contributes to the acute and chronic complications of the disease1. Endotheliopathy and dysregulated blood coagulation are prominent in COVID-19 and are considered to be major causes of microvascular obstruction1,2. Here we demonstrate extensive endothelial cell (EC) death in the microvasculature of COVID-19 organs. Notably, EC death was not associated with fibrin formation or platelet deposition, but was linked to microvascular red blood cell (RBC) haemolysis. Importantly, this RBC microangiopathy was associated with ischaemia-reperfusion injury, and was prominent in the microvasculature of humans with myocardial infarction, gut ischaemia, stroke, and septic and cardiogenic shock. Mechanistically, ischaemia induced MLKL-dependent EC necroptosis and complement-dependent RBC haemolysis. Deposition of haemolysed RBC membranes at sites of EC death resulted in the development of a previously unrecognized haemostatic mechanism preventing microvascular bleeding. Exaggeration of this haemolytic response promoted RBC aggregation and microvascular obstruction. Genetic deletion of Mlkl from ECs decreased RBC haemolysis, microvascular obstruction and reduced ischaemic organ injury. Our studies demonstrate the existence of a RBC haemostatic mechanism induced by dying ECs, functioning independently of platelets and fibrin. Therapeutic targeting of this haemolytic process may reduce microvascular obstruction in COVID-19, and other major human diseases associated with organ ischaemia.
Collapse
Affiliation(s)
- Mike C L Wu
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Ethan Italiano
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Rocko Jarvis-Child
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- ThromBio, Sydney, New South Wales, Australia
| | - Imala Alwis
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- ThromBio, Sydney, New South Wales, Australia
| | - Rhyll Smythe
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Eduardo A Albornoz
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Jonathan Noonan
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Marie Portelli
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Marissa Baptista
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Jessica Maclean
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Pashtana Noori
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Jinglu Yang
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - John D Lee
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - James D McFadyen
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Clinical Haematology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Alexandra F Sharland
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Andre L Samson
- Walter and Eliza Hall Research Institute (WEHI), Parkville, Victoria, Australia
- University of Melbourne, Parkville, Victoria, Australia
| | - Amy Rapkiewicz
- New York University Winthrop Hospital, Department of Pathology, Long Island School of Medicine, Mineola, NY, USA
| | - Tessa J Barrett
- Grossman School of Medicine, New York University, New York, NY, USA
| | - Alan Pham
- Department of Anatomical Pathology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Simone M Schoenwaelder
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- ThromBio, Sydney, New South Wales, Australia
- School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Yuping Yuan
- The Heart Research Institute, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- ThromBio, Sydney, New South Wales, Australia
| | - Shaun P Jackson
- The Heart Research Institute, Sydney, New South Wales, Australia.
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia.
- ThromBio, Sydney, New South Wales, Australia.
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
4
|
Song J, Wang Y, Xie Z, Wei J, Wang J. Review of the mechanism of infection induced cerebral small vessel disease. Front Immunol 2025; 16:1594891. [PMID: 40491910 PMCID: PMC12146171 DOI: 10.3389/fimmu.2025.1594891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 05/06/2025] [Indexed: 06/11/2025] Open
Abstract
Cerebral small vessel disease (CSVD) refers to a group of pathological syndromes that affect the brain's microcirculation. These conditions involve damage to small arteries, arterioles, capillaries, venules, and small veins. Cerebrovascular risk factors, immunosenescence, and inflammatory responses contribute to the pathogenesis of cerebral small vessel disease. The global impact of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has drawn significant attention to chronic inflammation caused by infections. Research into the mechanisms by which infections induce CSVD has made continual advancements. It is imperative to reassess the importance of managing infections and the chronic inflammatory phase that follows, highlighting their critical role in the pathogenesis. Our focus encompasses SARS-CoV-2, Human Immunodeficiency Virus (HIV), Hepatitis C Virus (HCV), Zika Virus(ZIKV), Treponema pallidum, as well as the microbial communities within the gut and oral cavity. These pathogen infections and chronic inflammation can contribute to CSVD through mechanisms such as neuroinflammation, blood-brain barrier disruption, microthrombosis, and endothelial cell damage, thereby promoting the occurrence and progression of the disease. This highlights the need for detailed mechanistic research on CSVD associated with these pathogens. Furthermore, we hope that in the future, we will be able to devise targeted prevention and treatment strategies for CSVD based on the unique characteristics of the pathogenic mechanisms associated with various infections.
Collapse
Affiliation(s)
- Jiamei Song
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yiqin Wang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Zhaoxia Xie
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiayi Wei
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Jue Wang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Grinberg N, Wu MA, Moyon Q, Merceron S, Fedun Y, Gousseff M, Sonneville R, Lhote F, Azoulay E, Raphalen JH, Saadoun D, Benhamou Y, Mira JP, Dumas G, Bay P, Devaquet J, Argaud L, Lambert M, Aujayeb A, Henriot B, Bichon A, Bocar T, Harty J, Melchio R, Leibinger F, Calvet L, Urbina T, Bodson L, Tonnelier JM, Reuter D, Canet E, Blaison G, Maizel J, Sedillot N, Dangers L, Eble V, Verlicchi F, Faguer S, Montomoli J, Dingemans G, Mikulski M, Pochard J, Uhel F, Cohen-Aubart F, Luyt CE, Mathian A, Combes A, Colombo R, Amoura Z, Pineton de Chambrun M, EurêClark Study Group. Characteristics of SARS-CoV-2-associated severe episodes of monoclonal gammopathy-associated capillary leak syndrome (Clarkson disease). Ann Intensive Care 2025; 15:72. [PMID: 40419837 DOI: 10.1186/s13613-025-01483-7] [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: 02/11/2025] [Accepted: 04/26/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Monoclonal gammopathy-associated capillary leak syndrome (MG-CLS) is a rare condition characterized by recurrent episodes of hypovolemic shock caused by a sudden increase in capillary permeability. The COVID-19 pandemic has been associated with a rise in MG-CLS episodes and increased mortality. We aimed to explore the association between MG-CLS and SARS-CoV-2 infection. We conducted a multicenter retrospective observational study involving MG-CLS patients who were admitted to the intensive care unit (ICU). The primary endpoint was 28-day mortality according to whether SARS-CoV-2 was identified as a trigger. RESULTS The study included 84 patients (44% women) with a median age of 55 years [IQR 46-62], accounting for 127 ICU admissions. Most patients (88%) had monoclonal gammopathy, predominantly with an IgG heavy chain (98%). A trigger was identified in 63% of cases, primarily suspected or confirmed viral infections, including 26 episodes of SARS-CoV-2 infection. Within 28 days of ICU admission, 32% of patients died. Episodes triggered by SARS-CoV-2 were associated with a higher need for mechanical ventilation (69% vs. 38%, p = 0.004), renal replacement therapy (54% vs. 31%, p = 0.03), and increased 28-day mortality (42% vs. 17%, p = 0.005). Multivariable analysis revealed that SARS-CoV-2 infection was independently associated with 28-day mortality (OR 4.67 [1.08-20.1], p = 0.04). The use of intravenous immunoglobulins did not improve 28-day survival. CONCLUSION In this large cohort of MG-CLS episodes requiring ICU admission, SARS-CoV-2as a trigger was associated with significantly higher 28-day mortality compared to other triggers. Further research is essential to elucidate the specific mechanisms by which SARS-CoV-2 impacts MG-CLS patients.
Collapse
Affiliation(s)
- Nissim Grinberg
- Service de Médecine Intensive-Réanimation, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex, France
| | - Maddalena Alessandra Wu
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Quentin Moyon
- Service de Médecine Intensive-Réanimation, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex, France
- Service de Médecine Interne 2, Centre de Référence National Lupus Systémique, Syndrome des Anticorps Anti-Phospholipides et Autres Maladies Auto-Immunes Systémiques Rares, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Institut E3M, Paris, France
- Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM, UMRS_1166-ICAN, Paris, France
| | - Sybille Merceron
- Service de Médecine-Intensive Réanimation, Hôpital André Mignot, Le Chesnay, France
| | - Yannick Fedun
- Service de Réanimation Polyvalente, CH Bretagne Atlantique, Vannes, France
| | - Marie Gousseff
- Service de Médecine Interne, CH Bretagne Atlantique, Vannes, France
| | - Romain Sonneville
- Service de Médecine-Intensive Réanimation, CHU Bichat, AP-HP, Paris, France
| | - François Lhote
- Service de Médecine Interne, Hôpital Delafontaine, Saint-Denis, France
| | - Elie Azoulay
- Service de Médecine Intensive-Réanimation, AP-HP, Hôpital Saint-Louis, Paris, France
| | | | - David Saadoun
- Department of Internal Medicine and Clinical Immunology, Sorbonne Universités, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Centre National de Références Maladies Autoimmunes et Systémiques Rares, Centre National de Références Maladies Autoinflammatoires Rares et Amylose Inflammatoire, and INSERM, UMR S 959, Immunology- Immunopathology-Immunotherapy (I3), Paris, France
| | - Ygal Benhamou
- Service de Médecine Interne, Normandie Université, UNIROUEN, 76031, Rouen, France
| | - Jean-Paul Mira
- Service de Médecine Intensive-Réanimation, AP-HP, CHU Cochin, Paris, France
| | - Guillaume Dumas
- Service de Réanimation Médicale, CHU Grenoble Alpes, La Tronche, France
| | - Pierre Bay
- Service de Réanimation Médicale, CHU Henri-Mondor, AP-HP, Créteil, France
| | - Jérôme Devaquet
- Service de Réanimation Polyvalente, CH Foch, Suresnes, France
| | - Laurent Argaud
- Service de Médecine Intensive-Réanimation, CHU Edouard-Herriot, Lyon, France
| | - Marc Lambert
- Département de Médecine Interne et d'Immunologie Clinique, Univ. Lille, CHU Lille, 59000, Lille, France
| | - Avinash Aujayeb
- Northumbria Specialist Emergency Care Hospital, Newcastle, England
| | - Basile Henriot
- Service de Médecine Interne, GHT d'Armor, Saint-Brieuc, France
| | - Amandine Bichon
- Service de Médecine Intensive-Réanimation, Assistance Publique Hôpitaux de Marseille, Hôpital La TimoneMarseille, France
| | - Thomas Bocar
- Service d'Accueil des Urgences, CHU Pontchaillou, Rennes, France
| | - John Harty
- Southern Health and Social Care Trust, Craigavon, Northern Ireland
| | - Remo Melchio
- Divisione Di Medicina Interna, A.O. S. Croce E Carle, Cuneo, Italy
| | - Franck Leibinger
- Service de Réanimation Polyvalente, CH de Perpignan, Perpignan, France
| | - Laure Calvet
- Service de Médecine Intensive-Réanimation, CHU Gabriel-Montpied, Clermont-Ferrand, France
| | - Tomas Urbina
- Service de Médecine Intensive-Réanimation, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Laurent Bodson
- Service de Réanimation, Clinique Saint Gatien Alliance, Saint-Cyr-Sur-Loire, France
| | | | - Danielle Reuter
- Service de Médecine Intensive-Réanimation, Centre Hospitalier Sud Francilien, Corbeil-Essonnes, France
| | - Emmanuel Canet
- Service de Médecine Intensive-Réanimation, CHU de Nantes, Nantes, France
| | - Gilles Blaison
- Service de Médecine Interne, Hôpital de Colmar, Colmar, France
| | - Julien Maizel
- Service de Médecine Intensive-Réanimation, CHU d'Amiens, Amiens, France
| | - Nicholas Sedillot
- Service de Médecine Intensive-Réanimation, CH de Bourg-en-Bresse, Bourg-en-Bresse, France
| | - Laurence Dangers
- Service de Médecine Intensive-Réanimation, CHU de La Réunion, Saint-Denis de La Réunion, France
| | - Vincent Eble
- Service de Médecine Interne, CH d'Evreux, Evreux, France
| | - Franco Verlicchi
- Transfusion Medicine Faenza-Lugo, Transfusion Service Ravenna, AUSL Romagna, Ravenna, Italy
| | - Stanislas Faguer
- Département de Néphrologie et Transplantation D'organes, Centre de Référence des Maladies Rénales Rares, INSERM U1297 (I2MC, Équipe 12), CHU de Toulouse, France
| | - Jonathan Montomoli
- Department of Intensive Care, Ospedale "Infermi", Rimini, Romagna Local Health Authority, Italy
| | | | - Marc Mikulski
- Service de Réanimation Polyvalente, CHT Gaston-Bourret, Nouméa, France
| | - Jonas Pochard
- Service de Réanimation Chirurgicale, Hôpital de Bicètre, AP-HP, Le Kremlin-Bicêtre, France
| | - Fabrice Uhel
- Médecine Intensive Réanimation, AP-HP, Hôpital Louis Mourier, DMU ESPRIT, 92700, Colombes, France
| | - Fleur Cohen-Aubart
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
- Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Paris, France
| | - Charles-Edouard Luyt
- Service de Médecine Intensive-Réanimation, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex, France
- Service de Médecine Interne 2, Centre de Référence National Lupus Systémique, Syndrome des Anticorps Anti-Phospholipides et Autres Maladies Auto-Immunes Systémiques Rares, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Institut E3M, Paris, France
| | - Alexis Mathian
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
- Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Paris, France
| | - Alain Combes
- Service de Médecine Intensive-Réanimation, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex, France
- Service de Médecine Interne 2, Centre de Référence National Lupus Systémique, Syndrome des Anticorps Anti-Phospholipides et Autres Maladies Auto-Immunes Systémiques Rares, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Institut E3M, Paris, France
| | - Riccardo Colombo
- Division of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital-Polo Universitario-University of Milan, Milan, Italy
| | - Zahir Amoura
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
- Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Paris, France
| | - Marc Pineton de Chambrun
- Service de Médecine Intensive-Réanimation, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex, France.
- Division of Internal Medicine, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy.
- Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Paris, France.
| | | |
Collapse
|
6
|
Popa E, Popa AE, Poroch M, Poroch V, Ungureanu MI, Slanina AM, Bacusca A, Coman EA. The Molecular Mechanisms of Cognitive Dysfunction in Long COVID: A Narrative Review. Int J Mol Sci 2025; 26:5102. [PMID: 40507911 PMCID: PMC12154490 DOI: 10.3390/ijms26115102] [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: 04/05/2025] [Revised: 05/18/2025] [Accepted: 05/21/2025] [Indexed: 06/16/2025] Open
Abstract
Cognitive dysfunction represents one of the most persistent and disabling features of Long COVID, yet its molecular underpinnings remain incompletely understood. This narrative review synthesizes current evidence on the pathophysiological mechanisms linking SARS-CoV-2 infection to long-term neurocognitive sequelae. Key processes include persistent neuroinflammation, blood-brain barrier (BBB) disruption, endothelial dysfunction, immune dysregulation, and neuroendocrine imbalance. Microglial activation and cytokine release (e.g., IL-6, TNF-α) promote synaptic dysfunction and neuronal injury, while activation of inflammasomes such as NLRP3 amplifies CNS inflammation. Vascular abnormalities, including microthrombosis and BBB leakage, facilitate the infiltration of peripheral immune cells and neurotoxic mediators. Hypothalamic-pituitary-adrenal axis dysfunction and reduced vagal tone further exacerbate systemic inflammation and autonomic imbalance. Biomarkers such as GFAP, NFL, IL-6, and S100B have been associated with both neuroinflammation and cognitive symptoms. Notably, transcriptomic signatures in Long COVID overlap with those observed in Alzheimer's disease, highlighting shared pathways involving tau dysregulation, oxidative stress, and glial reactivity. Understanding these mechanisms is critical for identifying at-risk individuals and developing targeted therapeutic strategies. This review underscores the need for longitudinal research and integrative biomarker analysis to elucidate the molecular trajectory of cognitive impairment in Long COVID.
Collapse
Affiliation(s)
- Elena Popa
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.P.); (V.P.); (M.I.U.); (A.M.S.); (A.B.); (E.A.C.)
- Department of Family Medicine, Preventive Medicine and Interdisciplinary, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str. 16, 700115 Iasi, Romania
| | | | - Mihaela Poroch
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.P.); (V.P.); (M.I.U.); (A.M.S.); (A.B.); (E.A.C.)
- Department of Family Medicine, Preventive Medicine and Interdisciplinary, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str. 16, 700115 Iasi, Romania
| | - Vladimir Poroch
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.P.); (V.P.); (M.I.U.); (A.M.S.); (A.B.); (E.A.C.)
| | - Monica Iuliana Ungureanu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.P.); (V.P.); (M.I.U.); (A.M.S.); (A.B.); (E.A.C.)
- Department of Family Medicine, Preventive Medicine and Interdisciplinary, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str. 16, 700115 Iasi, Romania
| | - Ana Maria Slanina
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.P.); (V.P.); (M.I.U.); (A.M.S.); (A.B.); (E.A.C.)
- Department of Family Medicine, Preventive Medicine and Interdisciplinary, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str. 16, 700115 Iasi, Romania
| | - Agnes Bacusca
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.P.); (V.P.); (M.I.U.); (A.M.S.); (A.B.); (E.A.C.)
- Department of Family Medicine, Preventive Medicine and Interdisciplinary, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str. 16, 700115 Iasi, Romania
| | - Elena Adorata Coman
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Str., 700115 Iasi, Romania; (M.P.); (V.P.); (M.I.U.); (A.M.S.); (A.B.); (E.A.C.)
- Department of Family Medicine, Preventive Medicine and Interdisciplinary, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str. 16, 700115 Iasi, Romania
| |
Collapse
|
7
|
Li J, Geng Y, Luo Y, Sun X, Guo Y, Dong Z. Pathological roles of NETs-platelet synergy in thrombotic diseases: From molecular mechanisms to therapeutic targeting. Int Immunopharmacol 2025; 159:114934. [PMID: 40418882 DOI: 10.1016/j.intimp.2025.114934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 05/10/2025] [Accepted: 05/20/2025] [Indexed: 05/28/2025]
Abstract
The formation of neutrophil extracellular traps (NETs) is a novel way for neutrophils to perform organismal protective functions essential for protecting the host against infections. Nevertheless, an increasing amount of data shows that uncontrolled or excessive formation of NETs in the body leads to inflammation and thrombosis. Many serious human diseases, such as sepsis, stroke, cancer, and autoimmune diseases, are associated with thrombosis, and inhibiting its formation is essential to prevent the development of many inflammatory and thrombotic diseases. With deeper research, it has been found that there is a complex interaction between NETs and platelets: platelets activate neutrophils to form NETs, while NET components enhance platelet aggregation and activation. This self-perpetuating vicious cycle between them mediates pathological processes such as inflammation, coagulation, and thrombosis. A deeper comprehension of the underlying molecular mechanisms between them promises to be a new target for thrombotic diseases. In this review, we concentrate on a summary of NET formation and its mechanisms of action. Providing a thorough summary of how neutrophils are activated by platelets to form NETs, how NETs cause platelet activation, and how this close interaction during inflammatory events affects the course of the disease, with the aim of providing fresh targets and ideas for thrombotic disease clinical prevention and therapy.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
| | - Yifei Geng
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; Beijing Key Laboratory of Neuro-Innovative Drug Research and Development of Traditional Chinese Medicine (Natural Medicines), No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; Beijing Key Laboratory of Neuro-Innovative Drug Research and Development of Traditional Chinese Medicine (Natural Medicines), No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; Beijing Key Laboratory of Neuro-Innovative Drug Research and Development of Traditional Chinese Medicine (Natural Medicines), No. 151, Malianwa North Road, Haidian District, Beijing 100193, China.
| | - Zhengqi Dong
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; Beijing Key Laboratory of Neuro-Innovative Drug Research and Development of Traditional Chinese Medicine (Natural Medicines), No. 151, Malianwa North Road, Haidian District, Beijing 100193, China.
| |
Collapse
|
8
|
Hostiuc M, Negoi I. Etiology and Risk Factors for Splanchnic Vein Thrombosis in Non-Cirrhotic, Non-Neoplastic Patients: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:933. [PMID: 40428892 PMCID: PMC12113251 DOI: 10.3390/medicina61050933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/13/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Splanchnic vein thrombosis (SVT) is a heterogeneous group of disorders affecting the portal, mesenteric, splenic, and hepatic veins. While frequently associated with liver cirrhosis and malignancy, SVT also occurs in non-cirrhotic, non-neoplastic patients. This narrative review evaluates the epidemiology and risk factors for SVT in this population. The prevalence and incidence of SVT in non-cirrhotic, non-neoplastic patients remain incompletely characterized, with estimates varying widely across studies. The clinical significance of SVT relates to potential complications, including intestinal ischemia, portal hypertension, and a possible underlying systemic disorder. Risk factors for SVT can be categorized into local abdominal conditions, thrombophilias, and systemic disorders. Local factors include inflammatory bowel disease, pancreatitis, abdominal surgery, and trauma. Thrombophilias, both inherited and acquired, are significant contributors to SVT risk. Systemic conditions associated with SVT include autoimmune disorders, pregnancy, hematological diseases, and infections. The complex interplay of these risk factors highlights the need for a comprehensive evaluation of SVT patients. Early recognition and management of these conditions can prevent potentially life-threatening complications and guide decisions regarding anticoagulation and long-term follow-up.
Collapse
Affiliation(s)
- Mihaela Hostiuc
- Internal Medicine, Department 5, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ionut Negoi
- General Surgery, Department 10, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| |
Collapse
|
9
|
Imazio M, Basso C, Brucato A, Klingel K, Kuchynka P, Lazaros G, Merlo M, Sinagra G, Adler Y, Bucciarelli Ducci C, Cameli M, Castelletti S, Caforio AL, Crotti L, Dagna L, Frustaci A, Klein A, Kuusisto J, Lopez Sainz A, Marcolongo R, Pantazis A, Rigopoulos AG, Ristic A, Seferovic P, Sheppard M, Tschöpe C, Lüscher T. Myopericardial complications following COVID-19 disease and vaccination: a clinical consensus statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J 2025:ehaf222. [PMID: 40390594 DOI: 10.1093/eurheartj/ehaf222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/21/2025] Open
Abstract
The aim of the present clinical consensus statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases is to review the current knowledge on the epidemiology, pathogenesis, diagnosis, therapy, and outcomes of myocardial and pericardial complications of coronavirus disease 2019 (COVID-19) and vaccination in order to improve the awareness and clinical confidence on the management of patients with these complications. The risk of myopericardial complications is especially higher within 1 month of COVID-19 disease and vaccination. Forms related to the disease are generally more common and severe than those related to vaccination. Even if vaccination against COVID-19 increases myocarditis risk, this risk is lower in vaccinated than non-vaccinated COVID-19 individuals, supporting the vaccine use. Overall, COVID-19 related complications, especially myocarditis, are relatively rare.
Collapse
Affiliation(s)
- Massimo Imazio
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Cardiothoracic Department, University Hospital Santa Maria della Misericordia, ASUFC, Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy
| | - Cristina Basso
- Cardiovascular Pathology Unit, Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Antonio Brucato
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital of Tübingen, Tübingen, Germany
| | - Petr Kuchynka
- 2nd Department of Medicine, Department of Cardiovascular Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - George Lazaros
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University, Athens, Greece
| | - Marco Merlo
- CardioThoracoVascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina, Trieste, Italy
- University of Trieste (Italy), member of ERN-Guard Heart, Trieste, Italy
| | - Gianfranco Sinagra
- CardioThoracoVascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina, Trieste, Italy
- University of Trieste (Italy), member of ERN-Guard Heart, Trieste, Italy
| | - Yehuda Adler
- Sackler Faculty of Medicine, Tel Aviv University, Israel
| | | | | | - Silvia Castelletti
- IRCCS, Istituto Auxologico Italiano, Department of Cardiology, San Luca Hospital, Cardiomyopathy Unit, Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Alida Linda Caforio
- Cardiology, Dept of Cardiac Thoracic Vascular Science and Public Health, University of Padova, *member of ERN Guard-Heart, Padova, Italy
| | - Lia Crotti
- IRCCS, Istituto Auxologico Italiano, Department of Cardiology, San Luca Hospital, Cardiomyopathy Unit, Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Frustaci
- Cellular and Molecular Cardiology Lab, IRCCS L. Spallanzani, Rome, Italy
| | - Allan Klein
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Johanna Kuusisto
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Angela Lopez Sainz
- Department of Cardiology, Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain
| | - Renzo Marcolongo
- Cardiology, Dept of Cardiac Thoracic Vascular Science and Public Health, University of Padova, *member of ERN Guard-Heart, Padova, Italy
| | - Antonis Pantazis
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | | | - Arsen Ristic
- Department of Cardiology, University Clinical Center of Serbia and Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Petar Seferovic
- Serbian Academy of Sciences and Arts, University of Belgrade Faculty of Medicine and Heart Failure Center, Belgrade University Medical Center, Belgrade, Serbia
| | - Mary Sheppard
- Department of Cardiovascular Pathology, Cardiovascular and Genetics Research Institute, City St Georges University of London, London, UK
| | - Carsten Tschöpe
- Department of Cardiology, Angiology, and Intensive Medicine (CVK), German Heart Center at Charite (DHZC), Berlin, Germany
- Berlin Institute of Health (BIH) at Charite and Berlin-Berlin Brandenburger Center for Regenerative Therapies (BCRT), Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHC), Partner Side Berlin, Germany
| | - Thomas Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and Cardiovascular Academic Group, King's College and National Heart and Lung Institute, Imperial College, London, UK
- Center for Molecualr Cardiology, University of Zurich, Switzerland
| |
Collapse
|
10
|
Liu Q, Chen R, Zhang Z, Sha Z, Wu H. Mechanisms and immune crosstalk of neutrophil extracellular traps in response to infection. mBio 2025; 16:e0018925. [PMID: 40237474 PMCID: PMC12077121 DOI: 10.1128/mbio.00189-25] [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] [Indexed: 04/18/2025] Open
Abstract
Neutrophil extrusion of neutrophil extracellular traps (NETs) in a process called NETosis provides immune defense against extracellular bacteria. It has been observed that bacteria are capable of activating neutrophils to release NETs that subsequently kill them or at least prevent their local spread within host tissue. However, existing studies have mainly focused on the isolated function of NETs, with less attention given to their anti-bacterial mechanisms through interactions with other immune cell populations. The net effect of these complex intercellular interactions, which may act additively, synergistically, or antagonistically, is a critical determinant in the outcomes of host-pathogen interactions. This review summarizes the mechanisms underlying classic NET formation and their crosstalk with the immune system, offering novel insights aimed at balancing the anti-microbial function with their potential inflammatory risks.
Collapse
Affiliation(s)
- Qi Liu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Ruke Chen
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Ziyan Zhang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
11
|
Pekayvaz K, Kilani B, Joppich M, Eivers L, Brambs S, Knottenberg V, Akgöl S, Yue K, Li L, Martinez-Navarro A, Kaiser R, Meißner N, Schulz H, Belz L, Akhalkatsi A, Stockhausen S, Mueller TT, Millonig S, Hartelt L, Gold C, Janjic A, Polewka V, Wendler F, Droste Zu Senden A, Titova A, Leunig A, Voelkl M, Engelmann B, Hernandez Petzsche MR, Boeckh-Behrens T, Liebig T, Winning S, Fandrey J, Dichgans M, Enard W, Zimmer R, Tiedt S, Massberg S, Nicolai L, Stark K. Immunothrombolytic monocyte-neutrophil axes dominate the single-cell landscape of human thrombosis and correlate with thrombus resolution. Immunity 2025; 58:1343-1358.e13. [PMID: 40280129 DOI: 10.1016/j.immuni.2025.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Thrombotic diseases remain the major cause of death and disability worldwide, and the contribution of inflammation is increasingly recognized. Thromboinflammation has been identified as a key pathomechanism, but an unsupervised map of immune-cell states, trajectories, and intercommunication at a single-cell level has been lacking. Here, we reveal innate leukocyte substates with prominent thrombolytic properties by employing single-cell omics measures on human stroke thrombi. Using in vivo and in vitro thrombosis models, we propose a pro-resolving monocyte-neutrophil axis, combining two properties: (1) NR4A1hi non-classical monocytes acquire a thrombolytic and neutrophil-chemoattractive phenotype, and (2) blood neutrophils are thereby continuously recruited to established thrombi through CXCL8-CXCR1 and CXCR2 and adopt a hypoxia-induced thrombus-resolving urokinase receptor (PLAUR)+ phenotype. This immunothrombolytic axis results in thrombus resolution. Together, with this immune landscape of thrombosis, we provide a valuable resource and introduce the concept of "immunothrombolysis" with broad mechanistic and translational implications at the crossroad of inflammation and thrombosis.
Collapse
Affiliation(s)
- Kami Pekayvaz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Badr Kilani
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luke Eivers
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Sophia Brambs
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sezer Akgöl
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Keyang Yue
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Li
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Rainer Kaiser
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Nina Meißner
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Heiko Schulz
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Larissa Belz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sven Stockhausen
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Tonina T Mueller
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Simon Millonig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lea Hartelt
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Gold
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Vivien Polewka
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Franziska Wendler
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Anna Titova
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Leunig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Voelkl
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Moritz R Hernandez Petzsche
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boeckh-Behrens
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Liebig
- Institute for Diagnostic and Interventional Neuroradiology, University Hospital, LMU Munich, Munich, Germany
| | - Sandra Winning
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Joachim Fandrey
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Martin Dichgans
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Massberg
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Konstantin Stark
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
12
|
Ignacio-Mejía I, Bandala C, González-Zamora JF, Chavez-Galan L, Buendia-Roldan I, Pérez-Torres K, Rodríguez-Díaz MZ, Pacheco-Tobón DX, Quintero-Fabián S, Vargas-Hernández MA, Carrasco-Vargas H, Falfán-Valencia R, Pérez-Rubio G, Hernández-Lara KA, Gómez-Manzo S, Ortega-Cuellar D, Ignacio-Mejía F, Cárdenas-Rodríguez N. Association of Vitamin D Supplementation with Glutathione Peroxidase (GPx) Activity, Interleukine-6 (IL-6) Levels, and Anxiety and Depression Scores in Patients with Post-COVID-19 Condition. Int J Mol Sci 2025; 26:4582. [PMID: 40429727 PMCID: PMC12110956 DOI: 10.3390/ijms26104582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) presents with various symptoms, and some patients develop post-COVID-19 condition (PCC). Vitamin D has shown therapeutic potential in COVID-19 and may offer benefits for PCC. The aim of this study was to evaluate the differences associated with two supplementation strategies (bolus and daily) on interleukin-6 (IL-6) levels, glutathione peroxidase (GPx) activity, and clinical outcomes in PCC patients, regardless of whether target 25 (OH) D levels reached the ideal range. We conducted a self-controlled study in which 54 participants with PCC were supplemented with vitamin D3 (n = 28 bolus and n = 26 daily) for 2 months. Blood samples were collected to measure IL-6 levels and GPx activity using spectrophotometric methods. The Hospital Anxiety and Depression Scale (HADS) was used to assess mental function. Both bolus and daily vitamin D supplementation were significantly associated with increased GPx activity and decreased IL-6 levels. Daily supplementation was additionally associated with a significant reduction in anxiety and depression scores. However, neither regimen was associated with improvements in cough, dyspnea, or fatigue. These findings suggest a potential association between vitamin D supplementation and improvements in antioxidant and neuropsychiatric parameters in PCC, possibly mediated by its immunomodulatory and antioxidant properties. Further placebo-controlled trials are warranted to determine whether these observed associations reflect causal relationships.
Collapse
Affiliation(s)
- Iván Ignacio-Mejía
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico; (I.I.-M.); (S.Q.-F.)
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Cindy Bandala
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | | | - Leslie Chavez-Galan
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico;
| | - Ivette Buendia-Roldan
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibróticas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (M.Z.R.-D.); (D.X.P.-T.)
| | - Karina Pérez-Torres
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibróticas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (M.Z.R.-D.); (D.X.P.-T.)
| | - María Zobeida Rodríguez-Díaz
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibróticas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (M.Z.R.-D.); (D.X.P.-T.)
| | - Denilson Xipe Pacheco-Tobón
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibróticas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (K.P.-T.); (M.Z.R.-D.); (D.X.P.-T.)
| | - Saray Quintero-Fabián
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico; (I.I.-M.); (S.Q.-F.)
| | - Marco Antonio Vargas-Hernández
- Subdirección de Investigación, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico;
| | - Humberto Carrasco-Vargas
- Dirección de la Escuela Militar de Medicina, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico;
| | - Ramcés Falfán-Valencia
- Laboratorio de HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (R.F.-V.)
| | - Gloria Pérez-Rubio
- Laboratorio de HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (R.F.-V.)
| | - Kevin Alexis Hernández-Lara
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Daniel Ortega-Cuellar
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | | | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| |
Collapse
|
13
|
Ma YN, Ma SR, Yang L, Wu J, Wang YR, Bao LJ, Ma L, Wu QQ, Wang ZH. Diagnostic biomarkers and immune infiltration profiles common to COVID-19, acute myocardial infarction and acute ischaemic stroke using bioinformatics methods and machine learning. BMC Neurol 2025; 25:201. [PMID: 40340571 PMCID: PMC12060493 DOI: 10.1186/s12883-025-04212-6] [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: 11/13/2024] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND COVID-19 is a disease that affects people globally. Beyond affecting the respiratory system, COVID-19 patients are at an elevated risk for both venous and arterial thrombosis. This heightened risk contributes to an increased probability of acute complications, including acute myocardial infarction (AMI) and acute ischemic stroke (AIS). Given the unclear relationship between COVID-19, AMI, and AIS, it is crucial to gain a deeper understanding of their associations and potential molecular mechanisms. This study aims to utilize bioinformatics to analyze gene expression data, identify potential therapeutic targets and biomarkers, and explore the role of immune cells in the disease. METHODS This study employed three Gene Expression Omnibus (GEO) datasets for analysis, which included data on COVID-19, AMI and AIS. We performed enrichment analysis on the co-DEGs for these three diseases to clarify gene pathways and functions, and also examined the relationship between co-DEGs and immune infiltration. Machine learning techniques and protein-protein interaction networks (PPI) were used to identify hub genes within the co-DEGs. Finally, we employed a dual validation strategy integrating independent GEO datasets and in vitro experiments with human blood samples to comprehensively assess the reliability of our experimental findings. RESULTS We identified 88 co-DEGs associated with COVID-19, AMI and AIS. Enrichment analysis results indicated that co-DEGs were significantly enriched in immune inflammatory responses related to leukocytes and neutrophils. Immune infiltration analysis revealed significant differences in immune cell populations between the disease group and the normal group. Finally, genes selected through machine learning methods included: CLEC4E, S100A12, and IL1R2. Based on the PPI network, the top ten most influential DEGs were identified as MMP9, TLR2, TLR4, ITGAM, S100A12, FCGR1A, CD163, FCER1G, FPR2, and CLEC4D. The integration of the protein-protein interaction (PPI) network with machine learning techniques facilitated the identification of S100A12 as a potential common biomarker for early diagnosis and a therapeutic target for all three diseases. Ultimately, validation of S100A12 showed that it was consistent with our experimental results, confirming its reliability as a biomarker. Moreover, it demonstrated good diagnostic performance for the three diseases. CONCLUSION We employed bioinformatics methods and machine learning to investigate common diagnostic biomarkers and immune infiltration characteristics of COVID-19, AMI and AIS. Functional and pathway analyses indicated that the co-DEGs were primarily enriched in immune inflammatory responses related to leukocytes and neutrophils. Through two machine learning approaches and the PPI network, and subsequent validation and evaluation, we identified S100A12 as a potential common therapeutic target and biomarker related to immune response that may influence these three diseases.
Collapse
Affiliation(s)
- Ya-Nan Ma
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Si-Rong Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li Yang
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Juan Wu
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ya-Rong Wang
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li-Jia Bao
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li Ma
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Qing-Qiu Wu
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Zhen-Hai Wang
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
- Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China.
- Neurology Center, Ningxia Medical University General Hospital, Yinchuan, Ningxia, China.
| |
Collapse
|
14
|
Zhao Z, Wei TT, Zhang WX, Zhang SS, Wu R, Li F, Yang H, Zhang Q, Xi J, Zhou Y, Wang T, Du J, Lu QB, Ge Q. Association of homoarginine with arginine and disease severity in COVID-19 patients. Amino Acids 2025; 57:24. [PMID: 40332615 PMCID: PMC12058869 DOI: 10.1007/s00726-025-03453-6] [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: 01/22/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
This study explored the relationship between the concentrations of homoarginine and arginine and between homoarginine concentration and laboratory parameters in coronavirus disease 2019 (COVID-19) patients with different severity to demonstrate the role of homoarginine in the progress of COVID-19. The laboratory-confirmed COVID-19 patients were included from Peking University Third Hospital during December 2022 to January 2023. Serum, urine, and stool samples were collected from the patients and detected by liquid chromatography-mass spectrometry. Totally 46 patients were recruited, including 18 in the mild group, 19 in the severe group, and 9 fatal. The concentration of homoarginine was positively correlated with the concentration of arginine in serum (r = 0.50), urine (r = 0.55), and stool samples (r = 0.39), respectively (all P < 0.001). The serum concentration and urine concentration of homoarginine were lower in severe patients than in mild patients (both P < 0.05). 13 indicators reflecting immunity and coagulation, including but not limited to T cell, white blood cell, natural killer cell, interleukin 6 (IL-6), and IL-8, had statistically significant correlations with both disease severity and the homoarginine concentration. Patients with hypertension were significantly associated with the decreased serum homoarginine (odds ratio 10.905, 95% confidence interval 1.454 - 137.144). Our results suggest that the homoarginine plays a role in the progress of COVID-19, which may be achieved by influencing arginine metabolism.
Collapse
Affiliation(s)
- Zhiling Zhao
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Ting-Ting Wei
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Wan-Xue Zhang
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Shan-Shan Zhang
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Rui Wu
- Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Fei Li
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Han Yang
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Qiang Zhang
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Jingjing Xi
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Yiguo Zhou
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China
| | - Tiehua Wang
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Juan Du
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Qing-Bin Lu
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China.
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China.
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
- Center for Infectious Disease and Policy Research and Department of Laboratorial of Science and Technology, School of Public Health, Peking University, No 38 Xue-Yuan Rd, Haidian District, Beijing, 100191, China.
| | - Qinggang Ge
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China.
| |
Collapse
|
15
|
Backman E, Gröning R, Lind A, Granvik C, Eilers H, Lange A, Ahlm C, Cajander S, Forsell MNE, Normark J, Urban CF. Elevated Plasma Levels of NET Components in Men with Severe COVID-19 Correlates to Increased Amounts of IL-18. Eur J Immunol 2025; 55:e202451546. [PMID: 40346759 PMCID: PMC12064869 DOI: 10.1002/eji.202451546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/12/2025]
Abstract
Severe COVID-19 disease is accompanied by high plasma levels of proinflammatory, prothrombotic NETs, and NET-inducing cytokine IL-18. We found that both, IL-18 and NETs, are elevated in men with severe disease, but not in women of the same category. Our findings warrant further investigation of sex-related differences in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Emelie Backman
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| | - Remigius Gröning
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| | - Alicia Lind
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
- Department of Diagnostics and InterventionUmeå UniversityUmeåSweden
| | | | - Hinnerk Eilers
- Laboratory MedicineRegion VästerbottenNorrland University HospitalUmeåSweden
| | - Anna Lange
- Department of Infectious DiseasesFaculty of Medicine and HealthÖrebro UniversityÖrebroSweden
| | - Clas Ahlm
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| | - Sara Cajander
- Department of Infectious DiseasesFaculty of Medicine and HealthÖrebro UniversityÖrebroSweden
| | - Mattias N. E. Forsell
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| | - Johan Normark
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
| | - Constantin F. Urban
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| |
Collapse
|
16
|
Gotelli E, Campitiello R, Pizzorni C, Sammorì S, Aitella E, Ginaldi L, De Martinis M, Carubbi F, Di Ruscio E, Cuomo G, Martinelli E, Marrone S, De Angelis R, Giuggioli D, Guiducci S, Ingegnoli F, Riccieri V, Sebastiani M, Sulli A, Smith V, Cutolo M. Multicentre retrospective detection of nailfold videocapillaroscopy abnormalities in long covid patients. RMD Open 2025; 11:e005469. [PMID: 40295119 PMCID: PMC12039021 DOI: 10.1136/rmdopen-2025-005469] [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: 01/17/2025] [Accepted: 04/06/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND SARS-CoV-2 induces acute non-specific endothelial/microvascular alterations that have been identified by nailfold videocapillaroscopy (NVC). Details on NVC abnormalities in long covid (LC) patients (pts) are unknown. METHODS LC pts without and with systemic sclerosis (non-SSc-LC and SSc-LC), recovered COVID-19 (RC) pts that did not develop LC and healthy matched control subjects (CNT) that underwent NVC examinations were evaluated in a multicentre national study from the Capillaroscopy and Microcirculation in Rheumatic Diseases Study Group of the Italian Society of Rheumatology. Retrospective collection was performed for demographic data, course of SARS-CoV-2 infection, comorbidities, concomitant drugs. NVC alterations were quantified by validated scores. Pre-COVID-19 and post-COVID-19 microvascular status was analysed by NVC. RESULTS 62 non-SSc-LC pts (49 female/13 male, 51±16 years old), 24 SSc-LC pts (21 female/3 male, 59±17 years old), 23 RC pts (18 female/5 male, 51±18 years old) and 84 CNT (68 female/16 male, 52±12 years old) were analysed. Non-SSc-LC pts showed significantly more dilated capillaries (p<0.01, p multivariate<0.01), microhaemorrhages (p=0.01, p multivariate<0.05), abnormal shapes (p<0.05, p multivariate<0.05) than CNT and of note, lower mean capillary number per linear millimetre (p<0.01, p multivariate<0.01) than both RC pts and CTN (p<0.01, p multivariate<0.05).Of highest interest, 16 non-SSc-LC pts showed statistically significantly more dilated capillaries (p<0.05) and microhaemorrhages (p<0.05) in NVC examinations after COVID-19, compared with pre-COVID-19 status.Similarly, SSc-LC pts (24) showed significantly lower capillary density (p=0.01) and more dilated capillaries (p<0.01) in NVC examinations after COVID-19, compared with pre-COVID-19 status. CONCLUSIONS LC pts show more microvascular alterations at NVC as compared with RC patients and CNT, which may contribute to the pathogenesis of persistent organ/systems dysfunction.
Collapse
Affiliation(s)
- Emanuele Gotelli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Rosanna Campitiello
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Sammorì
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Ernesto Aitella
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- Allergy and Clinical Immunology Unit, G. Mazzini Hospital, Teramo, Italy
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
- Long-Term Care Unit, Maria SS dello Splendore Hospital, Giulianova, Italy
| | - Francesco Carubbi
- Internal Medicine and Nephrology Division, Department of Clinical Medicine, Life, Health and Environmental Sciences, San Salvatore Hospital, L'Aquila, Italy
| | - Evy Di Ruscio
- Internal Medicine and Nephrology Division, Department of Clinical Medicine, Life, Health and Environmental Sciences, San Salvatore Hospital, L'Aquila, Italy
| | - Giovanna Cuomo
- Department of Precision Medicine, University Hospital Luigi Vanvitelli, Naples, Italy
| | - Emanuela Martinelli
- Department of Precision Medicine, University Hospital Luigi Vanvitelli, Naples, Italy
| | - Sabrina Marrone
- Department of Precision Medicine, University Hospital Luigi Vanvitelli, Naples, Italy
| | - Rossella De Angelis
- Rheumatology Unit, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Carlo Urbani Hospital, Jesi, Italy
- IRCCS INRCA, Ancona, Italy
| | - Dilia Giuggioli
- Rheumatology Unit, Department of Medical and Surgical Sciences for Children and Adults, UNIMORE, Modena, Italy
| | - Serena Guiducci
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesca Ingegnoli
- Clinical Rheumatology Unit, Department of Clinical Science and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Valeria Riccieri
- Department of Internal Medicine, Anesthesiology and Cardiovascular Sciences, University of Rome La Sapienza, Rome, Italy
| | - Marco Sebastiani
- Rheumatology Unit, AUSL Piacenza, Piacenza, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Alberto Sulli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Vanessa Smith
- Department of Rheumatology, Ghent University, Ghent, Belgium
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, Flemish Institute for Biotechnology (VIB), Ghent, Belgium
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
17
|
Modiga A, Butiurca VO, Boeriu CM, Truta TS, Turucz E, Halațiu VB, Rodean IP, Russu PC, Gherghinescu MC, Molnar C. Pathophysiological Mechanisms Linking COVID-19 and Acute Surgical Abdomen: A Literature Review. Life (Basel) 2025; 15:707. [PMID: 40430138 PMCID: PMC12113513 DOI: 10.3390/life15050707] [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: 03/18/2025] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Acute surgical abdomen is characterized by intense, sudden abdominal pain due to intra-abdominal conditions requiring prompt surgical intervention. The coronavirus disease 2019 (COVID-19) pandemic has led to various complications related to the disease's complex pathophysiological mechanisms, hence the hypothesis of COVID-19-induced acute abdominal surgical pathologies. The connection between acute surgical abdomen and COVID-19 involves two primary mechanisms. First, there is the presence of angiotensin-converting enzyme 2 (ACE2) receptors in multiple abdominal organs. This facilitates the cytokine storm through direct viral injury and inflammation. Second, the hypercoagulable state induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) increases the thrombotic risk within abdominal vessels, which can subsequently lead to ischemia. ACE2 receptors are notably expressed in the gastric, duodenal, and rectal epithelium, with SARS-CoV-2 viral RNA and nucleocapsid proteins detected in these tissues. The inflammatory response results in significant endothelial damage, activating coagulation pathways that cause monocellular infiltration, lymphocytic inflammation, and uncontrolled coagulation. These findings highlight the need for further research to clarify how COVID-19 leads to acute abdominal pathologies. Understanding these mechanisms is vital for improving clinical management and patient outcomes during future health crises and in the aftermath of the pandemic.
Collapse
Affiliation(s)
- Andrei Modiga
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Vlad-Olimpiu Butiurca
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Cristian Marius Boeriu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Teodora Sorana Truta
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Emilia Turucz
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Vasile-Bogdan Halațiu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
| | - Ioana-Patricia Rodean
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
| | - Paul Cristian Russu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Mircea Constantin Gherghinescu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Călin Molnar
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| |
Collapse
|
18
|
Smadja DM, Massonnaud CR, Philippe A, Rosa M, Luneau S, Rauch A, Peiffer-Smadja N, Gagneux-Brunon A, Poissy J, Gruest M, Ung A, Pourcher V, Raffi F, Piroth L, Bouiller K, Esperou H, Delmas C, Belhadi D, Diallo A, Saillard J, Dechanet A, Mercier N, Dupont A, Lescure FX, Goehringer F, Jaureguiberry S, Danion F, Tolsma V, Cabie A, Courjon J, Leroy S, Mootien J, Mourvillier B, Gallien S, Lanoix JP, Botelho-Nevers E, Wallet F, Richard JC, Reuter J, Gaymard A, Greil R, Martin-Blondel G, Andrejak C, Yazdanpanah Y, Burdet C, Diehl JL, Hites M, Ader F, Susen S, Mentré F, Dupont A. sST2 is a key outcome biomarker in COVID-19: insights from discovery randomized trial. Sci Rep 2025; 15:14348. [PMID: 40274842 PMCID: PMC12022249 DOI: 10.1038/s41598-025-95122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
We investigated whether baseline levels of biomarkers related to endotheliopathy, thromboinflammation, and fibrosis were associated with clinical outcomes in hospitalized COVID-19 patients. We analyzed the associations between baseline levels of 21 biomarkers and time to hospital discharge and change in NEWS-2 score in patients from DisCoVeRy trial. We fitted multivariate models adjusted for baseline ISARIC 4C score, disease severity, D-dimer values, and treatment regimen. Between March 22 and June 29, 2020, 603 participants were randomized; 454 had a sample collected at baseline and analyzed. The backward selection of multivariate models showed that higher baseline levels of soluble suppressor of tumorigenicity 2 (sST2) and nucleosomes were statistically associated with a lower chance of hospital discharge before day 29 (sST2: aHR 0.24, 95% CI [0.15-0.38], p < 10-9; nucleosomes: aHR 0.62, 95% CI [0.48-0.81], p < 10-3). Likewise, higher levels of baseline sST2 were statistically associated with lower changes in the NEWS-2 score between baseline and day 15 (adjusted beta 4.47, 95% CI [2.65-6.28], p < 10-5). Moreover, we evaluated sST2 involvement in a confirmation cohort (SARCODO study, 103 patients) and found that elevated baseline sST2 levels were significantly associated with lower rates of hospital discharge before day 29 and a higher model performance (AUC at day 29 of 92%) compared to models without sST2. sST2 emerged as an independent predictor of clinical outcomes in two large cohort of hospitalized COVID-19 patients, warranting further investigation to elucidate its role in disease progression and potential as a therapeutic target.
Collapse
Affiliation(s)
- David M Smadja
- Innovative Therapies in Hemostasis, INSERM, University Paris Cité, 75006, Paris, France.
- Hematology Department, AP-HP Centre, Georges Pompidou European Hospital, Université Paris Cité, 20 Rue Leblanc, 75015, Paris, France.
| | - Clément R Massonnaud
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | - Aurélien Philippe
- Innovative Therapies in Hemostasis, INSERM, University Paris Cité, 75006, Paris, France
- Hematology Department, AP-HP Centre, Georges Pompidou European Hospital, Université Paris Cité, 20 Rue Leblanc, 75015, Paris, France
| | - Mickael Rosa
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - Sophie Luneau
- Hematology Department, AP-HP Centre, Georges Pompidou European Hospital, Université Paris Cité, 20 Rue Leblanc, 75015, Paris, France
| | - Antoine Rauch
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - Nathan Peiffer-Smadja
- IAME, INSERM, Université de Paris, 75018, Paris, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - Amandine Gagneux-Brunon
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC-INSERM 1408, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Julien Poissy
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - Maxime Gruest
- Hematology Department, AP-HP Centre, Georges Pompidou European Hospital, Université Paris Cité, 20 Rue Leblanc, 75015, Paris, France
| | - Alexandre Ung
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - Valérie Pourcher
- Assistance Publique - Hôpitaux de Paris, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Service de Maladies Infectieuses et Tropicales, INSERM 1136, Institut Pierre Louis d'Épidémiologie et de Santé Publique, Sorbonne Université, 75013, Paris, France
| | - François Raffi
- Department of Infectious Diseases, CIC 1413, INSERM, CHU Nantes, Nantes Université, Nantes, France
| | - Lionel Piroth
- Infectious Diseases Department, CHU, Dijon, and INSERM CIC 1432, Université de Bourgogne-Franche Comté, Dijon, France
| | - Kévin Bouiller
- UMR-CNRS 6249 Chrono-Environnement, Department of Infectious and Tropical Diseases, CHU Besançon, Université de Franche-Comté, 25000, Besançon, France
| | - Hélène Esperou
- Institut de Santé Publique, INSERM, Pôle Recherche Clinique, Paris, France
| | | | - Drifa Belhadi
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | - Alpha Diallo
- ANRS, Maladies Infectieuses Émergentes, Paris, France
| | | | - Aline Dechanet
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | | | - Axelle Dupont
- IAME, INSERM, Université de Paris, 75018, Paris, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - François-Xavier Lescure
- IAME, INSERM, Université de Paris, 75018, Paris, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - François Goehringer
- Service de Maladies Infectieuses et Tropicales, CHRU-Nancy, Université de Lorraine, 54000, Nancy, France
| | - Stéphane Jaureguiberry
- AP-HP, Service Des Maladies Infectieuses, Hôpital Bicêtre, 94270, Le Kremlin Bicêtre, France
- AP-HP, Centre National de Référence du Paludisme, Paris, France
| | - François Danion
- Service des Maladies Infectieuses et Tropicales, Hôpitaux Universitaires de Strasbourg, 67091, Strasbourg, France
| | - Violaine Tolsma
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Annecy Genevois, 74374, Annecy, France
| | - André Cabie
- PCCEI, Inserm, EFS, Univ Montpellier, Univ Antilles, Montpellier, France
- Service des Maladies Infectieuses et Tropicales, Inserm CIC1424, CHU de Martinique, Martinique, France
| | - Johan Courjon
- Service des Maladies Infectieuses et Tropicales, CHU de Nice, Nice, France
- U1065, Centre Méditerranéen de Médecine Moléculaire, C3M, Virulence Microbienne et Signalisation Inflammatoire, INSERM, Université Côte d'Azur, Nice, France
| | - Sylvie Leroy
- Fédération Hospitalo-Universitaire OncoAge, Nice, France
- Département de Pneumologie et d'Oncologie, CHU de Nice, 06000, Nice, France
- CNRS UMR 7275, IPMC, Université Côte d'Azur, Sophia Antipolis, France
| | - Joy Mootien
- Service de Réanimation Médicale, Groupe Hospitalier de la Région Mulhouse Sud-Alsace, Mulhouse, France
| | - Bruno Mourvillier
- CHU de Reims, Service de Réanimation Médicale, Université de Reims Champagne-Ardenne, Reims, France
| | - Sébastien Gallien
- Service d'Immunologie et Maladies Infectieuses, AP-HP, Hôpital Henri Mondor, 94000, Créteil, France
- INSERM U955, Université Paris-Est Créteil, 94000, Créteil, France
| | - Jean-Philippe Lanoix
- Service de Maladies Infectieuses et Tropicales, CHU Amiens-Picardie, 80000, Amiens, France
- AGIR UR UPJV 4294, CURS, Université Picardie Jules Verne, 80000, Amiens, France
| | - Elisabeth Botelho-Nevers
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC-INSERM 1408, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Florent Wallet
- Département de Soins Intensifs, Hospices Civils de Lyon, Hôpital Lyon-Sud Pierre-Bénite, 69000, Lyon, France
| | - Jean-Christophe Richard
- Service de Réanimation Médicale, Hospices Civils de Lyon, Hôpital de la Croix-Rousse, 69000, Lyon, France
- CREATIS, CNRS UMR5220, INSERM U1044, INSA, Université Lyon I, 69000, Lyon, France
| | - Jean Reuter
- Service de Réanimation-Soins Intensifs, Centre Hospitalier de Luxembourg, 1210, Luxembourg, Luxembourg
| | - Alexandre Gaymard
- Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des Virus Respiratoires France Sud, Hospices Civils de Lyon, 69317, Lyon, France
- Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, 69372, Lyon, France
| | - Richard Greil
- Department of Internal Medicine III With Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University Salzburg, 5020, Salzburg, Austria
| | - Guillaume Martin-Blondel
- Service des Maladies Infectieuses et Tropicales, CHU de Toulouse, 31320, Toulouse, France
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity) INSERM UMR1291 - CNRS UMR5051, Université Toulouse III, 31320, Toulouse, France
| | - Claire Andrejak
- Département de Pneumologie, CHU d'Amiens, 80000, Amiens, France
| | - Yazdan Yazdanpanah
- IAME, INSERM, Université de Paris, 75018, Paris, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - Charles Burdet
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | - Jean-Luc Diehl
- Innovative Therapies in Hemostasis, INSERM, University Paris Cité, 75006, Paris, France
- Intensive Care Unit, AP-HP Centre Université Paris Cité, Georges Pompidou European Hospital, 75015, Paris, France
| | - Maya Hites
- Clinic of Infectious Diseases, Hôpital Universitaire de Bruxelles (H.U.B), 1070, Brussels, Belgium
| | - Florence Ader
- Département des Maladies Infectieuses et Tropicales, Hospices Civils de Lyon, Lyon, France
- Département des Maladies Infectieuses et Tropicales, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Sophie Susen
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - France Mentré
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | - Annabelle Dupont
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Santibanez V, Mathur A, Zatakia J, Ng N, Cohen M, Bagiella E, Brown SA, Rosas IO, Patel NM, Olson A, Li P, Padilla M. Early nintedanib deployment in COVID-19 interstitial lung disease (ENDCOV-I): study protocol of a randomised, double-blind, placebo-controlled trial. BMJ Open Respir Res 2025; 12:e002323. [PMID: 40216412 PMCID: PMC12185894 DOI: 10.1136/bmjresp-2024-002323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
INTRODUCTION In December 2019, the novel SARS-CoV-2 triggered a global pneumonia outbreak, leading to millions of deaths worldwide. A subset of survivors faces increased morbidity and mortality, particularly due to subacute lung injury evolving to chronic fibrosing interstitial lung disease. While nintedanib, a tyrosine-kinase inhibitor, shows promise in treating progressive fibrotic lung disease, limited randomised trial data exists for post-COVID-19-induced lung injury. We hypothesise that treatment with nintedanib may attenuate advancement to the fibrotic stages, offering a potential avenue for improving outcomes in this specific patient subset. METHODS AND ANALYSIS We describe the design of a multicentre, randomised, double-blind, placebo-controlled trial involving approximately 170 patients with subacute lung injury secondary to COVID-19, who required respiratory support with oxygen supplementation. Patients are randomised by site and disease phenotype (fibrotic vs non-fibrotic) in a 1:1 ratio to either oral nintedanib or placebo. Patients will be followed for 180 days. The primary endpoint is to assess change from baseline in forced vital capacity (FVC, mL) at 180 days. Secondary objectives include change in FVC (mL) at 90 days; diffusing capacity of carbon monoxide (% of predicted) and 6-min walk test (feet) at 180 days; and mortality at 90 and 180 days. Qualitative and quantitative changes in high-resolution computerised tomography (HRCT), change in patient-reported outcome measures (PROMs) and safety endpoints will also be assessed. Analysis will be performed according to the intention-to-treat principle. ETHICS AND DISSEMINATION The study is conducted in accordance with the Good Clinical Practices as outlined by the Food and Drug Administration and the Declaration of Helsinki 2008. This study received approval from participating sites' Institutional Review Boards and committees, including The Ethics Committee of the Medical Board at the Mount Sinai Hospital (ID: HS#20-01166). The Independent Oversight Committee oversees study conduct, data and patient safety for the duration of the study investigation. The trial details presented align with the trial protocol V.8. (April 2022). Results will be presented at national and international conferences, published in a peer-reviewed journal and disseminated to patients, funders and researchers on data analysis completion. TRIAL REGISTRATION NUMBER NCT04619680. First posted 6 November 2020.
Collapse
Affiliation(s)
| | - Aditi Mathur
- Pulmonary, Hackensack Meridian Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Jigna Zatakia
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nicole Ng
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michele Cohen
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emilia Bagiella
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Nina M Patel
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Amy Olson
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Peide Li
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Maria Padilla
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
22
|
Maier CL, Nakahara H, Barker NA, Auld SC, Truong AD, Friend S, Caridi-Scheible M, Connor M, Gaddh M, Cobb J, Polly DM, Guarner J, Powell C, Kempton CL, Daniels L, Wynn AT, Sniecinski R, Duncan A, Roback J, Masud T, Conlon S, Wade J, Wong A, Verkerke H, Zerra PE, Butler H, Sullivan HC, Easley KA, Josephson CD, Stowell SR. Therapeutic plasma exchange for fibrinogen-associated hyperviscosity: results of the COVID-19 PLasma EXchange (COPLEX) randomized controlled trial. J Thromb Haemost 2025; 23:1393-1400. [PMID: 39746400 DOI: 10.1016/j.jtha.2024.12.021] [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: 07/11/2024] [Revised: 11/01/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Therapeutic plasma exchange (TPE) is the primary intervention for treating symptomatic hyperviscosity from hypergammaglobulinemia, yet its efficacy for treating hyperviscosity related to hyperfibrinogenemia is unclear. OBJECTIVES Define the safety and efficacy of TPE for critically ill COVID-19 patients with elevated blood viscosity from hyperfibrinogenemia. METHODS We performed a prospective randomized controlled trial in critically ill COVID-19 patients in a single US healthcare system. Patients with hyperfibrinogenemia (>800 mg/dL) or elevated plasma viscosity (2.3-3.5 centipoise [cP]) were randomized to receive TPE on 2 consecutive days or continued standard of care (SOC). RESULTS Twenty participants were enrolled, with 10 receiving TPE and 10 receiving SOC alone. Mean (±SEM) plasma viscosity decreased significantly from 2.35 cP (±0.12) to 1.61 cP (±0.03) in the TPE group and was unchanged in the SOC group (2.47 cP [±0.11] to 2.47 cP [±0.15]). Mean fibrinogen decreased from 934.0 mg/dL (±25.1) to 359.1 mg/dL (±22.5) after TPE vs from 859.6 mg/dL (±57.6) to 807.3 mg/dL (±63.1) in SOC. There was no significant difference in 28-day all-cause mortality between groups, with 2 deaths in the TPE cohort and 5 deaths in the SOC cohort (P = .13). No serious safety events related to TPE were reported. TPE significantly decreased biomarkers of inflammation (erythrocyte sedimentation rate and C-reactive protein) and endothelial activation (von Willebrand factor and factor VIII) but not hemostatic activation (prothrombin fragment 1.2, thrombin-antithrombin complex, and fibrin monomer) or immunoglobulin (IgG and IgM) levels. CONCLUSION TPE is safe and effective for normalizing elevated blood viscosity from hyperfibrinogenemia in COVID-19 patients. Additional studies are needed to determine the impact of TPE on overall patient outcomes, including in those with non-COVID-19 conditions associated with hyperfibrinogenemia.
Collapse
Affiliation(s)
- Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA.
| | - Hirotomo Nakahara
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicholas A Barker
- Department of Pharmaceutical Services, Emory Healthcare, Atlanta, Georgia, USA
| | - Sara C Auld
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA; Departments of Epidemiology and Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | | | - Sarah Friend
- Department of Hematology and Medical Oncology, Emory School of Medicine, Atlanta, Georgia, USA
| | | | - Michael Connor
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Manila Gaddh
- Department of Hematology and Medical Oncology, Emory School of Medicine, Atlanta, Georgia, USA
| | - Jason Cobb
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Derek M Polly
- Department of Pharmaceutical Services, Emory Healthcare, Atlanta, Georgia, USA
| | - Jeannette Guarner
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Cindy Powell
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Christine L Kempton
- Department of Hematology and Medical Oncology, Emory School of Medicine, Atlanta, Georgia, USA
| | - Lisa Daniels
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - A Thanushi Wynn
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Roman Sniecinski
- Department of Anesthesiology, Emory School of Medicine, Atlanta, Georgia, USA
| | - Alexander Duncan
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - John Roback
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Tahsun Masud
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Shaun Conlon
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Jenna Wade
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Andrew Wong
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Hans Verkerke
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Patricia E Zerra
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Hailly Butler
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - H Cliff Sullivan
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Kirk A Easley
- Department of Biostatistics and Bioinformatics, Emory Rollins School of Public Health, Atlanta, Georgia, USA
| | - Cassandra D Josephson
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Sean R Stowell
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Zhang B, Yao Z, Li P, Niu G, Yan Z, She K, Cheng G, Yang M. Causal Relationships of Circulating Inflammatory Proteins and Portal Vein Thrombosis: A Mendelian Randomization Study. Semin Thromb Hemost 2025; 51:272-278. [PMID: 39293485 DOI: 10.1055/s-0044-1790259] [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: 09/20/2024]
Abstract
Portal vein thrombosis (PVT) is commonly encountered in patients with cirrhosis, challenging our understanding of its development, particularly the ambiguous contribution of inflammation. This study utilized Mendelian randomization (MR) to explore the causal impact of circulating inflammatory markers on PVT.Employing a two-sample MR framework, we merged genome-wide association study (GWAS) meta-analysis findings of 91 inflammation-associated proteins with independent PVT data from the FinnGen consortium's R10 release. A replication analysis was performed using a distinct GWAS dataset from the UK Biobank. Inverse variance weighting, MR-Egger regression, weighted median estimator, and Mendelian Randomization Pleiotropy RESidual Sum and Outlier were used for analysis, supplemented by multivariable MR (MVMR) to adjust for cirrhosis effects.Findings indicate a significant inverse association between the genetically inferred concentration of eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and PVT risk, evidenced by an odds ratio (OR) of 0.37 (95% confidence interval [CI]: 0.21-0.67; p = 9.2 × 10-4; adjusted for multiple testing p = 0.084). This association was corroborated in the replication phase (OR = 0.39, 95% CI: 0.17-0.93; p = 0.03) and through MVMR analysis (OR = 0.34, 95% CI: 0.15-0.79; p = 0.012). Sensitivity analyses disclosed no evidence of heterogeneity or pleiotropy.Our investigation emphasizes the 4E-BP1 as a protective factor against PVT, underscoring its potential relevance in understanding PVT pathogenesis and its implications for diagnosis and therapy.
Collapse
Affiliation(s)
- Bihui Zhang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Ziping Yao
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Pengyu Li
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Guochen Niu
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Ziguang Yan
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Kang She
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Gong Cheng
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Min Yang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
24
|
Kruger A, Joffe D, Lloyd-Jones G, Khan MA, Šalamon Š, Laubscher GJ, Putrino D, Kell DB, Pretorius E. Vascular Pathogenesis in Acute and Long COVID: Current Insights and Therapeutic Outlook. Semin Thromb Hemost 2025; 51:256-271. [PMID: 39348850 PMCID: PMC11906225 DOI: 10.1055/s-0044-1790603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Long coronavirus disease 2019 (COVID-19)-a postacute consequence of severe acute respiratory syndrome coronavirus 2 infection-manifests with a broad spectrum of relapsing and remitting or persistent symptoms as well as varied levels of organ damage, which may be asymptomatic or present as acute events such as heart attacks or strokes and recurrent infections, hinting at complex underlying pathogenic mechanisms. Central to these symptoms is vascular dysfunction rooted in thrombotic endothelialitis. We review the scientific evidence that widespread endothelial dysfunction (ED) leads to chronic symptomatology. We briefly examine the molecular pathways contributing to endothelial pathology and provide a detailed analysis of how these cellular processes underpin the clinical picture. Noninvasive diagnostic techniques, such as flow-mediated dilation and peripheral arterial tonometry, are evaluated for their utility in identifying ED. We then explore mechanistic, cellular-targeted therapeutic interventions for their potential in treating ED. Overall, we emphasize the critical role of cellular health in managing Long COVID and highlight the need for early intervention to prevent long-term vascular and cellular dysfunction.
Collapse
Affiliation(s)
- Arneaux Kruger
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - David Joffe
- Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, Australia
- World Health Network, Cambridge, Massachusetts
| | - Graham Lloyd-Jones
- Department of Radiology, Salisbury District Hospital, Salisbury NHS Foundation Trust, United Kingdom
| | - Muhammed Asad Khan
- World Health Network, Cambridge, Massachusetts
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | | | | | - David Putrino
- Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, Australia
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- World Health Network, Cambridge, Massachusetts
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
25
|
Hu B, Jiang J, Pan W, Chung CS, Gray C, Chen Y, Guo J, Ayala A. V-domain Ig Suppressor of T cell Activation Expression During Hemorrhage or Sepsis-Induced Acute Respiratory Distress Syndrome: Insights From a Mouse Model. J Surg Res 2025; 308:73-85. [PMID: 40086004 DOI: 10.1016/j.jss.2025.02.001] [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: 07/19/2024] [Revised: 01/15/2025] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Acute respiratory distress syndrome (ARDS) is a life-threatening pulmonary condition with significant mortality, largely due to a lack of therapeutic interventions grounded in its molecular pathophysiology. Immune checkpoint regulators, such as the V-domain Ig Suppressor of T cell Activation (VISTA), may provide novel immunotherapeutic strategies for ARDS by modulating the immune response, a concept extensively explored in cancer and autoimmune diseases. Investigating VISTA in the context of ARDS could unveil new therapeutic avenues. METHODS We used a mouse model of indirect ARDS by subjecting C57BL/6J mice to hemorrhage followed by cecal ligation and puncture. Systemic and localized inflammatory conditions were assessed using samples from blood, lung, and peritoneal fluid. Lung pathology was quantified by scoring hematoxylin and eosin-stained sections. Flow cytometry, enzyme-linked immunosorbent assay, and reverse transcription-polymerase chain reaction analyses concentrated on macrophages, neutrophils, endothelial cells, and epithelial cells to elucidate VISTA expression patterns. RESULTS Hemorrhage or cecal ligation and puncture-treated mice exhibited hallmark symptoms of indirect ARDS, including elevated levels of inflammatory cytokines and chemokines. Notably, VISTA expression was substantially upregulated on various cell types, including blood monocytes, lung macrophages, and both circulating and lung-infiltrating neutrophils, as well as on pulmonary epithelial cells and endothelial cells. CONCLUSIONS Our model replicates critical inflammatory and physiologic changes leading to ARDS, with the elevated expression of VISTA on immune and parenchymal cells suggesting its central involvement in lung injury. The findings propose VISTA as both a potential biomarker for lung damage and as a promising target for ARDS therapy.
Collapse
Affiliation(s)
- Baoji Hu
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China; Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China; Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island
| | - Jihong Jiang
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Pan
- Division of Infectious Diseases, Rhode Island Hospital/ The Warren Alpert Medical School at Brown University, Providence, Rhode Island
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island
| | - Chyna Gray
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island
| | - Yaping Chen
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island
| | - Jianrong Guo
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China; Department of Anesthesiology, Shanghai Gongli Hospital, Naval Military Medical University, Shanghai, China.
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island.
| |
Collapse
|
26
|
Zhou F, Guo Y, Li W, Hu Y, Yang L, Fu S, Bao X, Tong H, Ye Y, Ding Z. Tetrastigma hemsleyanum polysaccharide protects against "two-hit" induced severe pneumonia via TLR4/NF-κB signaling pathway. Int J Biol Macromol 2025; 303:140639. [PMID: 39909274 DOI: 10.1016/j.ijbiomac.2025.140639] [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: 11/10/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Severe pneumonia, frequently accompanied by cytokine storms, stands as a perilous respiratory condition with alarmingly high mortality rates. Tetrastigma hemsleyanum polysaccharide (THP), a pivotal constituent derived from Tetrastigma hemsleyanum Diels et Gilg (TH), has demonstrated efficacy in treating lung inflammation. However, its precise efficacy and underlying mechanisms in the context of severe pneumonia remain elusive. Our research aims to elucidate THP's protective effects in a "two-hit" severe pneumonia model. Our observations indicate that THP administration markedly shields the lungs from injury, reduces pulmonary apoptosis, balances the formation of immune thrombus and alleviates oxidative stress in pneumonia-induced mice. Furthermore, THP significantly decreases the levels of pro-inflammatory cytokines, suggesting its robust anti-inflammatory capabilities. Notably, THP also plays a crucial role in normalizing gut microbiota imbalance, which is vital in the pathogenesis of severe pneumonia. Metabolomic analysis further validates THP's restorative effects on plasma metabolites, indicating its involvement in regulating energy metabolism and immune homeostasis. Mechanistically, THP targets the TLR4/NF-κB signaling pathway, a core mediator of inflammation, thereby dampening the inflammatory cascade. In summary, our findings underscore that THP, through its multifaceted actions targeting inflammation, oxidative stress, immune thrombus formation, gut microbiota regulation, and metabolic modulation, emerges as a promising therapeutic approach for severe pneumonia. This study provides invaluable insights into the potential applications of natural polysaccharides in treating severe pneumonia and highlights the significance of the TLR4/NF-κB pathway in the disease's progression.
Collapse
Affiliation(s)
- Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ying Guo
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wenxuan Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yiwen Hu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Liu Yang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Siyu Fu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Xiaodan Bao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongbin Tong
- Hangzhou HealthBank Medical Laboratory Co., Ltd., Hangzhou, Zhejiang 310053, China
| | - Yujian Ye
- Department of Dermatology, Third People's Hospital of Hangzhou, Hangzhou, China.
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
27
|
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
|
28
|
Cui N, Wang J, Feng X, Zhang L, Yang Y. Deep vein thrombosis in severe community-acquired pneumonia patients undergoing thromboprophylaxis: Prevalence, risk factors, and outcome. Thromb J 2025; 23:23. [PMID: 40075406 PMCID: PMC11905501 DOI: 10.1186/s12959-025-00706-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Even with adherence to thromboprophylaxis recommended by guidelines, the incidence of deep vein thrombosis (DVT) remains high among patients with severe community-acquired pneumonia (SCAP). There is an urgent need to identify the risk factors for DVT in these patients to optimize preventive strategies. STUDY DESIGN AND METHODS We retrospectively enrolled 309 adults with SCAP admitted to Beijing Chao-Yang Hospital between 1 January 2015 and 30 June 2023. All patients received guideline-recommended thromboprophylaxis and lower extremity venous compression ultrasound scanning. Clinical characteristics, including demographic information, clinical history, vital signs, laboratory findings, treatments, complications, and outcomes, were analyzed for patients with and without DVT in these two cohorts. RESULTS Of the 309 patients, 110 (35.6%) developed 1ower extremity DVT. There was no significant difference in the incidence of DVT among the different prophylactic measures (P = 0.393). Multivariate logistic regression analysis showed an association between a history of VTE (OR, 13.388, 95% CI: 2.179 ~ 82.257; P = 0.005), bedridden time > 3 days (OR, 17.672, 95% CI: 5.686 ~ 54.929; P < 0.001), D-dimer levels ≥ 1.0 µg/mL (OR, 2.109, 95% CI: 1.018 ~ 4.372; P = 0.045), LDH levels ≥ 400 U/L (OR, 2.548, 95% CI: 1.308 ~ 4.965; P = 0.006), IMV (OR, 2.479, 95% CI: 1.233 ~ 4.986; P = 0.011) and the occurrence of DVT. A new prediction model, including history of VTE, bedridden time, D-dimer levels, LDH levels and IMV, showed a better performance in predicting DVT (AUC = 0.856; 95% CI: 0.766 ~ 0.921; sensitivity: 80.6%; specificity: 81.4%) than Padua prediction score (AUC = 0.666) and Caprini prediction score (AUC = 0.688) for patients with SCAP. The 30-day mortality and in-hospital mortality in the DVT group were significantly higher than those in the non-DVT group. CONCLUSIONS Even received guideline-recommended thromboprophylaxis, the prevalence of DVT among patients with SCAP remains unexpectedly high which is also associated with a poor prognosis. It is necessary to identify people at high risk of DVT early and refine the preventive strategies accordingly to improve patient outcomes.
Collapse
Affiliation(s)
- Na Cui
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlua, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Jing Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlua, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Xiaokai Feng
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlua, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Liming Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlua, Chaoyang District, Beijing, 100020, People's Republic of China.
| | - Yuanhua Yang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlua, Chaoyang District, Beijing, 100020, People's Republic of China.
| |
Collapse
|
29
|
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
|
30
|
Wang Z, Zhao L, Xie K. Development and validation of a nomogram to assess the occurrence of liver dysfunction in patients with COVID-19 pneumonia in the ICU. BMC Infect Dis 2025; 25:332. [PMID: 40065225 PMCID: PMC11892215 DOI: 10.1186/s12879-025-10684-1] [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: 08/12/2024] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
The global pandemic of novel coronavirus pneumonia (COVID-19) has resulted in millions of deaths over the past three years. As one of the most commonly affected extra-pulmonary organs, numerous studies have reported varying degrees of liver injury in a significant proportion of patients with COVID-19, particularly in severe and critically ill patients. Early prediction of liver dysfunction in hospitalized patients would facilitate the clinical management of COVID-19 and improve clinical prognosis, but reliable and valid predictive models are still lacking.MethodsWe collected data from 286 patients with RT-PCR confirmed COVID-19 admitted to various ICUs from the case system. These patients were randomly divided into a training cohort (50%) and a validation cohort (50%). In the training cohort, we first used ROC curves to measure the predictive efficiency of each of the variables for the development of liver damage during hospitalization in patients with COVID-19, followed by LASSO regression analysis to screen the variables for predictive models and logistic regression analysis to identify relevant risk factors. A nomogram based on these variables was created following the above model. Finally, the efficiency of the prediction models in the training and validation cohorts was assessed using AUC, consistency index (C index), calibration curves and Decision Curve Analysis.ResultsOut of a total of 80 parameters for COVID-19 patients admitted to the ICUs, 10 were determined to be significantly associated with the occurrence of liver dysfunction during hospitalization. Based on these predictors, further prediction models were used to construct and develop a nomogram that was offered for practical clinical application. The C-index of the column line graphs for the training and validation cohorts was 0.956 and 0.844 respectively. in addition, the calibration curves for the model showed a high degree of agreement between the predicted and actual incidence of liver dysfunction in patients with COVID-19.ConclusionBy developing a predictive model and associated nomogram, we predicted the incidence of liver dysfunction during hospitalization in patients with COVID-19 in the ICU. The model's predictive performance was determined in both the training and validation cohorts, contributing to the clinical management of COVID-19.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Lina Zhao
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China.
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifangaq, Weifang, Shandong, 261053, China.
| |
Collapse
|
31
|
Semina E, Popov V, Khabibullin N, Klimovich P, Sysoeva V, Kurilina E, Tsokolaeva Z, Tkachuk V, Rubina K. New evidence for T-cadherin in COVID-19 pathogenesis, endothelial dysfunction, and lung fibrosis. Front Cell Dev Biol 2025; 13:1476329. [PMID: 40109358 PMCID: PMC11920143 DOI: 10.3389/fcell.2025.1476329] [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: 08/05/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
The COVID-19 pandemic had an unprecedented impact on all aspects of human activity worldwide, frequently resulting in post-acute sequelae and affecting multiple organ systems. The underlying mechanisms driving both acute and post-acute manifestations of COVID-19 are still poorly understood, warranting further investigation for new targets. The study represents the first attempt to explore the role of T-cadherin in COVID-19 pathogenesis as well as its implications in pulmonary fibrosis and endothelial dysfunction. First, we revealed a significant decrease in T-cadherin expression in post-mortem lung samples from COVID-19 patients. This downregulated T-cadherin expression correlated with the elevated levels of VE-cadherin and reduced levels of β-catenin, suggesting a disruption in endothelial cell-cell contact integrity and function. Second, the reciprocal relation of T-cadherin and VE-cadherin expression was further confirmed using cultured human endothelial Ea.hy926 cells. T-cadherin overexpression caused a decrease in VE-cadherin mRNA expression in cultured endothelial cells providing additional evidence in favor of their interplay. Third, employing Cdh13 -/- mice, we unveiled the protective role of T-cadherin deficiency against bleomycin-induced lung fibrosis. Fourth, we demonstrated the mice lacking T-cadherin to have downregulated reactive oxygen species production and Nox2 mRNA expression in an angiotensin II-mediated endothelial dysfunction model. Our findings provide rationale for further studies into T-cadherin-mediated mechanisms in these processes.
Collapse
Affiliation(s)
- Ekaterina Semina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir Popov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | | | - Polina Klimovich
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Veronika Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Ella Kurilina
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Zoya Tsokolaeva
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Kseniya Rubina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
32
|
Jung C, Stueber T, Mirus M, Heubner L, Spieth PM. Anticoagulation in venovenous extracorporeal membrane oxygenation. Front Med (Lausanne) 2025; 12:1530411. [PMID: 40103791 PMCID: PMC11913846 DOI: 10.3389/fmed.2025.1530411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Venovenous extracorporeal membrane oxygenation (VV-ECMO) is a lifesaving therapy in severe acute respiratory distress syndrome (ARDS). Unfortunately, bleeding and thrombotic complications occur regularly due to coagulation disorders associated with the device, the underlying disease, and the anticoagulation management. To facilitate a personalized approach to hemostasis in individuals receiving ECMO support, it is essential to assess the coagulative state of the patient while simultaneously taking into account the underlying medical condition and administered therapies.
Collapse
Affiliation(s)
- Carolin Jung
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Thomas Stueber
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Martin Mirus
- Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Lars Heubner
- Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Peter Markus Spieth
- Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| |
Collapse
|
33
|
Zalghout S, Martinod K. Therapeutic potential of DNases in immunothrombosis: promising succor or uncertain future? J Thromb Haemost 2025; 23:760-778. [PMID: 39667687 DOI: 10.1016/j.jtha.2024.11.028] [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: 09/24/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024]
Abstract
Sepsis, a life-threatening condition characterized by systemic inflammation and multiorgan dysfunction, is closely associated with the excessive formation of neutrophil extracellular traps (NETs) and the release of cell-free DNA. Both play a central role in sepsis progression, acting as major contributors to immunothrombosis and associated complications. Endogenous DNases play a pivotal role in degrading NETs and cell-free DNA, yet their activity is often dysregulated during thrombotic disease. Although exogenous DNase1 administration has shown potential in reducing NET burden and mitigating the detrimental effects of immunothrombosis, its therapeutic efficacy upon intravenous administration remains uncertain. The development of engineered DNase formulations and combination therapies may further enhance its therapeutic effectiveness by modifying its pharmacodynamic properties and avoiding the adverse effects associated with NET degradation, respectively. Although NETs are well-established targets of DNase1, it remains uncertain whether the positive effects of DNase1 on immunothrombosis are exclusively related to it's targeting of NETs or if other components contributing to immunothrombosis are also affected. This review examines the endogenous regulation of NETs in circulation and the therapeutic potential of DNases in immunothrombosis, underscoring the necessity for further investigation to optimize their clinical application.
Collapse
Affiliation(s)
- Sara Zalghout
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| |
Collapse
|
34
|
Liu J, Li N, Wang B, Zhao W, Zhi J, Jia X, Jia Y, Tie Y. SARS-CoV-2 Infection Aggravates Physical and Mental Health in Cancer Patients Compared to Co-Living Individuals. Cancer Med 2025; 14:e70795. [PMID: 40129313 PMCID: PMC11933713 DOI: 10.1002/cam4.70795] [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: 10/09/2024] [Revised: 02/19/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
INTRODUCTION Cancer patients are particularly vulnerable to the psychological sequels of COVID-19 due to their immunocompromised state and the disruptions to their regular oncological care. There is limited research comparing the effects of SARS-CoV-2 on cancer patients and their co-living individuals. This study aims to explore the similarities and differences in physical and psychological outcomes between these two groups, with a 1-year follow-up to assess long-term effects. METHODS This retrospective observational study was conducted between January and February 2023. A total of 107 participants were included: 72 cancer patients and 35 co-living individuals, all diagnosed with COVID-19. Clinical and laboratory data were collected. Depression, anxiety, and fatigue were assessed at two timepoints: shortly after COVID-19 diagnosis and 1 year later. RESULTS Cancer patients exhibited higher rates of gastrointestinal symptoms, such as diarrhea (20.83% vs. 5.71%, p = 0.045), which were associated with increased anxiety and depression (p < 0.05). Advanced-stage cancer (p < 0.01) and lack of vaccination (p < 0.01) correlated with worse psychological outcomes. Female cancer patients reported higher depression scores (p < 0.05). Laboratory findings indicated higher neutrophil percentages (p < 0.001), fibrinogen (p < 0.001), and D-dimer levels (p = 0.015) in cancer patients, signaling a higher risk of inflammation and thrombosis. Both groups showed improvements in depression and fatigue over the 1-year follow-up, but cancer patients continued to report greater psychological distress (p < 0.001) and fatigue (p = 0.024). CONCLUSION Cancer patients infected with COVID-19 experienced more severe physical and psychological symptoms compared to their co-living individuals, with persistent differences 1 year after infection. TRIAL REGISTRATION ChiCTR2300067577.
Collapse
Affiliation(s)
- Jiayao Liu
- The Third Department of OncologyHebei General HospitalShijiazhuangChina
- Hebei Medical UniversityShijiazhuangChina
| | - Na Li
- The Third Department of OncologyHebei General HospitalShijiazhuangChina
| | - Bin Wang
- The Third Department of OncologyHebei General HospitalShijiazhuangChina
| | - Wujie Zhao
- The Third Department of OncologyHebei General HospitalShijiazhuangChina
| | - Jie Zhi
- The Third Department of OncologyHebei General HospitalShijiazhuangChina
| | - Xiaojing Jia
- The Third Department of OncologyHebei General HospitalShijiazhuangChina
| | - Yitao Jia
- The Third Department of OncologyHebei General HospitalShijiazhuangChina
| | - Yanqing Tie
- Department of Clinical LaboratoryHebei General HospitalShijiazhuangChina
| |
Collapse
|
35
|
Hwang S, Kang SW, Choi J, Park KA, Lim DH, Shin JY, Kang D, Cho J, Kim SJ. Ocular Adverse Events Following Coronavirus Disease 2019 Infection: A Self-controlled Case Series Study from the Entire Korean Population. OPHTHALMOLOGY SCIENCE 2025; 5:100638. [PMID: 39639889 PMCID: PMC11616028 DOI: 10.1016/j.xops.2024.100638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/26/2024] [Accepted: 10/21/2024] [Indexed: 12/07/2024]
Abstract
Purpose This study aimed to assess the risk of ocular adverse events, including retinal artery occlusion (RAO), retinal vein occlusion (RVO), noninfectious uveitis (NIU), noninfectious scleritis (NIS), optic neuritis (ON), ischemic optic neuropathy (ION), and ocular motor cranial nerve palsy (OMCNP), after coronavirus disease 2019 (COVID-19) infection. Design Population-based self-controlled case series (SCCS). Participants The study included patients from the entire Korean population of 52 million who experienced incident RAO, RVO, anterior NIU, nonanterior NIU, NIS, ON, ION, or OMCNP between January 1, 2021, and October 29, 2022. Methods This nationwide SCCS utilized data from the Korea National Health Insurance Service and the Korea Disease Control and Prevention Agency. The risk period after infection was defined as up to 24 weeks after COVID-19 infection. Conditional Poisson regression was used to calculate the relative incidence rate ratios (IRRs) for RAO, RVO, anterior NIU, nonanterior NIU, NIS, ON, ION, and OMCNP during the designated risk periods. Main Outcome Measures The IRRs for RAO, RVO, anterior NIU, nonanterior NIU, NIS, ON, ION, and OMCNP during the risk periods. Results The study included 9336, 103 362, 201 010, 25 428, 23 744, 3026, 69 933, and 16 335 cases of incident RAO, RVO, anterior NIU, nonanterior NIU, NIS, ON, ION, and OMCNP, respectively. The IRRs (95% confidence interval) during the early risk period (1-8 weeks) were 0.94 (0.83-1.07), 1.01 (0.97-1.04), 1.00 (0.98-1.03), 0.96 (0.90-1.03), 1.00 (0.94-1.07), 0.97 (0.81-1.17), 0.97 (0.93-1.01), and 1.02 (0.94-1.11), respectively. In the late risk period (9-24 weeks), the IRRs were 1.02 (0.92-1.12), 1.01 (0.98-1.04), 1.01 (0.99-1.03), 1.02 (0.97-1.08), 1.02 (0.97-1.08), 0.99 (0.85-1.15), 1.02 (0.99-1.06), and 0.97 (0.90-1.03), respectively. Stratified analyses showed that in patients with a history of cerebro-cardiovascular disease, the risk of RAO increased during the late risk period, with an IRR (95% confidence interval) of 1.19 (1.02-1.40). Conclusions The risk of incident RVO, anterior NIU, nonanterior NIU, NIS, ON, ION, or OMCNP did not increase after COVID-19 infection. The risk of incident RAO increased only in individuals with preexisting cardio-cerebrovascular disease. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Sungsoon Hwang
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se Woong Kang
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jaehwan Choi
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- Department of Ophthalmology, Kyung Hee University Medical Center, Kyung Hee University, Seoul, Republic of Korea
| | - Kyung-Ah Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Hui Lim
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Ju-Young Shin
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea
| | - Danbee Kang
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Juhee Cho
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang Jin Kim
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
36
|
Costa ADSD, Vadym K, Park K. Engineered endothelium model enables recapitulation of vascular function and early atherosclerosis development. Biomaterials 2025; 314:122889. [PMID: 39423515 DOI: 10.1016/j.biomaterials.2024.122889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Human health relies heavily on the vascular endothelium. Here, we propose a novel engineered endothelium model (EEM), which recapitulated both normal vascular function and pathology. An artificial basement membrane (aBM), where porous polyvinyl alcohol hydrogel was securely integrated with human fibroblast-derived, decellularized extracellular matrix on both sides was fabricated first and followed by endothelial cells (ECs) and pericytes (PCs) adhesion, respectively. Our EEM formed robust adherens junction (VE-cad) and built an impermeable barrier with time, along with the nitric oxide (NO) secretion. In our EEM, ECs and PCs interacted each other via aBM and led to hemoglobin alpha 1 (Hb-α1) development, which was involved in NO control and was strongly interconnected with VE-cad as well. A resilient property of EEM under inflammatory milieu was also confirmed by VE-cad and barrier recovery with time. In particular interest, foam cells formation, a hallmark of atherosclerotic initiation was successfully recapitulated in our EEM, where a series of sequential events were confirmed: human monocytes adhesion, transendothelial migration, and oxidized low-density lipoprotein uptake by macrophages. Collectively, our EEM is excellent in recapitulating not only normal endothelium but early pathologic one, thereby enabling EEM to be a physiologically relevant model for vascular study and disease modeling.
Collapse
Affiliation(s)
| | - Kopych Vadym
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
37
|
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
|
38
|
Ragni A, Biamonte E, Cavigiolo B, Mollero ELM, Bendotti G, Gabellieri E, Leporati P, Gallo M. COVID19 infection and vaccination and the risk of pituitary apoplexy: an entangled yarn. Endocrine 2025; 87:459-467. [PMID: 39433700 DOI: 10.1007/s12020-024-04078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024]
Abstract
PURPOSE Pituitary apoplexy (PA) has been increasingly reported in association with both infection from and vaccination for COVID19. Our aim was to analyse the available published cases and compare the clinical characteristics in the two groups (infection vs vaccination). METHODS We systematically reviewed the published literature for all cases of PA associated with COVID19 infection or vaccination. We also presented two cases managed at our Centre. RESULTS Collectively, fortythree cases were analysed. Patients with PA after COVID19 vaccination (n = 7), compared with patients with PA after COVID19 infection (n = 36), were significantly younger (p = 0.009) and had a more abrupt onset of PA (p = 0.022), but showed a milder hormonal involvement (p = 0.008) and a lower rate of persistent hypopituitarism during follow-up (p = 0.001). Patients in the vaccination group did not have clinical risk factors for PA, although this difference did not reach statistical significance. CONCLUSIONS PA associated with COVID19 is a rare but clinically significant entity, although pathophysiological details of this association are lacking. Given the significantly different clinical presentation, we could speculate that PA induced by COVID19 vaccination might represent a distinct clinical entity, with different pathophysiological mechanism, compared to PA from COVID19 infection.
Collapse
Affiliation(s)
- Alberto Ragni
- Endocrinology and Metabolic Diseases Unit, SS. Antonio e Biagio e Cesare Arrigo Teaching Hospital, Alessandria, Italy.
| | - Emilia Biamonte
- Endocrinology and Metabolic Diseases Unit, SS. Antonio e Biagio e Cesare Arrigo Teaching Hospital, Alessandria, Italy
| | - Beatrice Cavigiolo
- Endocrinology Unit, Maggiore della Carità Teaching Hospital, University of Eastern Piedmont, Novara, Italy
| | | | - Giulia Bendotti
- Endocrinology and Metabolic Diseases Unit, SS. Antonio e Biagio e Cesare Arrigo Teaching Hospital, Alessandria, Italy
| | - Enrico Gabellieri
- Endocrinology and Metabolic Diseases Unit, SS. Antonio e Biagio e Cesare Arrigo Teaching Hospital, Alessandria, Italy
| | - Paola Leporati
- Endocrinology and Metabolic Diseases Unit, SS. Antonio e Biagio e Cesare Arrigo Teaching Hospital, Alessandria, Italy
| | - Marco Gallo
- Endocrinology and Metabolic Diseases Unit, SS. Antonio e Biagio e Cesare Arrigo Teaching Hospital, Alessandria, Italy
| |
Collapse
|
39
|
Anzalone N, Gerevini S, del Poggio A, Gaudino S, Causin F, Politi LS, Triulzi FM, Pero G, Pichiecchio A, Bastianello S, Baruzzi FM, Bianchini E, Foti G, Ricciardi GK, Sponza M, Menozzi R, Cosottini M, Chirico PD, Saba L, Gasparotti R. Neuroradiological manifestations in hospitalized patients with COVID-19: An Italian national multicenter study on behalf of AINR (Associazione Italiana di Neuroradiologia) and SIRM (Società Italiana di Radiologia Medica). Neuroradiol J 2025; 38:44-51. [PMID: 38897216 PMCID: PMC11562890 DOI: 10.1177/19714009241240312] [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: 06/21/2024] Open
Abstract
PURPOSE This multicentric study aims to characterize and assess the occurrence of neuroradiological findings among patients with SARS-CoV-2 infection during the first Italian wave of the pandemic outbreak. MATERIALS AND METHODS Patients' data were collected between May 2020 and June 2020. Clinical and laboratory data, chest imaging, brain CT, and MRI imaging were included. Acquired data were centralized and analyzed in two hospitals: ASST Spedali Civili, Brescia, and IRRCS San Raffaele Research Hospital, Milan, Italy. COVID-19 patients were classified into two different subgroups, vascular and nonvascular. The vascular pattern was further divided into ischemic and hemorrhagic stroke groups. RESULTS Four hundred and fifteen patients from 20 different Italian Centers were enrolled in the study. The most frequent symptom was focal neurological deficit, found in 143 patients (34.5%). The most frequent neuroradiological finding was ischemic stroke in 122 (29.4%) patients. Forty-four (10.6%) patients presented a cerebral hemorrhage. Forty-seven patients had non-stroke neuroimaging lesions (11.3%). The most common was PRES-like syndrome (28%), SWI hypointensities (22%), and encephalitis (19%). The stroke group had higher CAD risk (37.5% vs 20%, p = .016) and higher D-dimer levels (1875 ng/mL vs 451 ng/mL, p < .001) compared to the negative group. CONCLUSION Our study describes the biggest cohort study in Italy on brain imaging of COVID-19 patients and confirms that COVID-19 patients are at risk of strokes, possibly due to a pro-thrombotic microenvironment. Moreover, apart from stroke, the other neuroradiological patterns described align with the ones reported worldwide.
Collapse
Affiliation(s)
- Nicoletta Anzalone
- Neuroradiology Department, IRCCS San Raffaele Scientific Institute, Italy
- Vita-Salute San Raffaele University, Italy
| | | | - Anna del Poggio
- Neuroradiology Department, IRCCS San Raffaele Scientific Institute, Italy
| | - Simona Gaudino
- Radiology Department, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Italy
| | | | | | - Fabio Maria Triulzi
- Neuroradiology Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Italy
| | - Guglielmo Pero
- Neuroradiology Department, ASST Grande Ospedale Metropolitano Niguarda, Italy
| | | | | | | | - Elena Bianchini
- Neuroradiology Unit, Radiology Department, Ospedale Legnano, Italy
| | - Giovanni Foti
- Radiology Department, Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | | | - Massimo Sponza
- Neuroradiology Department, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Roberto Menozzi
- Neuroradiology Department, Azienda Ospedaliera Universitaria, Parma, Italy
| | - Mirco Cosottini
- Neuroradiology Department, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | | | - Luca Saba
- Radiology Department, Azienda Ospedaliero Universitaria, Cagliari, Italy
| | | |
Collapse
|
40
|
Lei B, Su Y, Chen R, Chen Z, Liu B, Chen Y, Zhou M, Wang X, Ma Q. Uncovering the Mechanisms of BaBaoWuDanYaoMo Against Influenza A Virus and Virus-Induced Inflammation Based on Network Pharmacology and Pharmacological Evaluation. Infect Drug Resist 2025; 18:567-587. [PMID: 39902273 PMCID: PMC11789520 DOI: 10.2147/idr.s491101] [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: 08/12/2024] [Accepted: 01/11/2025] [Indexed: 02/05/2025] Open
Abstract
Purpose The pro-inflammatory response triggered by influenza viruses can contribute to viral pneumonia, even death. The effect and mechanism of BaBaoWuDanYaoMo (BB) against influenza virus remains obscure. To predict its therapeutic targets via network pharmacology and verify the therapeutic effect and the mechanism of BB against influenza virus in vitro and in vivo. Material and Methods The potential active and underlying mechanism of BB in the treatment of influenza virus was predicted through network pharmacological strategies and Molecular Docking. The protective and anti-inflammatory effects of BB were determined by cytopathic effect (CPE), quantitative real-time PCR, mitochondrial membrane potentials and Western blotting assay in vitro. BALB/c mice were intranasally infected with A/PR/8/34 (H1N1) (PR8) and orally administrated BB (500 mg/kg, 250 mg/kg and 125 mg/kg) or oseltamivir daily. The normal group was orally administrated PBS for 5 days. Lung indexes, histological changes and cytokines were determined on the 6th day. Results BB could effectively inhibit A/Puerto Rico/8/1934 (H1N1), A/GZ/GIRD07/09 (H1N1), A/HK/Y280/97 (H9N2) and A/Aichi/68 (H3N2) with IC50 values of 116.5 ± 2.2, 59.86 ± 8.33, 102.87 ± 6.66 and 66.43 ± 6.785 μg/mL, respectively. It could inhibit PR8-induced apoptosis and inhibit the expression of apoptosis markers (cleaved PARP). BB inhibited the mRNA expression of MCP-1/CCL-2, IL-6, CXCL-8/IL-8, TNF-α and CXCL-10/IP-10, and downregulated the protein expression of phosphorylated AKT/p38 and TLR3 in vitro. BB (500 and 250 mg/kg) could improve pulmonary histopathological changes, decrease the lung index and suppress the mRNA expression of CXCL1/KC, TNF-α, CXCL9/MIG and IL-1β in vivo. Conclusion BB has a protective effect on PR8-induced acute lung injury (ALI) probably via inhibition of apoptosis process and interfering with TLR3, p38 MAPK and PI3K-AKT signaling pathways. This study provided a potential treatment for influenza from BB, and network pharmacology provided a strategy to explore active components and mechanism of TCMs against influenza virus infection.
Collapse
Affiliation(s)
- Biao Lei
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yongjie Su
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Ruihan Chen
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zexing Chen
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Bin Liu
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yuou Chen
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
- King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Meihua Zhou
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xinhua Wang
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
- Institute of Integration of Traditional and Western Medicine, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Qinhai Ma
- 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 Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
41
|
Bakay OSK, Cetin N, Bakay U, Cinar G, Goksin S. A Window into the Vascular Endothelium in Covid-19: Nails. Dermatol Pract Concept 2025; 15:dpc.1501a4927. [PMID: 39853249 PMCID: PMC11928108 DOI: 10.5826/dpc.1501a4927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2024] [Indexed: 01/26/2025] Open
Abstract
INTRODUCTION Endothelial damage is associated with acute and long-term coronavirus disease 2019 (COVID-19) complications. Proximal nailfold capillaries and nail beds give important clues to microvascular changes associated with endothelial dysfunction. OBJECTIVE We aimed to use dermoscopy to examine the proximal nailfold capillaries and nail bed of COVID-19 patients and identify microvascular changes. METHODS A prospective study was designed to evaluate the dermoscopic features of proximal nail fold capillaries and nail bed in mild-to-moderate COVID-19 patients and healthy controls between June 2022 and December 2023. The patients underwent their initial dermoscopic examination two weeks after the onset of symptoms, followed by a follow-up evaluation 10-14 months later. RESULTS The study included 46 patients with mild-to-moderate COVID-19 and 62 healthy controls. The presence of avascular areas (P <0.001), meandering capillaries (P = 0.016), microhemorrhages (P = 0.007), and enlarged capillaries (P = 0.009) in the proximal nail fold was significantly higher in COVID-19 patients than in healthy controls. The capillary architecture was disorganized (P = 0.002) and density reduced (P <0.001) in COVID-19 patients compared to healthy controls. In the follow-up examination, microvascular changes were observed to have regressed. CONCLUSIONS Proximal nailfold dermoscopy is an effective, low-cost, easily accessible method that enables observation of microvascular changes in COVID-19 patients.
Collapse
Affiliation(s)
| | - Niyazi Cetin
- Pamukkale University Faculty of Medicine, Department of Dermatology, Denizli, Turkey
| | - Umut Bakay
- Denizli State Hospital, Department of Rheumatology, Denizli, Turkey
| | - Gokhan Cinar
- Agrı Training And Research Hospital, Department of Dermatology, Agrı, Turkey
| | - Sule Goksin
- Pamukkale University Faculty of Medicine, Department of Dermatology, Denizli, Turkey
| |
Collapse
|
42
|
Spinelli R, Sanchis I, Rietmann Á, Húmpola MV, Siano Á. Amphibian-Derived Peptides as Natural Inhibitors of SARS-CoV-2 Main Protease (M pro): A Combined In Vitro and In Silico Approach. Chem Biodivers 2025:e202403202. [PMID: 39854653 DOI: 10.1002/cbdv.202403202] [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: 12/03/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/26/2025]
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has highlighted the urgent need for novel therapeutic agents targeting viral enzymes such as the main protease (Mpro), which plays a crucial role in viral replication. In this study, we investigate the inhibitory potential of 23 peptides isolated from the skin of amphibians belonging to the Hylidae and Leptodactylidae families against SARS-CoV-2 Mpro. Five peptides demonstrated significant inhibition using a colorimetric Mpro inhibition assay, with IC50 values ranging from 41 to 203 µM. Among these, peptides Hp-1081 and Hp-1971, derived from Boana pulchella, exhibited the strongest activity, comparable to the natural Mpro inhibitor quercetin. The binding mechanism of the most potent peptide, Hp-1081, was further investigated through docking and molecular dynamics (MDs) simulations and energetic analysis, which revealed key Mpro residues involved in the binding process. Moreover, because SARS-CoV-2 infection can induce ROS overproduction, the antioxidant activity of Hp-1081 was assessed, reaching 48% of DPPH radical scavenging activity at 100 µM. The most potent peptides also showed no toxicity against human erythrocytes and Artemia salina. This study provides insight into the antiviral potential of amphibian-derived peptides and highlights their applicability as natural templates for drug development targeting coronaviruses.
Collapse
Affiliation(s)
- Roque Spinelli
- Laboratorio de Péptidos Bioactivos, Departamento de Química Orgánica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Iván Sanchis
- Laboratorio de Péptidos Bioactivos, Departamento de Química Orgánica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Álvaro Rietmann
- Laboratorio de Péptidos Bioactivos, Departamento de Química Orgánica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Verónica Húmpola
- Laboratorio de Péptidos Bioactivos, Departamento de Química Orgánica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Álvaro Siano
- Laboratorio de Péptidos Bioactivos, Departamento de Química Orgánica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
43
|
Zaaqoq AM, Mazzeffi MA, Vogelsong MA, Roeser M, Cho SM. Lung injury in myocardial infarction-associated cardiogenic shock supported by venoarterial extracorporeal membrane oxygenation: a scoping review. BMC Cardiovasc Disord 2025; 25:40. [PMID: 39849351 PMCID: PMC11756158 DOI: 10.1186/s12872-025-04472-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Acute lung injury and acute respiratory failure are frequent complications of cardiogenic shock and are associated with increased morbidity and mortality. Even with increased use of temporary mechanical circulatory support, such as venoarterial extracorporeal membrane oxygenation (VA-ECMO), acute lung injury related to cardiogenic shock continues to have a determinantal effect on patient outcomes. OBJECTIVES To summarize potential mechanisms of acute lung injury described in patients with cardiogenic shock supported by VA-ECMO and determine current knowledge gaps. METHODS We searched literature from January 1st, 2010, to December 31st, 2023, using MEDLINE, EMBASE, and Web of Science databases on February 27th, 2024. The search strategy was split into two main domains: (a) cardiogenic shock and ECMO and (b) Acute respiratory failure and ECMO. RESULTS The search yielded 2246 citations. After 743 duplicates were removed, 1465 citations remained. Of these studies, 1456 were excluded based on the exclusion criteria, leaving the final eight studies we included in our scoping review. We identified disruption of the pulmonary blood flow in patients with cardiogenic shock, with cardiac arrest being an extreme form of cardiogenic shock. Placing the patient on VA-ECMO could intensify this process of lung injury. CONCLUSION Acute lung injury in patients with cardiogenic shock, especially when supported by VA ECMO, is a significant complication that is associated with increased morbidity and mortality. There is a limited understanding of the underlying mechanisms that could represent opportunities for future research to mitigate its development and provide the best approach to protecting and monitoring lung function.
Collapse
Affiliation(s)
- Akram M Zaaqoq
- Department of Anesthesiology, Division of Critical Care, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA.
| | - Michael A Mazzeffi
- Department of Anesthesiology, Division of Critical Care, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA
| | - Melissa A Vogelsong
- Department of Anesthesiology, Division of Cardiac Anesthesia, Stanford University, Stanford, CA, USA
| | - Mark Roeser
- Department of Surgery, Division of Cardiac Surgery, University of Virginia, Charlottesville, VA, USA
| | - Sung-Min Cho
- Department of Surgery, Division of Cardiac Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Departments of Neurology, Anesthesiology, Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
44
|
Cambon A, Guervilly C, Delteil C, Potere N, Bachelier R, Tellier E, Abdili E, Leprince M, Giani M, Polidoro I, Albanese V, Ferrante P, Coffin L, Schiffrin M, Arnaud L, Lacroix R, Roque S, Forel JM, Hraiech S, Daniel L, Papazian L, Dignat-George F, Kaplanski G. Caspase-1 activation, IL-1/IL-6 signature and IFNγ-induced chemokines in lungs of COVID-19 patients. Front Immunol 2025; 15:1493306. [PMID: 39882243 PMCID: PMC11774885 DOI: 10.3389/fimmu.2024.1493306] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025] Open
Abstract
Rationale COVID-19-associated acute-respiratory distress syndrome (C-ARDS) results from a direct viral injury associated with host excessive innate immune response mainly affecting the lungs. However, cytokine profile in the lung compartment of C-ARDS patients has not been widely studied, nor compared to non-COVID related ARDS (NC-ARDS). Objectives To evaluate caspase-1 activation, IL-1 signature, and other inflammatory cytokine pathways associated with tissue damage using post-mortem lung tissues, bronchoalveolar lavage fluids (BALF), and serum across the spectrum of COVID-19 severity. Methods Histological features were described and activated-caspase-1 labeling was performed in 40 post-mortem biopsies. Inflammatory cytokines were quantified in BALF and serum from 19 steroid-treated-C-ARDSand compared to 19 NC-ARDS. Cytokine concentrations were also measured in serum from 128 COVID-19 patients at different severity stages. Measurements and main results Typical "diffuse alveolar damage" in lung biopsies were associated with activated caspase-1 expression and vascular lesions. Soluble Caspase-1p20, IL-1β, IL-1Ra, IL-6 and at lower level IFNγ and CXCL-10, were highly elevated in BALF from steroid-treated-C-ARDS as well as in NC-ARDS. IL-1β appeared concentrated in BALF, whereas circulating IL-6 and IL-1Ra concentrations were comparable to those in BALF and correlated with severity. TNFα, TNFR1 and CXCL8 however, were significantly higher in NC-ARDS compared to C-ARDS, treated by steroid. Conclusions In the lungs of C-ARDS, both caspase-1 activation with a predominant IL-1β/IL-6 signature and IFNγ -associated chemokines are elevated despite steroid treatment. These pathways may be specifically targeted in ARDS to improve response to treatment and to limit alveolar and vascular lung damage.
Collapse
Affiliation(s)
- Audrey Cambon
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Christophe Guervilly
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Clémence Delteil
- Département de Médecine légale, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille University, Marseille, France
| | - Nicola Potere
- School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | | | - Edwige Tellier
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Evelyne Abdili
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Marine Leprince
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| | - Marco Giani
- School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Ildo Polidoro
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | - Valentina Albanese
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | - Paolo Ferrante
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | | | | | - Laurent Arnaud
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Romaric Lacroix
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Sandrine Roque
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| | - Jean-Marie Forel
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Sami Hraiech
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Laurent Daniel
- Service d’Anatomopathologie, APHM, Aix Marseille University, Marseille, France
| | - Laurent Papazian
- Service de Réanimation, Centre Hospitalier de Bastia, Bastia, France
| | - Françoise Dignat-George
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Gilles Kaplanski
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| |
Collapse
|
45
|
Murad D, Paracha RZ, Nisar M. Unravelling the impact of SARS-CoV-2 on hemostatic and complement systems: a systems immunology perspective. Front Immunol 2025; 15:1457324. [PMID: 39885991 PMCID: PMC11781117 DOI: 10.3389/fimmu.2024.1457324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025] Open
Abstract
The hemostatic system prevents and stops bleeding, maintaining circulatory integrity after injury. It directly interacts with the complement system, which is key to innate immunity. In coronavirus disease 2019 (COVID-19), dysregulation of the hemostatic and complement systems has been associated with several complications. To understand the essential balance between activation and regulation of these systems, a quantitative systems immunology model can be established. The dynamics of the components are examined under three distinct conditions: the disease state representing symptomatic COVID-19 state, an intervened disease state marked by reduced levels of regulators, and drug interventions including heparin, tranexamic acid, avdoralimab, garadacimab, and tocilizumab. Simulation results highlight key components affected, including thrombin, tissue plasminogen activator, plasmin, fibrin degradation products, interleukin 6 (IL-6), the IL-6 and IL-6R complex, and the terminal complement complex (C5b-9). We explored that the decreased levels of complement factor H and C1-inhibitor significantly elevate these components, whereas tissue factor pathway inhibitor and alpha-2-macroglobulin have more modest effects. Furthermore, our analysis reveals that drug interventions have a restorative impact on these factors. Notably, targeting thrombin and plasmin in the early stages of thrombosis and fibrinolysis can improve the overall system. Additionally, the regulation of C5b-9 could aid in lysing the virus and/or infected cells. In conclusion, this study explains the regulatory mechanisms of the hemostatic and complement systems and illustrates how the biopathway machinery sustains the balance between activation and inhibition. The knowledge that we have acquired could contribute to designing therapies that target the hemostatic and complement systems.
Collapse
Affiliation(s)
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering and Sciences (SINES), Department of Sciences,
National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | | |
Collapse
|
46
|
Yan X, Zhang X, Song W, Qi T, Wang Z, Tang Y, Sun J, Xu S, Yang J, Wang J, Chen J, Zhang R, Liu L, Shen Y. Metabolomic Profiling Reveals Potential Biomarkers and Prominent Features in HIV/AIDS Patients Co-Infected with SARS-CoV-2. Microorganisms 2025; 13:144. [PMID: 39858912 PMCID: PMC11767690 DOI: 10.3390/microorganisms13010144] [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: 11/30/2024] [Revised: 12/19/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The underlying mechanisms and diagnostic biomarkers for the progress of COVID-19 in HIV patients have not been fully elucidated. In this study, the aim is to analyze the metabolomic profiles of HIV/AIDS patients co-infected with SARS-CoV-2 and to identify biomarkers indicative of co-infection. In this study, we conducted a retrospective cohort analysis of peripheral blood samples collected from 30 HIV/AIDS patients co-infected with SARS-CoV-2 (pc group) and 30 patients without SARS-CoV-2 (nc group). In this study, through non-targeted metabolomics and lipidomics analysis, 77 differential metabolites were identified in the plasma of patients co-infected with HIV and SARS-CoV-2 compared to the nc group, with vitamin K1 emerging as a significant feature. Moreover, the plasma of the pc group showed disturbances in lipid metabolism, with elevated triglycerides (TG) and phosphatidylcholine (PC) and decreased phosphatidylglycerol (PG) compared to the control group. Vitamin K1 may be a biomarker for SARS-CoV-2 in HIV/AIDS patients, and changes in the levels of TG, PC, and PG molecules appear to be the main features following HIV co-infection with COVID-19. The emphasis in our study is on the power of using comprehensive metabolomics (lipidomics) approaches to identify metabolic biomarkers and potential mechanisms of COVID-19 in HIV/AIDS patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Li Liu
- Department of Infection and Immunity, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (X.Y.); (X.Z.); (W.S.); (T.Q.); (Z.W.); (Y.T.); (J.S.); (S.X.); (J.Y.); (J.W.); (J.C.); (R.Z.)
| | - Yinzhong Shen
- Department of Infection and Immunity, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (X.Y.); (X.Z.); (W.S.); (T.Q.); (Z.W.); (Y.T.); (J.S.); (S.X.); (J.Y.); (J.W.); (J.C.); (R.Z.)
| |
Collapse
|
47
|
Honchar O, Ashcheulova T, Chumachenko T, Chumachenko D. Early prediction of long COVID-19 syndrome persistence at 12 months after hospitalisation: a prospective observational study from Ukraine. BMJ Open 2025; 15:e084311. [PMID: 39762090 PMCID: PMC11748775 DOI: 10.1136/bmjopen-2024-084311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVE To identify the early predictors of a self-reported persistence of long COVID syndrome (LCS) at 12 months after hospitalisation and to propose the prognostic model of its development. DESIGN A combined cross-sectional and prospective observational study. SETTING A tertiary care hospital. PARTICIPANTS 221 patients hospitalised for COVID-19 who have undergone comprehensive clinical, sonographic and survey-based evaluation predischarge and at 1 month with subsequent 12-month follow-up. The final cohort included 166 patients who had completed the final visit at 12 months. MAIN OUTCOME MEASURE A self-reported persistence of LCS at 12 months after discharge. RESULTS Self-reported LCS was detected in 76% of participants at 3 months and in 43% at 12 months after discharge. Patients who reported incomplete recovery at 1 year were characterised by a higher burden of comorbidities (Charlson index of 0.69±0.96 vs 0.31±0.51, p=0.001) and residual pulmonary consolidations (1.56±1.78 vs 0.98±1.56, p=0.034), worse blood pressure (BP) control (systolic BP of 138.1±16.2 vs 132.2±15.8 mm Hg, p=0.041), renal (estimated glomerular filtration rate of 59.5±14.7 vs 69.8±20.7 mL/min/1.73 m2, p=0.007) and endothelial function (flow-mediated dilation of the brachial artery of 10.4±5.4 vs 12.4±5.6%, p=0.048), higher in-hospital levels of liver enzymes (alanine aminotransferase (ALT) of 76.3±60.8 vs 46.3±25.3 IU/L, p=0.002) and erythrocyte sedimentation rate (ESR) (34.3±12.1 vs 28.3±12.6 mm/h, p=0.008), slightly higher indices of ventricular longitudinal function (left ventricular (LV) global longitudinal strain (GLS) of 18.0±2.4 vs 17.0±2.3%, p=0011) and higher levels of Hospital Anxiety and Depression Scale anxiety (7.3±4.2 vs 5.6±3.8, p=0.011) and depression scores (6.4±3.9 vs 4.9±4.3, p=0.022) and EFTER-COVID study physical symptoms score (12.3±3.8 vs 9.2±4.2, p<0.001). At 1 month postdischarge, the persisting differences included marginally higher LV GLS, mitral E/e' ratio and significantly higher levels of both resting and exertional physical symptoms versus patients who reported complete recovery. Logistic regression and machine learning-based binary classification models have been developed to predict the persistence of LCS symptoms at 12 months after discharge. CONCLUSIONS Compared with post-COVID-19 patients who have completely recovered by 12 months after hospital discharge, those who have subsequently developed 'very long' COVID were characterised by a variety of more pronounced residual predischarge abnormalities that had mostly subsided by 1 month, except for steady differences in the physical symptoms levels. A simple artificial neural networks-based binary classification model using peak ESR, creatinine, ALT and weight loss during the acute phase, predischarge 6-minute walk distance and complex survey-based symptoms assessment as inputs has shown a 92% accuracy with an area under receiver-operator characteristic curve 0.931 in prediction of LCS symptoms persistence at 12 months.
Collapse
Affiliation(s)
- Oleksii Honchar
- Department of Propedeutics of Internal Medicine, Nursing and Bioethics, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Tetiana Ashcheulova
- Department of Propedeutics of Internal Medicine, Nursing and Bioethics, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Tetyana Chumachenko
- Department of Epidemiology, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Dmytro Chumachenko
- Department of Mathematical Modelling and Artificial Intelligence, National Aerospace University Kharkiv Aviation Institute, Kharkiv, Ukraine
| |
Collapse
|
48
|
Luo X, Zhang N, Liu Y, Du B, Wang X, Zhao T, Liu B, Zhao S, Qiu J, Wang G. Resolving the developmental mechanisms of cardiac microthrombosis of SARS-CoV-2 based on single-cell transcriptome analysis. SCIENCE CHINA. LIFE SCIENCES 2025; 68:103-115. [PMID: 39470924 DOI: 10.1007/s11427-023-2624-9] [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: 03/21/2024] [Accepted: 05/21/2024] [Indexed: 11/01/2024]
Abstract
The coronavirus disease 2019 (COVID-19) outbreak caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) developed into a global health emergency. Systemic microthrombus caused by SARS-CoV-2 infection is a common complication in patients with COVID-19. Cardiac microthrombosis as a complication of SARS-CoV-2 infection is the primary cause of cardiac injury and death in patietns with severe COVID-19. In this study, we performed single-cell sequencing analysis of the right ventricular free wall tissue from healthy donors, patients who died during the hypercoagulable period of characteristic coagulation abnormality (CAC), and patients who died during the fibrinolytic period of CAC. We collected 61,187 cells enriched in 24 immune cell subsets and 13 cardiac-resident cell subsets. We found that in the course of SARS-CoV-2 infected heart microthrombus, MYO1EhighRASGEF1Bhighmonocyte-derived macrophages promoted hyperactivation of the immune system and initiated the extrinsic coagulation pathway by activating chemokines CCL3, CCL5. This series of events is the main cause of cardiac microthrombi following SARS-CoV-2 infection. In a SARS-CoV-2 infected heart microthrombus, excessive immune activation is accompanied by an increase in cellular iron content, which in turn promotes oxidative stress and intensifies intercellular competition. This induces cells to alter their metabolic environment, resulting in increased sugar uptake via the glycosaminoglycan synthesis pathway. In addition, high levels of reactive oxygen species generated by elevated iron levels promote increased endogenous malondialdehyde synthesis in a subpopulation of cardiac endothelial cells. This exacerbates endothelial cell dysfunction and exacerbates the coagulopathy process.
Collapse
Affiliation(s)
- Xizi Luo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Basic Medicine, Jilin University, Changchun, 130012, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Nan Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Basic Medicine, Jilin University, Changchun, 130012, China
- College of Mathematics, Jilin University, Changchun, 130021, China
| | - Yuntao Liu
- Bioinformatic Lab, School of Mathematics, Shandong University, Jinan, 250100, China
| | - Beibei Du
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Xuan Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Basic Medicine, Jilin University, Changchun, 130012, China
| | - Tianxu Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Basic Medicine, Jilin University, Changchun, 130012, China
| | - Bingqiang Liu
- Bioinformatic Lab, School of Mathematics, Shandong University, Jinan, 250100, China
| | - Shishun Zhao
- College of Mathematics, Jilin University, Changchun, 130021, China
| | - Jiazhang Qiu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| | - Guoqing Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Basic Medicine, Jilin University, Changchun, 130012, China.
| |
Collapse
|
49
|
Zong X, Wang X, Liu Y, Wang X, Li D, Zhou Z, Li Z. Comparative analysis of compartment-specific immunothrombotic biomarker profiles in bronchoalveolar lavage fluid and serum of patients with pneumonia-related acute respiratory distress syndrome: A preliminary cross-sectional study. J Investig Med 2025; 73:104-115. [PMID: 39324185 DOI: 10.1177/10815589241288515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Immunothrombosis has emerged as a potential mechanistic link underlying the development and progression of acute respiratory distress syndrome (ARDS), but understanding its specific profile in patients, both locally and systemically, is limited. The objective of this study was to characterize and compare the immunothrombotic signatures in patients diagnosed with pneumonia-related ARDS (p-ARDS) at both the pulmonary and systemic levels and to evaluate their clinical relevance. The study included 23 consecutive patients diagnosed with p-ARDS admitted to the intensive care unit at a tertiary university hospital from July 2022 to May 2023, alongside 40 concurrently hospitalized patients with common pneumonia as controls. Paired bronchoalveolar lavage fluid (BALF) and serum samples were collected from the participants for the analysis of 15 biomarkers to assess and quantify the pulmonary and systemic immunothrombotic signatures. The study results revealed significant pulmonary inflammation and systemic endothelial injury in p-ARDS patients compared to pneumonia controls. These observations were maintained after adjustment for severity of illness (Acute Physiology and Chronic Health Evaluation II scores). In terms of clinical relevance, inflammatory biomarkers (interleukin [IL]-6, IL-8) in BALF were found to correlate with PaO2/FiO2 ratio, while serum levels of a disintegrin and metalloproteinase with thrombospondin type 1 motif 13 (ADAMTS-13) and thrombomodulin showed associations with Sequential Organ Failure Assessment and Disseminated Intravascular Coagulation scores. In conclusion, this preliminary investigation identified compartment-specific variations in the immunothrombotic signature between patients with p-ARDS and those with pneumonia alone, with inflammatory responses predominantly localized in the alveolar compartments and coagulation/endothelial injury biomarkers more pronounced in peripheral blood.
Collapse
Affiliation(s)
- Xiaolong Zong
- Department of Clinical Laboratory, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xuechao Wang
- Department of Clinical Laboratory, Tianjin Medical University Baodi Hospital, Tianjin, China
| | - Yaru Liu
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiao Wang
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Duanyang Li
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiqing Zhou
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhenyu Li
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
50
|
Zhao Z, Zhu P, Lou Y, Hou R, Sun H, Du Y, Xu G. Receptor-Interacting Protein Kinase 3-Mediated Modulation of Endothelial Cell Necroptosis and Mitochondrial Dysfunction through AMPK/Drp1 Signaling Pathway: Insights into the Pathophysiological Mechanisms of Lipopolysaccharide-Induced Acute Lung Injury. Int J Med Sci 2025; 22:71-86. [PMID: 39744171 PMCID: PMC11659830 DOI: 10.7150/ijms.104932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/08/2024] [Indexed: 02/01/2025] Open
Abstract
Receptor-interacting protein 3 (Ripk3) plays a crucial part in acute lung injury (ALI) by regulating inflammation-induced endothelial damage in the lung tissue. The precise mechanisms through which Ripk3 contributes to the endothelial injury in ALI still remain uncertain. In the current research, we employed Ripk3-deficient (Ripk3-/-) mice to examine the role of Ripk3 in ALI progression, focusing on its effects on endothelial cells (ECs), mitochondrial damage and necroptosis. Our study observed significant Ripk3 upregulation in lipopolysaccharide- (LPS-) treated lung tissues, as well as in murine pulmonary microvascular endothelial cells (PMVECs). Ripk3 deletion improved lung tissue morphology, reduced inflammation, oxidative stress and endothelial dysfunction under LPS challenge. It also mitigated LPS-induced necroptosis and mitochondrial damage in PMVECs. Ripk3 upregulation suppressed the AMP-activated protein kinase (AMPK) pathway and activated Drp1-mediated mitochondrial fission, increasing mitochondrial permeability transition pore (mPTP) opening and PMVEC necroptosis. Conversely, Ripk3 deletion activated the AMPK/Drp1-mitochondrial fission pathway, preventing mPTP opening and PMVEC necroptosis in ALI. These findings demonstrated that Ripk3 promotes necroptosis through the AMPK/Drp1/mPTP opening pathway, identifying a potential therapeutic target for ALI treatment.
Collapse
Affiliation(s)
- Zhaoning Zhao
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Pingjun Zhu
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
- Department of Respiratory and Critical Care Medicine, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Yue Lou
- The Second Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ruoyu Hou
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
- School of Biology, University of St Andrews, St Andrews, KY16 9ST, UK
| | - Heqiang Sun
- Department of Laboratory Medicine, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingzhen Du
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
- Department of Disease Control and Prevention, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Guogang Xu
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|