1
|
Giorgetti A, Santelli S, Filipuzzi I, Bonasoni MP, Basile G, Pirani F, Pelotti S. Post-mortem diagnosis of septic arthritis by Pasteurella multocida: a case report and literature review of fatal septic arthritis. Forensic Sci Med Pathol 2025:10.1007/s12024-024-00929-x. [PMID: 39797963 DOI: 10.1007/s12024-024-00929-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2024] [Indexed: 01/13/2025]
Abstract
The diagnosis of septic arthritis remains challenging in the clinical setting, often leading to a suspicion for medical liability. Our purpose is to describe an unusual case of a post-mortem diagnosis of P. multocida fatal septic arthritis, in a healthy 67-year-old woman presenting with pain in the right shoulder. Moreover, a literature review of cases of fatal septic arthritis is provided. The multidisciplinary approach consisted of a forensic autopsy and additional post-mortem analyses (microbiology, biochemical analyses, histopathology, and revision of radiological images) carried out during the prosecutor's investigation for medical liability. A systematic review of the literature was performed to collect cases of fatal septic arthritis and to understand its frequency and characteristics. No clear cause of death was determined after the autopsy, that only highlighted swelling and purulent exudation in the right glenohumeral joint. The microbiological swab performed on the shoulder tested positive for P. multocida, while histopathological and biochemical data were consistent with a sepsis. These results guided the interview with the woman's relatives, until a history of a previous contact with a stray cat emerged. The cause of death was deemed as fatal septic arthritis caused by P. multocida, occurred after cat scratches and bites and only diagnosed post-mortem. The review of the literature provided 15 articles about fatal septic arthritis, only 1 caused by P. multocida, and all with ante-mortem diagnosis. Given the nonspecific symptoms, usually including a localized pain, and the absence of a clear history, e.g. of animal contact, septic arthritis might represent an under-reported clinical and pathological diagnosis, leading to a judicial autopsy for the suspicion for medical liability. The post-mortem examination, following a multidisciplinary approach including integration of the clinical history, microbiological and histopathological analysis, could represent the only opportunity for the diagnosis of the cause of death.
Collapse
Affiliation(s)
- Arianna Giorgetti
- Department of Medical and Surgical Sciences, Unit of Legal Medicine, University of Bologna, Via Irnerio 49, 40126, Bologna, Italy
| | - Simone Santelli
- Department of Medical and Surgical Sciences, Unit of Legal Medicine, University of Bologna, Via Irnerio 49, 40126, Bologna, Italy
| | - Ilenia Filipuzzi
- Department of Medical and Surgical Sciences, Unit of Legal Medicine, University of Bologna, Via Irnerio 49, 40126, Bologna, Italy
| | - Maria Paola Bonasoni
- Department of Medical and Surgical Sciences, Unit of Legal Medicine, University of Bologna, Via Irnerio 49, 40126, Bologna, Italy
- Pathology Unit, Azienda USL-IRCCS Di Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy
| | - Giuseppe Basile
- Trauma Unit and Emergency Department, IRCCS Galeazzi Orthopedics Institute, Milan, Italy
| | - Filippo Pirani
- Department of Medical and Surgical Sciences, Unit of Legal Medicine, University of Bologna, Via Irnerio 49, 40126, Bologna, Italy.
| | - Susi Pelotti
- Department of Medical and Surgical Sciences, Unit of Legal Medicine, University of Bologna, Via Irnerio 49, 40126, Bologna, Italy
| |
Collapse
|
2
|
Foltan M, Dinh D, Gruber M, Müller T, Hart C, Krenkel L, Schmid C, Lehle K. Incidence of neutrophil extracellular traps (NETs) in different membrane oxygenators: pilot in vitro experiments in commercially available coated membranes. J Artif Organs 2025:10.1007/s10047-024-01486-4. [PMID: 39775204 DOI: 10.1007/s10047-024-01486-4] [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: 10/03/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Neutrophil extracellular traps (NETs) were detected in blood samples and in cellular deposits of oxygenator membranes during extracorporeal membrane oxygenation (ECMO) therapy and may be responsible for thrombogenesis. The aim was to evaluate the effect of the base material of gas fiber (GF, polymethylpentene) and heat exchange (HE) membranes and different antithrombogenic coatings on isolated granulocytes from healthy volunteers under static culture conditions. Contact of granulocytes with membranes from different ECMO oxygenators (with different surface coatings) and uncoated-GFs allowed detection of adherent cells and NETotic nuclear structures (normal, swollen, ruptured) using nuclear staining. Flow cytometry was used to identify cell activation (CD11b/CD62L, oxidative burst) of non-adherent cells. Uncoated-GFs were used as a reference. Within 3 h, granulocytes adhered to the same extent on all surfaces. In contrast, the ratio of normal to NETotic cells was significantly higher for uncoated-GFs (56-83%) compared to all coated GFs (34-72%) (p < 0.001) with no difference between the coatings. After material contact, non-adherent cells remained vital with unchanged oxidative burst function and the proportion of activated cells remained low. The expression of activation markers was independent of the origin of the GF material. In conclusion, the polymethylpentene surfaces of the GFs already induce NET formation. Antithrombogenic coatings can already reduce the proportion of NETotic nuclei. However, it cannot be ruled out that NET formation can induce thrombotic events. Therefore, new surfaces or coatings are required for future ECMO systems and long-term implantable artificial lungs.
Collapse
Affiliation(s)
- M Foltan
- Department for Cardiac, Thoracic and Cardiovascular Surgery, University Hospital Regensburg, Regensburg, Germany.
| | - D Dinh
- Department for Cardiac, Thoracic and Cardiovascular Surgery, University Hospital Regensburg, Regensburg, Germany
| | - M Gruber
- Department for Anaesthesiology, University Hospital Regensburg, Regensburg, Germany
| | - T Müller
- Department for Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - C Hart
- Department for Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - L Krenkel
- Regensburg Center of Biomedical Engineering, University and OTH Regensburg, Regensburg, Germany
| | - C Schmid
- Department for Cardiac, Thoracic and Cardiovascular Surgery, University Hospital Regensburg, Regensburg, Germany
| | - K Lehle
- Department for Cardiac, Thoracic and Cardiovascular Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
3
|
Tourn J, Crescence L, Bruzzese L, Panicot-Dubois L, Dubois C. Cellular and Molecular Mechanisms Leading to Air Travel-Induced Thrombosis. Circ Res 2025; 136:115-134. [PMID: 39745986 DOI: 10.1161/circresaha.124.325208] [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] [Indexed: 01/04/2025]
Abstract
Venous thromboembolism, characterized by deep vein thrombosis and pulmonary embolism, is the third cardiovascular disease in the world. Deep vein thrombosis occurs when a blood clot forms in areas of impaired blood flow, and it is significantly affected by environmental factors. Local hypoxia, caused by venous stasis, plays a critical role in deep vein thrombosis under normal conditions, and this effect is intensified when the Po2 decreases, such as during air travel or high-altitude exposure. The lower oxygen levels and reduced pressure at high altitudes further contribute to deep vein thrombosis development. These conditions increase the pro-coagulant activity of neutrophils, platelets, and red blood cells, which interact on the surface of activated endothelial cells, promoting clot formation. Understanding the mechanisms involved in thrombus formation when Po2 is reduced, with or without pressure reduction, is crucial for preventing the development of venous thromboembolisms in such conditions and identifying innovative therapeutic targets. This literature review explores the mechanisms involved in thrombus formation related to high-altitude conditions and discusses the pro-coagulant consequences induced by environmental disturbances.
Collapse
Affiliation(s)
- Julie Tourn
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
| | - Lydie Crescence
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Laurie Bruzzese
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Laurence Panicot-Dubois
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| |
Collapse
|
4
|
Andraska EA, Denorme F, Kaltenmeier C, Arivudainabi A, Mihalko EP, Dyer M, Annarapu GK, Zarisfi M, Loughran P, Ozel M, Williamson K, Mota Alvidrez RI, Thomas K, Shiva S, Shea SM, Steinman RA, Campbell RA, Rosengart MR, Neal MD. Alterations in visible light exposure modulate platelet function and regulate thrombus formation. J Thromb Haemost 2025; 23:123-138. [PMID: 39299611 DOI: 10.1016/j.jtha.2024.08.020] [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: 05/05/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Variations in light exposure are associated with changes in inflammation and coagulation. The impact of light spectra on venous thrombosis (VT) and arterial thrombosis is largely unexplored. OBJECTIVES To investigate the impact of altering light spectrum on platelet function in thrombosis. METHODS Wild-type C57BL/6J mice were exposed to ambient (micewhite, 400 lux), blue (miceblue, 442 nm, 1400 lux), or red light (micered, 617 nm, 1400 lux) with 12:12 hour light:dark cycle for 72 hours. After 72 hours of light exposure, platelet aggregation, activation, transcriptomic, and metabolomic changes were measured. The ability of released products of platelet activation to induce thrombosis-generating neutrophil extracellular trap formation was quantified. Subsequent thrombosis was measured using murine models of VT and stroke. To translate our findings to human patients, light-filtering cataract patients were evaluated over an 8-year period for rate of venous thromboembolism with multivariable logistic regression clustered by hospital. RESULTS Exposure to long-wavelength red light resulted in reduced platelet aggregation and activation. RNA-seq analysis demonstrated no significant transcriptomic changes between micered and micewhite. However, there were global metabolomic changes in platelets from micered compared with micewhite. Releasate from activated platelets resulted in reduced neutrophil extracellular trap formation. Micered also had reduced VT weight and brain infarct size following stroke. On subgroup analysis of cataract patients, patients with a history of cancer had a lower lifetime risk of venous thromboembolism after implantation with lenses that filter low-wavelength light. CONCLUSION Light therapy may be a promising approach to thrombus prophylaxis by specifically targeting the intersection between innate immune function and coagulation.
Collapse
Affiliation(s)
- Elizabeth A Andraska
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.
| | - Frederik Denorme
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Christof Kaltenmeier
- University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA; MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital, Washington, DC, USA
| | | | - Emily P Mihalko
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mitchell Dyer
- Division of Vascular and Endovascular Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Gowtham K Annarapu
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohammadreza Zarisfi
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patricia Loughran
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Mehves Ozel
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kelly Williamson
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Kimberly Thomas
- Vitalant Research Institute, Denver, Colorado, USA; Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sruti Shiva
- Division of Classical Hematology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Susan M Shea
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Richard A Steinman
- University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA; Division of Hematology and Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert A Campbell
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Matthew R Rosengart
- Division of Acute and Critical Care Surgery, Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Matthew D Neal
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Cao Z, Jiang X, He Y, Zheng X. Metabolic landscape in venous thrombosis: insights into molecular biology and therapeutic implications. Ann Med 2024; 56:2401112. [PMID: 39297312 DOI: 10.1080/07853890.2024.2401112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 03/20/2024] [Accepted: 05/12/2024] [Indexed: 09/21/2024] Open
Abstract
The findings of the last decade suggest a complex link between inflammatory cells, coagulation, and the activation of platelets and their synergistic interaction to promote venous thrombosis. Inflammation is present throughout the process of venous thrombosis, and various metabolic pathways of erythrocytes, endothelial cells, and immune cells involved in venous thrombosis, including glucose metabolism, lipid metabolism, homocysteine metabolism, and oxidative stress, are associated with inflammation. While the metabolic microenvironment has been identified as a marker of malignancy, recent studies have revealed that for cancer thrombosis, alterations in the metabolic microenvironment appear to also be a potential risk. In this review, we discuss how the synergy between metabolism and thrombosis drives thrombotic disease. We also explore the great potential of anti-inflammatory strategies targeting venous thrombosis and the complex link between anti-inflammation and metabolism. Furthermore, we suggest how we can use our existing knowledge to reduce the risk of venous thrombosis.
Collapse
Affiliation(s)
- Zheng Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yiyu He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoxin Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
6
|
Liu X, Fan W, Lin S, Chen J, Zhang S, Li X, Jin M, He Q. Anti-Thrombotic Effect of Protoparaxotriol Saponins From Panax notoginseng Using Zebrafish Model. J Cardiovasc Pharmacol 2024; 84:528-538. [PMID: 39027983 DOI: 10.1097/fjc.0000000000001604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/01/2024] [Indexed: 07/20/2024]
Abstract
ABSTRACT Panax notoginseng has the effect of stimulating circulation to end stasis. Our study was designed to evaluate the anti-thrombotic effect of protoparaxotriol saponins (PTS) from P. notoginseng and the involved mechanisms. A thrombosis model was constructed, and the anti-thrombotic activity of PTS was determined by erythrocyte staining, heart rate, and blood flow velocity. In addition, quantitative real-time polymerase chain reaction was used to identify changes in the expression of genes related to coagulation, inflammation, and apoptosis. PTS alleviated arachidonic acid-induced caudal vein thrombosis, restored blood flow, and increased the area of cardiac erythrocyte staining, heart rate, and blood flow velocity. It reduced the ponatinib-induced cerebral thrombus area and decreased the intensity of erythrocyte staining. The quantitative polymerase chain reaction data showed that the anti-thrombotic effect of PTS was mediated by suppression of genes related to coagulation, inflammation, and apoptosis and also involved inhibition of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathways.
Collapse
Affiliation(s)
- Xin Liu
- Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Calì B, Troiani M, Bressan S, Attanasio G, Merler S, Moscarda V, Mosole S, Ricci E, Guo C, Yuan W, Gallagher L, Lundberg A, Bernett I, Figueiredo I, Arzola RA, Abreut EB, D'Ambrosio M, Bancaro N, Brina D, Zumerle S, Pasquini E, Maddalena M, Lai P, Colucci M, Pernigoni N, Rinaldi A, Minardi D, Morlacco A, Moro FD, Sabbadin M, Galuppini F, Fassan M, Rüschoff JH, Moch H, Rescigno P, Francini E, Saieva C, Modesti M, Theurillat JP, Gillessen S, Wilgenbus P, Graf C, Ruf W, de Bono J, Alimonti A. Coagulation factor X promotes resistance to androgen-deprivation therapy in prostate cancer. Cancer Cell 2024; 42:1676-1692.e11. [PMID: 39303726 DOI: 10.1016/j.ccell.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/13/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
Although hypercoagulability is commonly associated with malignancies, whether coagulation factors directly affect tumor cell proliferation remains unclear. Herein, by performing single-cell RNA sequencing (scRNA-seq) of the prostate tumor microenvironment (TME) of mouse models of castration-resistant prostate cancer (CRPC), we report that immunosuppressive neutrophils (PMN-MDSCs) are a key extra-hepatic source of coagulation factor X (FX). FX activation within the TME enhances androgen-independent tumor growth by activating the protease-activated receptor 2 (PAR2) and the phosphorylation of ERK1/2 in tumor cells. Genetic and pharmacological inhibition of factor Xa (FXa) antagonizes the oncogenic activity of PMN-MDSCs, reduces tumor progression, and synergizes with enzalutamide therapy. Intriguingly, F10high PMN-MDSCs express the surface marker CD84 and CD84 ligation enhances F10 expression. Elevated levels of FX, CD84, and PAR2 in prostate tumors associate with worse survival in CRPC patients. This study provides evidence that FXa directly promotes cancer and highlights additional targets for PMN-MDSCs for cancer therapies.
Collapse
Affiliation(s)
- Bianca Calì
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Martina Troiani
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Silvia Bressan
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy
| | - Giuseppe Attanasio
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Sara Merler
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Section of Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | - Viola Moscarda
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Section of Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Simone Mosole
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Elena Ricci
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Christina Guo
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Wei Yuan
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Lewis Gallagher
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Arian Lundberg
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Ilona Bernett
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Ines Figueiredo
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Rydell Alvarez Arzola
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Department of Immunoregulation, Immunology and Immunotherapy Division, Center of Molecular Immunology, La Habana 3GGH+C9G, Cuba
| | - Ernesto Bermudez Abreut
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Department of Immunoregulation, Immunology and Immunotherapy Division, Center of Molecular Immunology, La Habana 3GGH+C9G, Cuba
| | - Mariantonietta D'Ambrosio
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Nicolò Bancaro
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Daniela Brina
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Sara Zumerle
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, University of Padova, 35121 Padova, Italy
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Martino Maddalena
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Ping Lai
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Manuel Colucci
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Nicolò Pernigoni
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Davide Minardi
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Alessandro Morlacco
- Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Fabrizio Dal Moro
- Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Marianna Sabbadin
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Francesca Galuppini
- Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Jan Hendrik Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich (USZ), 8091 Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich (USZ), 8091 Zurich, Switzerland
| | | | - Edoardo Francini
- Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy
| | - Calogero Saieva
- Cancer Risk Factors and Lifestyle Epidemiology Unit - ISPRO, 50139 Florence, Italy
| | - Mikol Modesti
- Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland
| | - Jean-Philippe Theurillat
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Silke Gillessen
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland
| | - Petra Wilgenbus
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Claudine Graf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Johann de Bono
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, University of Padova, 35121 Padova, Italy; Department of Health Sciences and Technology (D-HEST) ETH Zurich, 8092 Zurich, Switzerland.
| |
Collapse
|
8
|
Todorova VK, Azhar G, Stone A, Malapati SJ, Che Y, Zhang W, Makhoul I, Wei JY. Neutrophil Biomarkers Can Predict Cardiotoxicity of Anthracyclines in Breast Cancer. Int J Mol Sci 2024; 25:9735. [PMID: 39273682 PMCID: PMC11395913 DOI: 10.3390/ijms25179735] [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: 08/11/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Doxorubicin (DOX), a commonly used anticancer agent, causes cardiotoxicity that begins with the first dose and may progress to heart failure years after treatment. An inflammatory response associated with neutrophil recruitment has been recognized as a mechanism of DOX-induced cardiotoxicity. This study aimed to validate mRNA expression of the previously identified biomarkers of DOX-induced cardiotoxicity, PGLYRP1, CAMP, MMP9, and CEACAM8, and to assay their protein expression in the peripheral blood of breast cancer patients. Blood samples from 40 breast cancer patients treated with DOX-based chemotherapy were collected before and after the first chemotherapy cycle and > 2 years after treatment. The protein and gene expression of PGLYRP1/Tag7, CAMP/LL37, MMP9/gelatinase B, and CEACAM8/CD66b were determined using ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of each candidate biomarker. Patients with cardiotoxicity (n = 20) had significantly elevated levels of PGLYRP1, CAMP, MMP9, and CEACAM8 at baseline, after the first dose of DOX-based chemotherapy, and at > 2 years after treatment relative to patients without cardiotoxicity (n = 20). The first dose of DOX induced significantly higher levels of all examined biomarkers in both groups of patients. At > 2 years post treatment, the levels of all but MMP9 dropped below the baseline. There was a good correlation between the expression of mRNA and the target proteins. We demonstrate that circulating levels of PGLYRP1, CAMP, MMP9, and CEACAM8 can predict the cardiotoxicity of DOX. This novel finding may be of value in the early identification of patients at risk for cardiotoxicity.
Collapse
Affiliation(s)
- Valentina K Todorova
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Gohar Azhar
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Annjanette Stone
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Sindhu J Malapati
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yingni Che
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wei Zhang
- Department of Mathematics and Statistics, University of Arkansas at Little Rock, Little Rock, AR 72205, USA
| | - Issam Makhoul
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
9
|
Zhang Z, Zhou X, Zhou X, Cheng Z, Hu Y. Role of Platelets and Their Interaction with Immune Cells in Venous Thromboembolism. Semin Thromb Hemost 2024. [PMID: 39214148 DOI: 10.1055/s-0044-1789022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Venous thromboembolism (VTE) represents a significant global health challenge, ranking as the third leading cause of cardiovascular-related mortality. VTE pervades diverse clinical specialties, posing substantial risks to patient well-being and imposing considerable economic strains on health care systems. While platelets have long been recognized as pivotal players in hemostasis, emerging evidence underscores their multifaceted immune functions and their capacity to engage in crosstalk with other immune cells, such as neutrophils, thereby fostering immune-related thrombosis. Notably, investigations have elucidated the pivotal role of platelets in the pathogenesis of VTE. This review provides a comprehensive overview of platelet physiology, encompassing their activation, secretion dynamics, and implications in VTE. Moreover, it delineates the impact of platelet interactions with various immune cells on the initiation and progression of VTE, explores the correlation between platelet-related laboratory markers and VTE, and elucidates the role of platelets in thrombosis regression.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| | - Xianghui Zhou
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| | - Xin Zhou
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| | - Zhipeng Cheng
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| | - Yu Hu
- Department of Hematology, Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Wuhan, China
| |
Collapse
|
10
|
Liu W, Cheng G, Cui H, Tian Z, Li B, Han Y, Wu JX, Sun J, Zhao Y, Chen T, Yu G. Theoretical basis, state and challenges of living cell-based drug delivery systems. Theranostics 2024; 14:5152-5183. [PMID: 39267776 PMCID: PMC11388066 DOI: 10.7150/thno.99257] [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: 06/04/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
The therapeutic efficacy of drugs is determined, to a certain extent, by the efficiency of drug delivery. The low efficiency of drug delivery systems (DDSs) is frequently associated with serious toxic side effects and can even prove fatal in certain cases. With the rapid development of technology, drug delivery has evolved from using traditional frameworks to using nano DDSs (NDDSs), endogenous biomaterials DDSs (EBDDSs), and living cell DDSs (LCDDSs). LCDDSs are receiving widespread attention from researchers at present owing to the unique advantages of living cells in targeted drug delivery, including their excellent biocompatibility properties, low immunogenicity, unique biological properties and functions, and role in the treatment of diseases. However, the theoretical basis and techniques involved in the application of LCDDSs have not been extensively summarized to date. Therefore, this review comprehensively summarizes the properties and applications of living cells, elaborates the various drug loading approaches and controlled drug release, and discusses the results of clinical trials. The review also discusses the current shortcomings and prospects for the future development of LCDDSs, which will serve as highly valuable insights for the development and clinical transformation of LCDDSs in the future.
Collapse
Affiliation(s)
- Wei Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Guowang Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Zhen Tian
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Bowen Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yanhua Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jia-Xin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jie Sun
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yuyue Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Guangtao Yu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
11
|
Flora GD, Ghatge M, Nayak MK, Barbhuyan T, Kumskova M, Chauhan AK. Deletion of pyruvate dehydrogenase kinases reduces susceptibility to deep vein thrombosis in mice. Blood Adv 2024; 8:3906-3913. [PMID: 38838230 PMCID: PMC11321300 DOI: 10.1182/bloodadvances.2024013199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
ABSTRACT Neutrophils contribute to deep vein thrombosis (DVT) by releasing prothrombotic neutrophil extracellular traps (NETs). NET formation (known as NETosis) is an energy-intensive process that requires an increased rate of aerobic glycolysis. The metabolic enzymes pyruvate dehydrogenase kinases (PDKs) inhibit the pyruvate dehydrogenase complex to divert the pyruvate flux from oxidative phosphorylation toward aerobic glycolysis. Herein, we identified that the combined deletion of PDK2 and PDK4 (PDK2/4-/-) renders mice less susceptible to DVT (measured by thrombus incidence, weight, and length) in the inferior vena cava-stenosis model at day 2 after surgery. Compared with wild-type (WT) mice, the venous thrombus obtained from PDK2/4-/- mice exhibited reduced citrullinated histone content, a known marker of NETs. In line with in vivo observations, phorbol 12-myristate 13-acetate (PMA)-stimulated PDK2/4-/- neutrophils displayed reduced NETosis and secretion of cathepsin G and elastase compared with PMA-stimulated WT neutrophils. The formation of platelet aggregates mediated by PMA-stimulated PDK2/4-/- neutrophils were significantly reduced compared with PMA-stimulated WT neutrophils. Finally, PDK2/4-/- neutrophils exhibited reduced levels of intracellular Ca2+ concentration, extracellular signal-regulated kinase 1/2 (Erk1/2) phosphorylation, and glycolytic proton efflux rate (a measure of aerobic glycolysis), known to facilitate NETosis. Together, these findings elucidate, to our knowledge, for the first time, the fundamental role of PDK2/4 in regulating NETosis and acute DVT.
Collapse
Affiliation(s)
- Gagan D. Flora
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Madankumar Ghatge
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Manasa K. Nayak
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Tarun Barbhuyan
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Mariia Kumskova
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Anil K. Chauhan
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
12
|
Zhang S, Li Y, Zhang J, Sun Y, Chu X, Gui X, Tong H, Ding Y, Ju W, Xu M, Li Z, Zeng L, Xu K, Qiao J. Platelet-Derived TGF-β1 Promotes Deep Vein Thrombosis. Thromb Haemost 2024; 124:641-648. [PMID: 38151026 DOI: 10.1055/a-2235-7485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
BACKGROUND Transforming growth factor-β1 (TGF-β1) modulates multiple cellular functions during development and tissue homeostasis. A large amount of TGF-β1 is stored in platelet α-granules and released upon platelet activation. Whether platelet-derived TGF-β1 plays a role in venous thrombosis remains unclear. This study intends to assess the role of platelet-derived TGF-β1 in the development of venous thrombosis in mice. MATERIAL AND METHODS TGF-β1flox/flox and platelet-specific TGF-β1-/- mice were utilized to assess platelet function in vitro, arterial thrombosis induced by FeCl3, tail bleeding time, prothrombin time (PT), activated partial thromboplastin time (APTT), and deep vein thrombosis induced through ligation of the inferior vena cava (IVC). The IVC sample was collected to measure accumulation of neutrophils, monocytes, and the formation of neutrophil extracellular traps (NETs) by immunofluorescence staining. RESULTS TGF-β1 deficiency in platelets did not affect the number of circulating platelets, platelet aggregation, adenosine triphosphate release, and integrin αIIbβ3 activation. Meanwhile, TGF-β1 deficiency did not alter the arterial thrombus formation, hemostasis, and coagulation time (PT and APTT), but significantly impaired venous thrombus formation, inhibited the recruitment and accumulation of neutrophils and monocytes in thrombi, as well as reduced formation of NETs and platelet-neutrophil complex. In addition, adoptive transfer of TGF-β1flox/flox platelets to TGF-β1-/- mice rescued the impaired venous thrombus formation, recruitment of leukocytes and monocytes, as well as the NETs formation. CONCLUSION In conclusion, platelet-derived TGF-β1 positively modulates venous thrombus formation in mice, indicating that targeting TGF-β1 might be a novel approach for treating venous thrombosis without increasing the risk of bleeding.
Collapse
Affiliation(s)
- Sixuan Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yingying Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Jie Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yueyue Sun
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xiang Chu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xiang Gui
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Huan Tong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yangyang Ding
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Mengdi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| |
Collapse
|
13
|
Smood B, Smith C, Dori Y, Mavroudis CD, Fuller S, Gaynor JW, Maeda K. Lymphatic failure and lymphatic interventions: Knowledge gaps and future directions for a new frontier in congenital heart disease. Semin Pediatr Surg 2024; 33:151426. [PMID: 38820801 PMCID: PMC11229519 DOI: 10.1016/j.sempedsurg.2024.151426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Lymphatic failure is a broad term that describes the lymphatic circulation's inability to adequately transport fluid and solutes out of the interstitium and into the systemic venous circulation, which can result in dysfunction and dysregulation of immune responses, dietary fat absorption, and fluid balance maintenance. Several investigations have recently elucidated the nexus between lymphatic failure and congenital heart disease, and the associated morbidity and mortality is now well-recognized. However, the precise pathophysiology and pathogenesis of lymphatic failure remains poorly understood and relatively understudied, and there are no targeted therapeutics or interventions to reliably prevent its development and progression. Thus, there is growing enthusiasm towards the development and application of novel percutaneous and surgical lymphatic interventions. Moreover, there is consensus that further investigations are needed to delineate the underlying mechanisms of lymphatic failure, which could help identify novel therapeutic targets and develop innovative procedures to improve the overall quality of life and survival of these patients. With these considerations, this review aims to provide an overview of the lymphatic circulation and its vasculature as it relates to current understandings into the pathophysiology and pathogenesis of lymphatic failure in patients with congenital heart disease, while also summarizing strategies for evaluating and managing lymphatic complications, as well as specific areas of interest for future translational and clinical research efforts.
Collapse
Affiliation(s)
- Benjamin Smood
- Division of Cardiothoracic Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States of America; Division of Cardiovascular Surgery, Department of Surgery, The University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America.
| | - Christopher Smith
- Jill and Mark Fishman Center for Lymphatic Disorders, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Department of Cardiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104 United States of America
| | - Yoav Dori
- Jill and Mark Fishman Center for Lymphatic Disorders, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Department of Cardiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104 United States of America
| | - Constantine D Mavroudis
- Division of Cardiothoracic Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States of America; Division of Cardiovascular Surgery, Department of Surgery, The University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Stephanie Fuller
- Division of Cardiothoracic Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States of America; Division of Cardiovascular Surgery, Department of Surgery, The University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - J William Gaynor
- Division of Cardiothoracic Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States of America; Division of Cardiovascular Surgery, Department of Surgery, The University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Katsuhide Maeda
- Division of Cardiothoracic Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States of America; Division of Cardiovascular Surgery, Department of Surgery, The University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America; Jill and Mark Fishman Center for Lymphatic Disorders, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
14
|
Wu X, Yang Y. Neutrophil extracellular traps (NETs) and fibrotic diseases. Int Immunopharmacol 2024; 133:112085. [PMID: 38626550 DOI: 10.1016/j.intimp.2024.112085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/18/2024]
Abstract
Fibrosis, a common cause and serious outcome of organ failure that can affect any organ, is responsible for up to 45% of all deaths in various clinical settings. Both preclinical models and clinical trials investigating various organ systems have shown that fibrosis is a highly dynamic process. Although many studies have sought to gain understanding of the mechanism of fibrosis progression, their findings have been mixed. In recent years, increasing evidence indicates that neutrophil extracellular traps (NETs) are involved in many inflammatory and autoimmune disorders and participate in the regulation of fibrotic processes in various organs and systems. In this review, we summarize the current understanding of the role of NETs in fibrosis development and progression and their possibility as therapeutic targets.
Collapse
Affiliation(s)
- Xiaojiao Wu
- School of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yang Yang
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Pandey N, Kaur H, Chorawala MR, Anand SK, Chandaluri L, Butler ME, Aishwarya R, Gaddam SJ, Shen X, Alfaidi M, Wang J, Zhang X, Beedupalli K, Bhuiyan MS, Bhuiyan MAN, Buchhanolla P, Rai P, Shah R, Chokhawala H, Jordan JD, Magdy T, Orr AW, Stokes KY, Rom O, Dhanesha N. Interactions between integrin α9β1 and VCAM-1 promote neutrophil hyperactivation and mediate poststroke DVT. Blood Adv 2024; 8:2104-2117. [PMID: 38498701 PMCID: PMC11063402 DOI: 10.1182/bloodadvances.2023012282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/20/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
ABSTRACT Venous thromboembolic events are significant contributors to morbidity and mortality in patients with stroke. Neutrophils are among the first cells in the blood to respond to stroke and are known to promote deep vein thrombosis (DVT). Integrin α9 is a transmembrane glycoprotein highly expressed on neutrophils and stabilizes neutrophil adhesion to activated endothelium via vascular cell adhesion molecule 1 (VCAM-1). Nevertheless, the causative role of neutrophil integrin α9 in poststroke DVT remains unknown. Here, we found higher neutrophil integrin α9 and plasma VCAM-1 levels in humans and mice with stroke. Using mice with embolic stroke, we observed enhanced DVT severity in a novel model of poststroke DVT. Neutrophil-specific integrin α9-deficient mice (α9fl/flMrp8Cre+/-) exhibited a significant reduction in poststroke DVT severity along with decreased neutrophils and citrullinated histone H3 in thrombi. Unbiased transcriptomics indicated that α9/VCAM-1 interactions induced pathways related to neutrophil inflammation, exocytosis, NF-κB signaling, and chemotaxis. Mechanistic studies revealed that integrin α9/VCAM-1 interactions mediate neutrophil adhesion at the venous shear rate, promote neutrophil hyperactivation, increase phosphorylation of extracellular signal-regulated kinase, and induce endothelial cell apoptosis. Using pharmacogenomic profiling, virtual screening, and in vitro assays, we identified macitentan as a potent inhibitor of integrin α9/VCAM-1 interactions and neutrophil adhesion to activated endothelial cells. Macitentan reduced DVT severity in control mice with and without stroke, but not in α9fl/flMrp8Cre+/- mice, suggesting that macitentan improves DVT outcomes by inhibiting neutrophil integrin α9. Collectively, we uncovered a previously unrecognized and critical pathway involving the α9/VCAM-1 axis in neutrophil hyperactivation and DVT.
Collapse
Affiliation(s)
- Nilesh Pandey
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Harpreet Kaur
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Mehul R. Chorawala
- Department of Pharmacology and Pharmacy Practice, L.M. College of Pharmacy, Ahmedabad, India
| | - Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Lakshmi Chandaluri
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Megan E. Butler
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Richa Aishwarya
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Shiva J. Gaddam
- Department of Hematology and Oncology and Feist Weiller Cancer Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Xinggui Shen
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Mabruka Alfaidi
- Division of Cardiology, Department of Internal Medicine, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Jian Wang
- Bioinformatics and Modeling Core, Center for Applied Immunology and Pathological Processes, Department of Microbiology and Immunology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Xiaolu Zhang
- Bioinformatics and Modeling Core, Center for Applied Immunology and Pathological Processes, Department of Microbiology and Immunology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Kavitha Beedupalli
- Department of Hematology and Oncology and Feist Weiller Cancer Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Md. Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | | | - Prabandh Buchhanolla
- Department of Neurology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Prashant Rai
- Department of Neurology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Rahul Shah
- Department of Neurology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Himanshu Chokhawala
- Department of Neurology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - J. Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Tarek Magdy
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - A. Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Karen Y. Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Nirav Dhanesha
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| |
Collapse
|
16
|
Bo Y, Lu Q, Li B, Sha R, Yu H, Miao C. The role of platelets in central hubs of inflammation: A literature review. Medicine (Baltimore) 2024; 103:e38115. [PMID: 38728509 PMCID: PMC11081549 DOI: 10.1097/md.0000000000038115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Platelets are increasingly recognized for their multifaceted roles in inflammation beyond their traditional involvement in haemostasis. This review consolidates knowledge on platelets as critical players in inflammatory responses. This study did an extensive search of electronic databases and identified studies on platelets in inflammation, focusing on molecular mechanisms, cell interactions, and clinical implications, emphasizing recent publications. Platelets contribute to inflammation via surface receptors, release of mediators, and participation in neutrophil extracellular trap formation. They are implicated in diseases like atherosclerosis, rheumatoid arthritis, and sepsis, highlighting their interaction with immune cells as pivotal in the onset and resolution of inflammation. Platelets are central to regulating inflammation, offering new therapeutic targets for inflammatory diseases. Future research should explore specific molecular pathways of platelets in inflammation for therapeutic intervention.
Collapse
Affiliation(s)
- Yan Bo
- College of Medicine, Northwest Minzu University, Lanzhou, China
| | - Qingyang Lu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR of China
| | - Beilei Li
- Department of Rehabilitation Medicine, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Ren Sha
- School of Economics and Management, Henan Polytechnic University, Jiaozuo, China
| | - Haodong Yu
- School of Economic Crime Investigation, Jiangxi Police Academy, Nanchang, China
| | - Chuhan Miao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR of China
| |
Collapse
|
17
|
Kuang H, Zhu X, Chen H, Tang H, Zhao H. The immunomodulatory mechanism of acupuncture treatment for ischemic stroke: research progress, prospects, and future direction. Front Immunol 2024; 15:1319863. [PMID: 38756772 PMCID: PMC11096548 DOI: 10.3389/fimmu.2024.1319863] [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: 10/11/2023] [Accepted: 04/03/2024] [Indexed: 05/18/2024] Open
Abstract
Ischemic stroke (IS) is one of the leading causes of death and disability. Complicated mechanisms are involved in the pathogenesis of IS. Immunomodulatory mechanisms are crucial to IS. Acupuncture is a traditional non-drug treatment that has been extensively used to treat IS. The exploration of neuroimmune modulation will broaden the understanding of the mechanisms underlying acupuncture treatment. This review summarizes the immune response of immune cells, immune cytokines, and immune organs after an IS. The immunomodulatory mechanisms of acupuncture treatment on the central nervous system and peripheral immunity, as well as the factors that influence the effects of acupuncture treatment, were summarized. We suggest prospects and future directions for research on immunomodulatory mechanisms of acupuncture treatment for IS based on current progress, and we hope that these will provide inspiration for researchers. Additionally, acupuncture has shown favorable outcomes in the treatment of immune-based nervous system diseases, generating new directions for research on possible targets and treatments for immune-based nervous system diseases.
Collapse
Affiliation(s)
- Hongjun Kuang
- Department of Acupuncture and Moxibustion, Shanghai University of Traditional Chinese Medicine, Shenzhen Hospital, Shenzhen, China
- Department of Acupuncture and Moxibustion, Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xinzhou Zhu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Huan Chen
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Science, Beijing, China
| | - Han Tang
- Department of Acupuncture and Moxibustion, Shanghai University of Traditional Chinese Medicine, Shenzhen Hospital, Shenzhen, China
- Department of Acupuncture and Moxibustion, Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Hong Zhao
- Department of Acupuncture and Moxibustion, Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China
| |
Collapse
|
18
|
Grdinic AG, Radovanovic S, Gleditsch J, Jørgensen CT, Asady E, Pettersen HH, Delibasic B, Ghanima W. Developing a machine learning model for bleeding prediction in patients with cancer-associated thrombosis receiving anticoagulation therapy. J Thromb Haemost 2024; 22:1094-1104. [PMID: 38184201 DOI: 10.1016/j.jtha.2023.12.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND Only 1 conventional score is available for assessing bleeding risk in patients with cancer-associated thrombosis (CAT): the CAT-BLEED score. OBJECTIVES Our aim was to develop a machine learning-based risk assessment model for predicting bleeding in CAT and to evaluate its predictive performance in comparison to that of the CAT-BLEED score. METHODS We collected 488 attributes (clinical data, biochemistry, and International Classification of Diseases, 10th Revision, diagnosis) in 1080 unique patients with CAT. We compared CAT-BLEED score, Ridge and Lasso logistic regression, random forest, and Extreme Gradient Boosting (XGBoost) algorithms for predicting major bleeding or clinically relevant nonmajor bleeding occurring 1 to 90 days, 1 to 365 days, and 90 to 455 days after venous thromboembolism (VTE). RESULTS The predictive performances of Lasso logistic regression, random forest, and XGBoost were higher than that of the CAT-BLEED score in the prediction of bleeding occurring 1 to 90 days and 1 to 365 days after VTE. For predicting major bleeding or clinically relevant nonmajor bleeding 1 to 90 days after VTE, the CAT-BLEED score achieved a mean area under the receiver operating characteristic curve (AUROC) of 0.48 ± 0.13, while Lasso logistic regression and XGBoost both achieved AUROCs of 0.64 ± 0.12. For predicting bleeding 1 to 365 days after VTE, the CAT-BLEED score achieved a mean AUROC of 0.47 ± 0.08, while Lasso logistic regression and XGBoost achieved AUROCs of 0.64 ± 0.08 and 0.59 ± 0.08, respectively. CONCLUSION This is the first machine learning-based risk model for bleeding prediction in patients with CAT receiving anticoagulation therapy. Its predictive performance was higher than that of the conventional CAT-BLEED score. With further development, this novel algorithm might enable clinicians to perform personalized anticoagulation strategies with improved clinical outcomes.
Collapse
Affiliation(s)
- Aleksandra G Grdinic
- Department of Cardiology, Østfold Hospital, Sarpsborg, Norway; Department of Research, Østfold Hospital, Sarpsborg, Norway.
| | - Sandro Radovanovic
- Faculty of Organizational Sciences, University of Belgrade, Belgrade, Serbia
| | - Jostein Gleditsch
- Department of Radiology, Østfold Hospital, Sarpsborg, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Camilla Tøvik Jørgensen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Emergency Medicine, Østfold Hospital, Sarpsborg, Norway
| | - Elia Asady
- Department of Research, Østfold Hospital, Sarpsborg, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Boris Delibasic
- Faculty of Organizational Sciences, University of Belgrade, Belgrade, Serbia
| | - Waleed Ghanima
- Department of Research, Østfold Hospital, Sarpsborg, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Hematology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
19
|
Nguyen HT, Vu MP, Nguyen TTM, Nguyen TT, Kieu TVO, Duong HY, Pham PT, Hoang TH. Association of the neutrophil-to-lymphocyte ratio with the occurrence of venous thromboembolism and arterial thrombosis. J Int Med Res 2024; 52:3000605241240999. [PMID: 38606734 PMCID: PMC11015807 DOI: 10.1177/03000605241240999] [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: 11/29/2023] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
OBJECTIVE This study aimed to assess the association of the neutrophil-to-lymphocyte ratio (NLR) with the occurrence of venous thromboembolism (VTE) and arterial thrombosis (AT). METHODS This was a retrospective cross-sectional study including 585 medical records obtained from all consecutive patients who were suspected of having thrombosis. RESULTS The AT group had a higher neutrophil count and NLR and a lower lymphocyte count than the non-thrombosis group. Receiver operating characteristic curve analysis showed the ability of the NLR to predict the presence of AT. The cut-off value for the NLR was 4.44. No distinction was found in the NLR between the VTE and non-thrombosis groups. Regression analysis showed that a high NLR was an independent factor related to the presence of AT. Patients with an NLR ≥ 4.44 had a higher risk of AT than those with an NLR < 4.44 (odds ratio = 2.015, 95% confidence interval: 1.180-3.443). CONCLUSION A high NLR may be considered a predictive factor for the occurrence of AT, but an association with the presence of VTE was not found.
Collapse
Affiliation(s)
- Ha Thanh Nguyen
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
| | - Minh Phuong Vu
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Thi Tuyet Mai Nguyen
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Tuan Tung Nguyen
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Thi Van Oanh Kieu
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Hai Yen Duong
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Phuong Thao Pham
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Thi Hue Hoang
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| |
Collapse
|
20
|
Gao X, Zhao X, Li J, Liu C, Li W, Zhao J, Li Z, Wang N, Wang F, Dong J, Yan X, Zhang J, Hu X, Jin J, Mang G, Ma R, Hu S. Neutrophil extracellular traps mediated by platelet microvesicles promote thrombosis and brain injury in acute ischemic stroke. Cell Commun Signal 2024; 22:50. [PMID: 38233928 PMCID: PMC10795390 DOI: 10.1186/s12964-023-01379-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/01/2023] [Indexed: 01/19/2024] Open
Abstract
AIMS Neutrophil extracellular traps (NETs) have been implicated in thrombotic diseases. There is no definitive explanation for how NETs form during acute ischemic strokes (AIS). The purpose of our study was to investigate the potential mechanism and role of NETs formation in the AIS process. METHODS As well as 45 healthy subjects, 45 patients with AIS had ELISA tests performed to detect NET markers. Expression of high-mobility group box 1 (HMGB1) on platelet microvesicles (PMVs) was analyzed by flow cytometry in healthy subjects and AIS patients' blood samples. We established middle cerebral artery occlusion (MCAO) mice model to elucidate the interaction between PMPs and NETs. RESULTS A significant elevation in NET markers was found in patient plasma in AIS patients, and neutrophils generated more NETs from patients' neutrophils. HMGB1 expression was upregulated on PMVs from AIS patients and induced NET formation. NETs enhanced Procoagulant activity (PCA) through tissue factor and via platelet activation. Targeting lactadherin in genetical and in pharmacology could regulate the formation of NETs in MCAO model. CONCLUSIONS NETs mediated by PMVs derived HMGB1 exacerbate thrombosis and brain injury in AIS. Video Abstract.
Collapse
Affiliation(s)
- Xin Gao
- Department of Neurosurgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyi Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jiacheng Li
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Chang Liu
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Wenqiang Li
- Department of Vascular Surgery, Jinshan Hospital of Fudan University, Shanghai, China
| | - Junjie Zhao
- Department of General Surgery, Changsha Fourth Hospital, Changsha, China
| | - Zhixi Li
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Nan Wang
- Department of Neurosurgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fang Wang
- Department of Neurosurgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Dong
- Department of Neurosurgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiuwei Yan
- Department of Neurosurgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiheng Zhang
- Department of Neurosurgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xueyan Hu
- Department of Neurosurgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiaqi Jin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Ge Mang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang Province, China.
| | - Ruishuang Ma
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Ningbo University, Ningbo, China.
- Department of Radiotherapy and Chemotherapy, Ningbo First Hospital of Ningbo, Ningbo, 315000, China.
| | - Shaoshan Hu
- Department of Neurosurgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
21
|
Gao C, Wang H, Liu H. Intramuscular Hemangioma of the Triceps Brachii Muscle: A Case Report. Int Med Case Rep J 2024; 17:31-34. [PMID: 38249659 PMCID: PMC10800111 DOI: 10.2147/imcrj.s441846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction Intramuscular hemangioma (IMH) is a rare type of benign tumor that represents less than 1% of all hemangiomas. Chronic pain and a palpable mass are the most common symptoms. Due to the atypical clinical characteristics of the disease, accurate diagnosis is difficult. Misdiagnosis of IMH as malignancy can occur due to similarities in imaging features between IMH and malignancy. To diagnose IMH accurately, multiple imaging modalities, including X-ray, MRI, CT, and 18F-FDG PET/CT, can be used. However, the final diagnosis of IMH is confirmed through histopathological examination. Case This case reports a 16-year-old girl diagnosed with IMH in the triceps brachii muscle. Seek medical attention due to pain and discomfort in the left shoulder. Initial imaging with contrast-enhanced MRI and CT suggested synovial sarcomata. The moderate uptake of FDG on positron emission tomography/computed tomography (PET/CT) also raised suspicions of malignancy. The pathological findings revealed an intramuscular hemangioma with thrombosis and thrombus organization. Conclusion The accurate diagnosis of IMH can be challenging due to the absence of distinct clinical symptoms and imaging findings. When evaluating periarticular intramuscular lesions, IMH should be considered if the MRI shows mixed signals with heterogeneous enhancement. Despite the moderate uptake of FDG seen in some IMH cases, it should not automatically rule out the possibility of IMH. Hence, a combination of imaging modalities and histopathological examination is crucial in ensuring a correct diagnosis of IMH.
Collapse
Affiliation(s)
- Cailiang Gao
- Department of Nuclear Medicine, Three Gorges Hospital, Chongqing University, Chongqing, People’s Republic of China
| | - Hu Wang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huiting Liu
- Department of Nuclear Medicine, Three Gorges Hospital, Chongqing University, Chongqing, People’s Republic of China
| |
Collapse
|
22
|
Kryukov AI, Chumakov PL, Tovmasyan AS, Yanovsky VV, Kishinevskii AE, Shvedov NV, Mosin VV, Artemyeva-Karelova AV. [Comparative analysis of different types of tampons used after septoplasty, conchotomy of the inferior turbinates]. Vestn Otorinolaringol 2024; 89:16-22. [PMID: 39545756 DOI: 10.17116/otorino20248905116] [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: 11/17/2024]
Abstract
In otorhinolaryngology, the question of improving the tactics of early postoperative management in patients who have undergone surgery on the structures of the nasal cavity remains open. The timing of rehabilitation, the possible outcome of postoperative inflammation of the mucous membrane and the patient's ability to work depend on the choice of tactics. The nasal cavity differs from other anatomical areas in its rich blood supply, and therefore the problem of adequate hemostasis in the early stages after undergoing surgical treatment is still extremely relevant. OBJECTIVE To study the methods of postoperative hemostasis and the degree of traumatization of the nasal mucosa when using various types of tamponade in patients who underwent septoplasty and sparing lower conchotomy. MATERIAL AND METHODS We operated on and examined 40 patients. The patients were divided into four groups: in patients of group 1 (n=10), the operation was completed with nasal cavity tamponade with gauze swabs; in patients of group 2 (n=10), nasal cavity tamponade was performed with Merocel tampons (Medtronic Xomed, Inc., USA); in patients of group 3 (n=10) tamponade was performed with elastic finger tampons; for patients of group 4 (n=10) tamponade was performed with filling hydrotampons. In the postoperative period, the severity of pain during nasal tamponade was assessed using a visual analogue scale (VAS), the volume of nasal blood loss during tampon removal, depending on the type of tamponade, the presence/absence of recurrent nosebleeds in the early postoperative period. To objectively assess the severity of the inflammatory reaction of the nasal mucosa against the background of tamponade, a cytological examination of smear prints from the surface of the tampon was performed. RESULTS The severity of pain was insignificant in patients of all four groups. The average VAS score in patients of group 1 (gauze tampons) was 0.94±0.16 points; group 2 (Merocel tampons) - 0.92±0.14 points; group 3 (elastic tampons) - 0.72±0.09 points; group 4 (filling hydrotampons) - 0.96±0.13 points (p<0.05). The volume of blood loss after evacuation of gauze tampons and Merocel tampons was 114.3±35.1 ml and 140.1±22.2 ml, respectively, which exceeded the volume of blood loss when removing elastic tampons (30.4±13.1 ml) and hydrotampons (23.1±11.0 ml) by more than 80%; patients with intranasal elastic tampons and original hydrotampons with internal gluings had no recurrence of bleeding in the early postoperative period, unlike patients with tamponade with gauze turunds and Merocel; according to the results of cytological examination, more pronounced destructive changes in the ciliated epithelium were noted when using gauze turunds and Merocel tampons. CONCLUSIONS In the first 24 hours after surgery, regardless of the type of tampons used, there was consistent hemostasis and mild pain syndrome - less than 1.0 on a visual analogue scale. The maximum amount of blood loss after evacuation of tampons was noted when using Merocel tampons and gauze turundas, the minimum amount of blood loss was after removal of hydrotampons with internal gluings. The proportion of patients with recurrent bleeding in the early postoperative period after gauze tamponade and Merocel tampons was 10% and 20%, respectively, whereas recurrence of bleeding in patients with original tampons with internal gluings and elastic tampons was not observed.
Collapse
Affiliation(s)
- A I Kryukov
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - P L Chumakov
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - A S Tovmasyan
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - V V Yanovsky
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - A E Kishinevskii
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - N V Shvedov
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - V V Mosin
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | | |
Collapse
|
23
|
Maisat W, Hou L, Sandhu S, Sin YC, Kim S, Pelt HV, Chen Y, Emani S, Kong SW, Emani S, Ibla J, Yuki K. Neutrophil extracellular traps formation is associated with postoperative complications in neonates and infants undergoing congenital cardiac surgery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572768. [PMID: 38187754 PMCID: PMC10769315 DOI: 10.1101/2023.12.21.572768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Pediatric patients with congenital heart diseases (CHD) often undergo surgical repair on cardiopulmonary bypass (CPB). Despite a significant medical and surgical improvement, the mortality of neonates and infants remains high. Damage-associated molecular patterns (DAMPs) are endogenous molecules released from injured/damaged tissues as danger signals. We examined 101 pediatric patients who underwent congenital cardiac surgery on CPB. The mortality rate was 4.0%, and the complication rate was 31.6%. We found that neonates/infants experienced multiple complications most, consistent with the previous knowledge. Neonates and infants in the complication group had received more transfusion intraoperatively than the non-complication arm with lower maximum amplitude (MA) on rewarming CPB thromboelastography (TEG). Despite TEG profiles were comparable at ICU admission between the two groups, the complication arm had higher postoperative chest tube output, requiring more blood transfusion. The complication group showed greater neutrophil extracellular traps (NETs) formation at the end of CPB and postoperatively. Plasma histones and high mobility group box 1 (HMGB1) levels were significantly higher in the complication arm. Both induced NETs in vitro and in vivo . As histones and HMGB1 target Toll-like receptor (TLR)2 and TLR4, their mRNA expression in neutrophils was upregulated in the complication arm. Taken together, NETs play a major role in postoperative complication in pediatric cardiac surgery and would be considered a target for intervention. Key points Neonates and infants showed highest postoperative complications with more upregulation of inflammatory transcriptomes of neutrophils.Neonates and infants with organ dysfunction had more NETs formation with higher plasma histones and HMGB1 levels.
Collapse
|
24
|
Chooklin S, Chuklin S. The role of neutrophil extracellular traps in thrombosis. EMERGENCY MEDICINE 2023; 19:448-457. [DOI: 10.22141/2224-0586.19.7.2023.1627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
According to the cellular model of hemostasis, the process of blood coagulation is presented in the form of three phases: initiation, amplification and propagation, each of them includes several consecutive stages. At the same time, thrombus formation is often explained by Virchow’s triad: blood stasis, damage to the blood vessel walls, and hypercoagulation. Classically, the appearance of one of the three mentioned parameters can lead to thrombus formation. Over the past decade, our knowledge of the cross-talk between coagulation, inflammation, and innate immune activation and the involvement of neutrophil extracellular traps in these processes has expanded. This brief review shows their role in thrombosis through the mechanisms of activation of platelets, complement, interaction with blood coagulation factors and damage to the vascular endothelium. We searched the literature in the MEDLINE database on the PubMed platform.
Collapse
|
25
|
Flora GD, Nayak MK, Ghatge M, Chauhan AK. Metabolic targeting of platelets to combat thrombosis: dawn of a new paradigm? Cardiovasc Res 2023; 119:2497-2507. [PMID: 37706546 PMCID: PMC10676458 DOI: 10.1093/cvr/cvad149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/29/2023] [Accepted: 07/18/2023] [Indexed: 09/15/2023] Open
Abstract
Current antithrombotic therapies used in clinical settings target either the coagulation pathways or platelet activation receptors (P2Y12 or GPIIb/IIIa), as well as the cyclooxygenase (COX) enzyme through aspirin. However, they are associated with bleeding risk and are not suitable for long-term use. Thus, novel strategies which provide broad protection against platelet activation with minimal bleeding risks are required. Regardless of the nature of agonist stimulation, platelet activation is an energy-intensive and ATP-driven process characterized by metabolic switching toward a high rate of aerobic glycolysis, relative to oxidative phosphorylation (OXPHOS). Consequently, there has been considerable interest in recent years in investigating whether targeting metabolic pathways in platelets, especially aerobic glycolysis and OXPHOS, can modulate their activation, thereby preventing thrombosis. This review briefly discusses the choices of metabolic substrates available to platelets that drive their metabolic flexibility. We have comprehensively elucidated the relevance of aerobic glycolysis in facilitating platelet activation and the underlying molecular mechanisms that trigger this switch from OXPHOS. We have provided a detailed account of the antiplatelet effects of targeting vital metabolic checkpoints such as pyruvate dehydrogenase kinases (PDKs) and pyruvate kinase M2 (PKM2) that preferentially drive the pyruvate flux to aerobic glycolysis. Furthermore, we discuss the role of fatty acids and glutamine oxidation in mitochondria and their subsequent role in driving OXPHOS and platelet activation. While the approach of targeting metabolic regulatory mechanisms in platelets to prevent their activation is still in a nascent stage, accumulating evidence highlights its beneficial effects as a potentially novel antithrombotic strategy.
Collapse
Affiliation(s)
- Gagan D Flora
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA, USA
| | - Manasa K Nayak
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA, USA
| | - Madankumar Ghatge
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA, USA
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
26
|
Falcione S, Spronk E, Munsterman D, Joy T, Boghozian R, Jickling GC. Sex Differences in Thrombin Generation in Patients with Acute Ischemic Stroke. Transl Stroke Res 2023:10.1007/s12975-023-01200-1. [PMID: 37987986 DOI: 10.1007/s12975-023-01200-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/22/2023]
Abstract
Sex differences in stroke exist, including variation in stroke risk and outcome. Differences in thrombin generation may contribute to this variation between females and males. To examine this, we assessed sex differences in thrombin generation between females and males with acute ischemic stroke and the relationship to blood cell gene expression. In 97 patients with acute ischemic stroke, thrombin generation was measured by thrombin generation assay. Blood cell gene expression was measured by microarray. Differences in thrombin generation between sexes were identified and the relationship to blood cell gene expression examined. Genes associated with sex differences in thrombin generation were analyzed by functional pathway analysis. Females and males had similar overall capacity to generate thrombin. The peak thrombin generated in females was 468.8 nM (SD 91.6), comparable to males (479.3nM;SD 90.8; p = 0.58). Lag time, time to peak thrombin, and endogenous thrombin potential were also similar between females and males. While overall thrombin generation was comparable between females and males with stroke, differences in genes that promote this thrombin generation exist. Females with high peak thrombin had an increase in genes that promote thrombosis, and platelet activation. In contrast, males with high peak thrombin had a decrease in genes involved in thrombus degradation. Females and males with acute ischemic stroke have similar capacity to generate thrombin, however, differences may exist in how this thrombin generation is achieved, with females having increased thrombin signaling, and platelet activation, and males having decreased thrombus degradation. This suggests regulatory differences in thrombosis may exist between females and males that may contribute to sex differences in stroke.
Collapse
Affiliation(s)
- Sarina Falcione
- Department of Medicine, Division of Neurology, University of Alberta, 11315 87th Ave NW, Edmonton, T6G 2H5, Canada.
| | - Elena Spronk
- Department of Medicine, Division of Neurology, University of Alberta, 11315 87th Ave NW, Edmonton, T6G 2H5, Canada
| | - Danielle Munsterman
- Department of Medicine, Division of Neurology, University of Alberta, 11315 87th Ave NW, Edmonton, T6G 2H5, Canada
| | - Twinkle Joy
- Department of Medicine, Division of Neurology, University of Alberta, 11315 87th Ave NW, Edmonton, T6G 2H5, Canada
| | - Roobina Boghozian
- Department of Medicine, Division of Neurology, University of Alberta, 11315 87th Ave NW, Edmonton, T6G 2H5, Canada
| | - Glen C Jickling
- Department of Medicine, Division of Neurology, University of Alberta, 11315 87th Ave NW, Edmonton, T6G 2H5, Canada
| |
Collapse
|
27
|
Wang X, Chen H, Song F, Zuo K, Chen X, Zhang X, Liang L, Ta Q, Zhang L, Li J. Resveratrol: a potential medication for the prevention and treatment of varicella zoster virus-induced ischemic stroke. Eur J Med Res 2023; 28:400. [PMID: 37794518 PMCID: PMC10552394 DOI: 10.1186/s40001-023-01291-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/14/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Infection rate of varicella zoster virus (VZV) is 95% in humans, and VZV infection is strongly associated with ischemic stroke (IS). However, the underlying molecular mechanisms of VZV-induced IS are still unclear, and there are no effective agents to treat and prevent VZV-induced IS. OBJECTIVE By integrating bioinformatics, this study explored the interactions between VZV and IS and potential medication to treat and prevent VZV-induced IS. METHODS In this study, the VZV and IS datasets from the GEO database were used to specify the common genes. Then, bioinformatics analysis including Gene Ontology, Kyoto Encyclopedia Genes Genomes and Protein-Protein Interaction network analysis was performed. Further, the hub genes, transcription factor (TF) gene interactions, TF-miRNA co-regulatory network and potential drug were obtained. Finally, validation was performed using molecular docking and molecular dynamics simulations. RESULTS The potential molecular mechanisms of VZV-induced IS were studied using multiple bioinformatics tools. Ten hub genes were COL1A2, DCN, PDGFRB, ACTA2, etc. TF genes and miRNAs included JUN, FOS, CREB, BRCA1, PPARG, STAT3, miR-29, etc. A series of mechanism may be involved, such as inflammation, oxidative stress, blood-brain barrier disruption, foam cell generation and among others. Finally, we proposed resveratrol as a potential therapeutic medicine for the prevention and treatment of VZV-induced IS. Molecular docking and molecular dynamics results showed that resveratrol and hub genes exhibited strong binding score. CONCLUSIONS Resveratrol could be an alternative for the prevention and treatment of VZV-IS. More in vivo and in vitro studies are needed in the future to fully explore the molecular mechanisms between VZV and IS and for medication development.
Collapse
Affiliation(s)
- Xu Wang
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Hao Chen
- Department of Neurovascular Surgery, First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Feiyu Song
- Jilin Connell Pharmaceutical Co., Ltd, JilinJilin, 132013, China
| | - Kuiyang Zuo
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Xin Chen
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Xu Zhang
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Lanqian Liang
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Qiyi Ta
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Lin Zhang
- China-Japan Union Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Jinhua Li
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
28
|
Obradovic S, Dzudovic B, Subotic B, Salinger S, Matijasevic J, Benic M, Kovacevic T, Kovacevic-Kuzmanovic A, Mitevska I, Miloradovic V, Jevtic E, Neskovic A. Association of Blood Leukocytes and Hemoglobin with Hospital Mortality in Acute Pulmonary Embolism. J Clin Med 2023; 12:6269. [PMID: 37834913 PMCID: PMC10573828 DOI: 10.3390/jcm12196269] [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: 08/24/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
This study aimed to assess the prognostic significance of total leukocyte count (TLC) and hemoglobin (Hb) levels upon admission for patients with acute pulmonary embolism (PE), considering the European Society of Cardiology (ESC) model for mortality risk. 1622 patients from a regional PE registry were included. Decision tree statistics were employed to evaluate the prognostic value of TLC and Hb, both independently and in conjunction with the ESC model. The results indicated all-cause and PE-related in-hospital mortality rates of 10.7% and 6.5%, respectively. Subgrouping patients based on TLC cut-off values (≤11.2, 11.2-16.84, >16.84 × 109/L) revealed increasing all-cause mortality risks (7.0%, 11.8%, 30.2%). Incorporating Hb levels (≤126 g/L or above) further stratified the lowest risk group into two strata with all-cause mortality rates of 10.1% and 4.7%. Similar trends were observed for PE-related mortality. Notably, TLC improved risk assessment for intermediate-high-risk patients within the ESC model, while Hb levels enhanced mortality risk stratification for lower-risk PE patients in the ESC model for all-cause mortality. In conclusion, TLC and Hb levels upon admission can refine the ESC model's mortality risk classification for patients with acute PE, providing valuable insights for improved patient management.
Collapse
Affiliation(s)
- Slobodan Obradovic
- Clinic of Cardiology, Military Medical Academy of Belgrade, 11000 Belgrade, Serbia; (S.O.); (B.S.)
- School of Medicine, University of Defense, 11000 Belgrade, Serbia
| | - Boris Dzudovic
- School of Medicine, University of Defense, 11000 Belgrade, Serbia
- Clinic of Emergency Internal Medicine, Military Medical Academy, 11000 Belgrade, Serbia
| | - Bojana Subotic
- Clinic of Cardiology, Military Medical Academy of Belgrade, 11000 Belgrade, Serbia; (S.O.); (B.S.)
| | - Sonja Salinger
- Clinic of Cardiology, Clinical Center Nis, 18000 Nis, Serbia;
- School of Medicine, University of Nis, 18000 Nis, Serbia
| | - Jovan Matijasevic
- Institute of Pulmonary Diseases Vojvodina, Novi Sad, 21204 Sremska Kamenica, Serbia; (J.M.); (M.B.)
- School of Medicine, University of Novi Sad, 24000 Subotica, Serbia
| | - Marija Benic
- Institute of Pulmonary Diseases Vojvodina, Novi Sad, 21204 Sremska Kamenica, Serbia; (J.M.); (M.B.)
| | - Tamara Kovacevic
- Clinic of Cardiology, Clinical Center Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina;
- School of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | | | - Irena Mitevska
- Intensive Care Unit, University Cardiology Clinic, 1000 Skopje, North Macedonia;
| | - Vladimir Miloradovic
- Clinic of Cardiology, Clinical Center Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (E.J.)
- School of Medicine, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ema Jevtic
- Clinic of Cardiology, Clinical Center Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (E.J.)
| | - Aleksandar Neskovic
- Clinic of Cardiology, University Clinical Center Zemun, 11080 Belgrade, Serbia;
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
29
|
Alsubhi YM, Alhadi AH, Hammudah AM, Alahmadi RA, Aljohani AM, Dubai SA, Susi AI, Almuwallad K, Alwasaidi TA. Comparison of laboratory biomarkers for the prediction of in-hospital mortality and severity of acute pulmonary embolism: A multi-center study. Saudi Med J 2023; 44:898-903. [PMID: 37717976 PMCID: PMC10505296 DOI: 10.15537/smj.2023.44.9.20230441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
OBJECTIVES To assess the specificity and sensitivity of prognostic biomarkers in individuals diagnosed with acute pulmonary embolism (PE). METHODS This study retrospectively enrolled 162 patients from the 741 patients who were hospitalized with acute PE and diagnosed using pulmonary computed tomography (CT) angiogram at 5 hospitals in Saudi Arabia between January 2015 and December 2019. Pulmonary embolism patients classified into survivor and non-survivor groups. Neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and red cell distribution width (RDW) were all recorded and were compared between the groups. The evaluation of mortality prediction, sensitivity, and specificity was carried out by employing receiver operating characteristic curves. RESULTS The variables NLR and RDW exhibited a statistically significant correlation with increased mortality and disease severity. A total of 8 patients among the 162 patients died. At the cut-off value of 5.5, NLR was showed an association with all-cause mortality, demonstrating a sensitivity of 75% and a specificity of 82%. At the cut-off value of 18.15, RDW was found to be significantly associated with all-cause mortality, displaying a sensitivity of 63% and a specificity of 88%. CONCLUSION Multiple parameters have been implicated in the mortality and severity of PE. Our study revealed a statistically significant association between NLR, RDW, and PE mortality. These tests are easily accessible and may provide insights into the mortality associated with PE.
Collapse
Affiliation(s)
- Yasir M. Alsubhi
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Ahmed H. Alhadi
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Abdulaziz M. Hammudah
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Riyadh A. Alahmadi
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Azhar M. Aljohani
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Sami Al Dubai
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Abdulqader I. Susi
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Kholoud Almuwallad
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Turki A. Alwasaidi
- From the Department of Medicine (Alsubhi, Hammudah, Alahmadi); from the Department of Critical Care (Almuwallad), King Fahad General Hospital, from the Department of Medicine (Alhadi); from the Division of Hematology (Alwasaidi), Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, from the Department of Preventive Medicine (Aljohani), Division of Infection Prevention and Control; from the Preventive Medicine Joint Program (Al Dubai), Post Graduate Studies, Ministry of Health, from the Department of Medicine (Alwasaidi), College of Medicine, Taibah University, Al-Madinah Al-Munawarah, and from the Department of Emergency Medicine (Susi), King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
30
|
Wilhelm G, Mertowska P, Mertowski S, Przysucha A, Strużyna J, Grywalska E, Torres K. The Crossroads of the Coagulation System and the Immune System: Interactions and Connections. Int J Mol Sci 2023; 24:12563. [PMID: 37628744 PMCID: PMC10454528 DOI: 10.3390/ijms241612563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
The coagulation and immune systems, two vital systems in the human body, share intimate connections that fundamentally determine patient health. These systems work together through several common regulatory pathways, including the Tissue Factor (TF) Pathway. Immune cells expressing TF and producing pro-inflammatory cytokines can influence coagulation, while coagulation factors and processes reciprocally impact immune responses by activating immune cells and controlling their functions. These shared pathways contribute to maintaining health and are also involved in various pathological conditions. Dysregulated coagulation, triggered by infection, inflammation, or tissue damage, can result in conditions such as disseminated intravascular coagulation (DIC). Concurrently, immune dysregulation may lead to coagulation disorders and thrombotic complications. This review elucidates these intricate interactions, emphasizing their roles in the pathogenesis of autoimmune diseases and cancer. Understanding the complex interplay between these systems is critical for disease management and the development of effective treatments. By exploring these common regulatory mechanisms, we can uncover innovative therapeutic strategies targeting these intricate disorders. Thus, this paper presents a comprehensive overview of the mutual interaction between the coagulation and immune systems, highlighting its significance in health maintenance and disease pathology.
Collapse
Affiliation(s)
- Grzegorz Wilhelm
- Department of Plastic and Reconstructive Surgery and Microsurgery, Medical University of Lublin, 20-059 Lublin, Poland; (G.W.); (K.T.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Anna Przysucha
- Chair and Department of Didactics and Medical Simulation, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Jerzy Strużyna
- East Center of Burns Treatment and Reconstructive Surgery, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Kamil Torres
- Department of Plastic and Reconstructive Surgery and Microsurgery, Medical University of Lublin, 20-059 Lublin, Poland; (G.W.); (K.T.)
| |
Collapse
|
31
|
Inozemtsev V, Sergunova V, Vorobjeva N, Kozlova E, Sherstyukova E, Lyapunova S, Chernysh A. Stages of NETosis Development upon Stimulation of Neutrophils with Activators of Different Types. Int J Mol Sci 2023; 24:12355. [PMID: 37569729 PMCID: PMC10418979 DOI: 10.3390/ijms241512355] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Before NETs are released, the neutrophil undergoes structural changes. First, it flattens, accompanied by a change in cell shape and rearrangement of the cytoskeleton. Then, nuclear swelling begins, which ends with the ejection of NETs into the extracellular space. We used widefield and confocal fluorescence microscopy to register morphological and structural changes in neutrophils during activation and NETosis. Different types of activators were used, such as NOX-dependent PMA and calcium ionophore A23187. The measurements were performed in a series of sequential stages. In the first stage (30 s after addition of activators and immediately after stimulation of neutrophils), the response of neutrophils to A23187 and PMA exposure was studied. Subsequently, the characteristics of neutrophils in different phases of activation were examined over a longer period of time (30, 60, 120, 180, and 240 min). The specific features of NETosis development were analyzed separately. During the first 30 s, neutrophils appeared to be heterogeneous in shape and structure of the actin cytoskeleton. Characteristic cell shapes included 30″ type 1 cells, similar in shape to the control, with F-actin concentrated in the center of the cytoplasm, and 30″ type 2 cells, which had flattened (spread) shapes with increased frontal dimensions and F-actin distributed throughout the cell. Later, the development of nuclear swelling, the corresponding changes in neutrophil membranes, and NET release into the extracellular space were evaluated. The conditions determining the initiation of chromatin ejection and two characteristic types of decondensed chromatin ejection were revealed. The results obtained contribute to a better understanding of the biophysical mechanisms of neutrophil activation and NETosis development.
Collapse
Affiliation(s)
- Vladimir Inozemtsev
- Laboratory of Biophysics of Cell Membranes under Critical State, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Scientific Research Institute of General Reanimatology, Petrovka Street 25c2, 107031 Moscow, Russia; (V.S.); (E.K.); (E.S.); (S.L.); (A.C.)
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Viktoria Sergunova
- Laboratory of Biophysics of Cell Membranes under Critical State, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Scientific Research Institute of General Reanimatology, Petrovka Street 25c2, 107031 Moscow, Russia; (V.S.); (E.K.); (E.S.); (S.L.); (A.C.)
| | - Nina Vorobjeva
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, Lenin Hills 1/12, 119234 Moscow, Russia;
| | - Elena Kozlova
- Laboratory of Biophysics of Cell Membranes under Critical State, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Scientific Research Institute of General Reanimatology, Petrovka Street 25c2, 107031 Moscow, Russia; (V.S.); (E.K.); (E.S.); (S.L.); (A.C.)
- Department of Medical and Biological Physics, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Ekaterina Sherstyukova
- Laboratory of Biophysics of Cell Membranes under Critical State, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Scientific Research Institute of General Reanimatology, Petrovka Street 25c2, 107031 Moscow, Russia; (V.S.); (E.K.); (E.S.); (S.L.); (A.C.)
| | - Snezhanna Lyapunova
- Laboratory of Biophysics of Cell Membranes under Critical State, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Scientific Research Institute of General Reanimatology, Petrovka Street 25c2, 107031 Moscow, Russia; (V.S.); (E.K.); (E.S.); (S.L.); (A.C.)
| | - Aleksandr Chernysh
- Laboratory of Biophysics of Cell Membranes under Critical State, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Scientific Research Institute of General Reanimatology, Petrovka Street 25c2, 107031 Moscow, Russia; (V.S.); (E.K.); (E.S.); (S.L.); (A.C.)
- General Pathology Department, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Scientific Research Institute of General Reanimatology, Petrovka Street 25c2, 107031 Moscow, Russia
| |
Collapse
|
32
|
Harada T, Shimomura Y, Nishida O, Maeda M, Kato Y, Nakamura T, Kuriyama N, Komura H. Effects of recombinant human soluble thrombomodulin on neutrophil extracellular traps in the kidney of a mouse model of endotoxin shock. FUJITA MEDICAL JOURNAL 2023; 9:225-230. [PMID: 37554943 PMCID: PMC10405902 DOI: 10.20407/fmj.2022-026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/12/2022] [Indexed: 08/10/2023]
Abstract
OBJECTIVES Sepsis is a life-threatening condition characterized by multi-organ dysfunction due to host immune system dysregulation in response to an infection. During sepsis, neutrophils release neutrophil extracellular traps (NETs) as part of the innate immune response. However, excessive NETs play a critical role in the development of organ failure during sepsis. Although recombinant human soluble thrombomodulin (rTM) can inhibit NET formation in the lungs and liver of a mouse model of endotoxin shock, its effects on the kidneys are unclear. METHODS The specific effects of NETs and rTM on the renal cortex and renal medulla were examined in a mouse model of endotoxin shock generated by intraperitoneal (i.p.) injection of lipopolysaccharide (LPS), followed by i.p. injection of rTM or an identical volume of saline 1 h later. RESULTS LPS injection increased serum creatinine, blood urea nitrogen, and histone H3 levels. However, rTM administration significantly decreased histone H3 and citrullinated histone H3 (citH3) levels. Immunohistochemical analysis revealed no significant changes in citH3 quantity in the renal cortex of any group. However, in the renal medulla, the increase in citH3 induced by LPS was abolished in the LPS+rTM group. CONCLUSIONS Our findings demonstrate that rTM can suppress NETs in the renal medulla of mice with endotoxin-induced acute kidney injury.
Collapse
Affiliation(s)
- Tatsuhiko Harada
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Yasuyo Shimomura
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Osamu Nishida
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Munenori Maeda
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Yu Kato
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Tomoyuki Nakamura
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Naohide Kuriyama
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| | - Hidefumi Komura
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University, School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
33
|
Hocini H, Wiedemann A, Blengio F, Lefebvre C, Cervantes-Gonzalez M, Foucat E, Tisserand P, Surenaud M, Coléon S, Prague M, Guillaumat L, Krief C, Fenwick C, Laouénan C, Bouadma L, Ghosn J, Pantaleo G, Thiébaut R, Lévy Y. Neutrophil Activation and Immune Thrombosis Profiles Persist in Convalescent COVID-19. J Clin Immunol 2023; 43:882-893. [PMID: 36943669 PMCID: PMC10029801 DOI: 10.1007/s10875-023-01459-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/24/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE Following a severe COVID-19 infection, a proportion of individuals develop prolonged symptoms. We investigated the immunological dysfunction that underlies the persistence of symptoms months after the resolution of acute COVID-19. METHODS We analyzed cytokines, cell phenotypes, SARS-CoV-2 spike-specific and neutralizing antibodies, and whole blood gene expression profiles in convalescent severe COVID-19 patients 1, 3, and 6 months following hospital discharge. RESULTS We observed persistent abnormalities until month 6 marked by (i) high serum levels of monocyte/macrophage and endothelial activation markers, chemotaxis, and hematopoietic cytokines; (ii) a high frequency of central memory CD4+ and effector CD8+ T cells; (iii) a decrease in anti-SARS-CoV-2 spike and neutralizing antibodies; and (iv) an upregulation of genes related to platelet, neutrophil activation, erythrocytes, myeloid cell differentiation, and RUNX1 signaling. We identified a "core gene signature" associated with a history of thrombotic events, with upregulation of a set of genes involved in neutrophil activation, platelet, hematopoiesis, and blood coagulation. CONCLUSION The lack of restoration of gene expression to a normal profile after up to 6 months of follow-up, even in asymptomatic patients who experienced severe COVID-19, signals the need to carefully extend their clinical follow-up and propose preventive measures.
Collapse
Affiliation(s)
- Hakim Hocini
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Aurélie Wiedemann
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Fabiola Blengio
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Cécile Lefebvre
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Minerva Cervantes-Gonzalez
- Département Épidémiologie Biostatistiques Et Recherche Clinique, AP-HP, Hôpital Bichat, INSERM, Centre d'Investigation Clinique-Epidémiologie Clinique 1425, 75018, Paris, France
- UMR 1137, Université de Paris, INSERM, IAME, 75018, Paris, France
- APHP- Hôpital Bichat - Médecine Intensive et Réanimation des Maladies Infectieuses, Paris, France
| | - Emile Foucat
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Pascaline Tisserand
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Mathieu Surenaud
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Séverin Coléon
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Mélanie Prague
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
- Department of Public Health, Univ. Bordeaux, Inserm Bordeaux Population Health Research Centre, Inria SISTM, UMR 1219, Bordeaux, France
| | - Lydia Guillaumat
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Corinne Krief
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Cédric Laouénan
- Département Épidémiologie Biostatistiques Et Recherche Clinique, AP-HP, Hôpital Bichat, INSERM, Centre d'Investigation Clinique-Epidémiologie Clinique 1425, 75018, Paris, France
- UMR 1137, Université de Paris, INSERM, IAME, 75018, Paris, France
| | - Lila Bouadma
- UMR 1137, Université de Paris, INSERM, IAME, 75018, Paris, France
- APHP- Hôpital Bichat - Médecine Intensive et Réanimation des Maladies Infectieuses, Paris, France
| | - Jade Ghosn
- UMR 1137, Université de Paris, INSERM, IAME, 75018, Paris, France
- AP-HP, Hôpital Bichat, Service de Maladies Infectieuses Et Tropicales, 75018, Paris, France
| | - Giuseppe Pantaleo
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Rodolphe Thiébaut
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
- Department of Public Health, Univ. Bordeaux, Inserm Bordeaux Population Health Research Centre, Inria SISTM, UMR 1219, Bordeaux, France
- CHU de Bordeaux, Pôle de Santé Publique, Service d'Information Médicale, Bordeaux, France
| | - Yves Lévy
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France.
- Assistance Publique-Hôpitaux de Paris, Service Immunologie Clinique, Groupe Henri-Mondor Albert-Chenevier, Créteil, France.
| |
Collapse
|
34
|
Zhou J, Zhou N, Liu Q, Xie ZP, Xu Y, Dai SC, Lu J, Bao ZY, Wu LD. Prevalence of neutropenia in US residents: a population based analysis of NHANES 2011-2018. BMC Public Health 2023; 23:1254. [PMID: 37380948 DOI: 10.1186/s12889-023-16141-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/17/2023] [Indexed: 06/30/2023] Open
Abstract
AIMS Neutrophils play a pivotal in immunity and inflammation. We aim to investigate the prevalence of neutropenia in the United States. METHODS In this cross-sectional study, participants from the National Health and Nutrition Examination Survey (NHANES) (2011-2018) were enrolled. Demographic information, hematologic measurements, smoking status of all participants were collected for all participants. All statistical analyses were performed utilizing the NHANES survey weights. Covariate-adjusted linear regression was used to compare hematologic indices in different population grouped by age, sex, ethnicity, and smoking. We also employed multivariate-logistic regression to estimate the weighted odds ratio with a 95% confidence interval and predict the neutropenia risk among. RESULTS 32,102 participants from NHANES survey were included, represented 286.6 million multiracial population in the United States. Black participants had lower mean leukocyte count (mean difference (MD): 0.71 × 109/L; P < 0.001) and lower neutrophil count (MD: 0.83 × 109/L; P < 0.001) compared with white participants after adjusting for age and sex. Furthermore, t a notable observation was the significant downward shift in the distribution curves of leukocyte count and neutrophil count among black participants. Smokers had a higher mean leukocyte count (MD: 1.10 × 109 cells/L; P < 0.001) and a higher mean neutrophil count (MD: 0.75 × 109 cells/L; P < 0.001) comparing with nonsmokers. The estimated prevalence of neutropenia was 1.24% (95% CI: 1.11 - 1.37%), which corresponds to approximately 35.5 million individuals in the United States. The prevalence of neutropenia in black participants was significantly higher than other races. Results of logistic regression analysis showed that black individuals, male individuals, and children younger than 5 years had a higher risk of neutropenia. CONCLUSIONS Neutropenia is more common in the general population than we thought, especially in black individuals and children. More attention should be paid to neutropenia.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Infectious Diseases, Affiliated Wuxi Fifth Hospital of Jiangnan University, The Fifth People's Hospital of Wuxi, Wuxi, 214065, China
| | - Nan Zhou
- Department of Nursing, Huadong Sanatorium, Wuxi, 214065, China
| | - Qing Liu
- Department of Nursing, Huadong Sanatorium, Wuxi, 214065, China
| | - Zhi-Ping Xie
- Department of Nursing, Huadong Sanatorium, Wuxi, 214065, China
| | - Yun Xu
- Department of Anesthesiology, Huadong Sanatorium, Wuxi, 214065, China
| | - Si-Cheng Dai
- Department of Nursing, Huadong Sanatorium, Wuxi, 214065, China
| | - Juan Lu
- Department of Nursing, Huadong Sanatorium, Wuxi, 214065, China.
- Department of Anesthesiology, Huadong Sanatorium, Wuxi, 214065, China.
| | - Zheng-Yang Bao
- Department of Internal Medicine, Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, China.
| | - Li-Da Wu
- Clinical Medical College of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
35
|
Zhang H, Liang R, Yuan X, Zheng Z, Lai W. Serum IgA levels for predicting the development of rapidly progressive interstitial lung disease in dermatomyositis. Respir Med 2023:107322. [PMID: 37302423 DOI: 10.1016/j.rmed.2023.107322] [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] [Received: 12/25/2022] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Some patients with dermatomyositis (DM) can develop rapidly progressive interstitial lung disease (RPILD) that is resistant to treatment and life-threatening. Convenient and practical predictive factors for the development of RPILD are currently lacking. We aimed to identify independent risk factors for RPILD in patients with DM. METHODS A total of 71 patients with DM admitted to our hospital between July 2018 and July 2022 were retrospectively reviewed. Risk factors to predict RPILD were identified by univariate and multivariate regression analyses, and significant variates for RPILD were included to establish a risk model. RESULTS Multivariate regression analysis revealed that the risk of RPILD was significantly associated with serum IgA levels. The area under the risk model curve, established by IgA levels combined with other independent predictors including the anti-melanoma differentiation-associated gene 5 (MDA5) antibody, fever, and C-reactive protein, was 0.935 (P < 0.001). CONCLUSION A higher serum IgA level was identified as an independent risk factor for RPILD in patients with DM.
Collapse
Affiliation(s)
- Haoru Zhang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Renge Liang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xin Yuan
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zemao Zheng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Weinan Lai
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
36
|
Dhanesha N, Ansari J, Pandey N, Kaur H, Virk C, Stokes KY. Poststroke venous thromboembolism and neutrophil activation: an illustrated review. Res Pract Thromb Haemost 2023; 7:100170. [PMID: 37274177 PMCID: PMC10236222 DOI: 10.1016/j.rpth.2023.100170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 06/06/2023] Open
Abstract
Patients with acute ischemic stroke are at a high risk of venous thromboembolism (VTE), such as deep vein thrombosis (DVT), estimated to affect approximately 80,000 patients with stroke each year in the United States. The prevalence of symptomatic DVT after acute stroke is approximately 10%. VTE is associated with increased rates of in-hospital death and disability, with higher prevalence of in-hospital complications and increased 1-year mortality in patients with stroke. Current guidelines recommend the use of pharmacologic VTE prophylaxis in patients with acute ischemic stroke. However, thromboprophylaxis prevents only half of expected VTE events and is associated with high risk of bleeding, suggesting the need for targeted alternative treatments to reduce VTE risk in these patients. Neutrophils are among the first cells in blood to respond after ischemic stroke. Importantly, coordinated interactions among neutrophils, platelets, and endothelial cells contribute to the development of DVT. In case of stroke and other related immune disorders, such as antiphospholipid syndrome, neutrophils potentiate thrombus propagation through the formation of neutrophil-platelet aggregates, secreting inflammatory mediators, complement activation, releasing tissue factor, and producing neutrophil extracellular traps. In this illustrated review article, we present epidemiology and management of poststroke VTE, preclinical and clinical evidence of neutrophil hyperactivation in stroke, and mechanisms for neutrophil-mediated VTE in the context of stroke. Given the hyperactivation of circulating neutrophils in patients with stroke, we propose that a better understanding of molecular mechanisms leading to neutrophil activation may result in the development of novel therapeutics to reduce the risk of VTE in this patient population.
Collapse
Affiliation(s)
- Nirav Dhanesha
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Junaid Ansari
- Department of Neurology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Nilesh Pandey
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Harpreet Kaur
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Chiranjiv Virk
- Division of Vascular Surgery and Endovascular Surgery, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Karen Y. Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| |
Collapse
|
37
|
Yamashita Y, Kobayashi T, Mo M. Thrombosis and Anticoagulation Strategies in Patients with COVID-19 Including Japanese Perspective. J Atheroscler Thromb 2023; 30:311-320. [PMID: 36792179 PMCID: PMC10067339 DOI: 10.5551/jat.rv22002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has become a major health problem worldwide since 2020. Although the main pathophysiology of COVID-19 is a respiratory infectious disease, it could also cause cardiovascular complications, including thrombosis. Thus, anticoagulation therapy has been thought to help prevent thrombosis, leading to improved survival. However, to date, several aspects of the optimal anticoagulation strategies for COVID-19 remain unclear. Considering the status of COVID-19-related thrombosis and some domestic issues in Japan, the optimal anticoagulation strategies for COVID-19 might have to be based on Japanese domestic clinical data considering racial difference. Racial disparities in terms of thromboembolic risk have been well known in the pre-COVID-19 era, and the risk of COVID-19-associated thrombosis depending on race could be an important issue. Considering a potential higher risk of bleeding with anticoagulation therapy in the Asian population, it might be important to maintain a good balance between the risks of thrombosis and bleeding. Latest evidences of COVID-19-related thrombosis and anticoagulation strategies, including some domestic issues in Japan, showed a different status of COVID-19-related thrombosis in Japan from that in Western countries, suggesting the potential benefit of different anticoagulation strategies, specifically for the Japanese population. Although these insights could be useful for the consideration of anticoagulation strategies for the Japanese population, the final decision should be based on balancing the benefits and risks of anticoagulation therapy in each patient.
Collapse
Affiliation(s)
- Yugo Yamashita
- Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Japan
| | | | - Makoto Mo
- Department of Cardiovascular Surgery, Yokohama Minami Kyosai Hospital, Yokohama, Japan
| |
Collapse
|
38
|
Yu M, Xiao G, Han L, Peng L, Wang H, He S, Lyu M, Zhu Y. QiShen YiQi and its components attenuate acute thromboembolic stroke and carotid thrombosis by inhibition of CD62P/PSGL-1-mediated platelet-leukocyte aggregate formation. Biomed Pharmacother 2023; 160:114323. [PMID: 36738500 DOI: 10.1016/j.biopha.2023.114323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND QiShen YiQi (QSYQ) dropping pill, a component-based Chinese medicine consisting of benefiting Qi (YQ) and activating blood (HX) components, has been reported to exert a beneficial effect on cerebral ischemia-induced stroke. However, its efficacy and pharmacological mechanism on acute thromboembolic stroke is not clear. PURPOSE This study is to explore the preventative effect and pharmacological mechanism of QSYQ and its YQ/HX components on the formation of platelet-leukocyte aggregation (PLA) in acute thromboembolic stroke. STUDY DESIGN AND METHODS In vivo thromboembolic stroke model and FeCl3-induced carotid arterial occlusion models were used. Immunohistochemistry, Western blot, RT-qPCR, and flow cytometry experiments were performed to reveal the pharmacological mechanisms of QSYQ and its YQ/HX components. RESULTS In thromboembolic stroke rats, QSYQ significantly attenuated infarct area, improved neurological recovery, reduced PLA formation, and inhibited P-selection (CD62P)/ P-selectin glycoprotein ligand-1 (PSGL-1) expressions. The YQ component preferentially down-regulated PSGL-1 expression in leukocyte, while the HX component preferentially down-regulated CD62P expression in platelet. In carotid arterial thrombosis mice, QSYQ and its YQ/HX components inhibited thrombus formation, prolonged vessel occlusion time, reduced circulating leukocytes and P-selectin expression. PLA formation and platelet/leukocyte adhesion to endothelial cell were also inhibited by QSYQ and its YQ/HX components in vitro. CONCLUSION QSYQ and YQ/HX components attenuated thromboembolic stroke and carotid thrombosis by decreasing PLA formation via inhibiting CD62P/PSGL-1 expressions. This study shed a new light on the prevention of thromboembolic stroke.
Collapse
Affiliation(s)
- Mingxing Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Linhong Han
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Li Peng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Huanyi Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Ming Lyu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China.
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China.
| |
Collapse
|
39
|
The Role of Neutrophils in Lower Limb Peripheral Artery Disease: State of the Art and Future Perspectives. Int J Mol Sci 2023; 24:ijms24021169. [PMID: 36674682 PMCID: PMC9866688 DOI: 10.3390/ijms24021169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
In recent years, increasing attention has been paid to the role of neutrophils in cardiovascular (CV) disease (CVD) with evidence supporting their role in the initiation, progression, and rupture of atherosclerotic plaque. Although these cells have long been considered as terminally differentiated cells with a relatively limited spectrum of action, recent research has revealed intriguing novel cellular functions, including neutrophil extracellular trap (NET) generation and inflammasome activation, which have been linked to several human diseases, including CVD. While most research to date has focused on the role of neutrophils in coronary artery and cerebrovascular diseases, much less information is available on lower limb peripheral artery disease (PAD). PAD is a widespread condition associated with great morbidity and mortality, though physician and patient awareness of the disease remains low. To date, several studies have produced some evidence on the role of certain biomarkers of neutrophil activation in this clinical setting. However, the etiopathogenetic role of neutrophils, and in particular of some of the newly discovered mechanisms, has yet to be fully elucidated. In the future, complementary assessment of neutrophil activity should improve CV risk stratification and provide personalized treatments to patients with PAD. This review aims to summarize the basic principles and recent advances in the understanding of neutrophil biology, current knowledge about the role of neutrophils in atherosclerosis, as well as available evidence on their role of PAD.
Collapse
|
40
|
Pang X, He X, Qiu Z, Zhang H, Xie R, Liu Z, Gu Y, Zhao N, Xiang Q, Cui Y. Targeting integrin pathways: mechanisms and advances in therapy. Signal Transduct Target Ther 2023; 8:1. [PMID: 36588107 PMCID: PMC9805914 DOI: 10.1038/s41392-022-01259-6] [Citation(s) in RCA: 252] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 01/03/2023] Open
Abstract
Integrins are considered the main cell-adhesion transmembrane receptors that play multifaceted roles as extracellular matrix (ECM)-cytoskeletal linkers and transducers in biochemical and mechanical signals between cells and their environment in a wide range of states in health and diseases. Integrin functions are dependable on a delicate balance between active and inactive status via multiple mechanisms, including protein-protein interactions, conformational changes, and trafficking. Due to their exposure on the cell surface and sensitivity to the molecular blockade, integrins have been investigated as pharmacological targets for nearly 40 years, but given the complexity of integrins and sometimes opposite characteristics, targeting integrin therapeutics has been a challenge. To date, only seven drugs targeting integrins have been successfully marketed, including abciximab, eptifibatide, tirofiban, natalizumab, vedolizumab, lifitegrast, and carotegrast. Currently, there are approximately 90 kinds of integrin-based therapeutic drugs or imaging agents in clinical studies, including small molecules, antibodies, synthetic mimic peptides, antibody-drug conjugates (ADCs), chimeric antigen receptor (CAR) T-cell therapy, imaging agents, etc. A serious lesson from past integrin drug discovery and research efforts is that successes rely on both a deep understanding of integrin-regulatory mechanisms and unmet clinical needs. Herein, we provide a systematic and complete review of all integrin family members and integrin-mediated downstream signal transduction to highlight ongoing efforts to develop new therapies/diagnoses from bench to clinic. In addition, we further discuss the trend of drug development, how to improve the success rate of clinical trials targeting integrin therapies, and the key points for clinical research, basic research, and translational research.
Collapse
Affiliation(s)
- Xiaocong Pang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Xu He
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiwei Qiu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Hanxu Zhang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Ran Xie
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiyan Liu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Yanlun Gu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Nan Zhao
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Qian Xiang
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| |
Collapse
|
41
|
Blanch-Ruíz MA, Sánchez-López A, Ríos-Navarro C, Ortega-Luna R, Collado-Díaz V, Orden S, Martínez-Cuesta MA, Esplugues JV, Álvarez Á. Abacavir causes leukocyte/platelet crosstalk by activating neutrophil P2X7 receptors thus releasing soluble lectin-like oxidized low-density lipoprotein receptor-1. Br J Pharmacol 2022; 180:1516-1532. [PMID: 36541109 DOI: 10.1111/bph.16016] [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: 03/11/2022] [Revised: 11/02/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Abacavir, an antiretroviral drug used in HIV therapy associated with myocardial infarction, promotes thrombosis through P2X7 receptors. The role of platelets as pro-thrombotic cells is acknowledged whereas that of neutrophils-due to their secretory capacity-is gaining recognition. This study analyses the role of neutrophils-specifically the secretome of abacavir-treated neutrophils (SNABC )-in platelet activation that precedes thrombosis. EXPERIMENTAL APPROACH Effects of abacavir or SNABC on platelet activation and platelet-leukocyte interactions and expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) were analysed by flow cytometry. The secretome was analysed by proteomics. The role of leukocytes in the actions of abacavir was evaluated in a mouse model of thrombosis. KEY RESULTS Abacavir induced platelet-leukocyte interactions, not directly via effects of abacavir on platelets, but via activation of neutrophils, which triggered interactions between platelet P-selectin and neutrophil P-selectin glycoprotein ligand-1 (PSGL-1). SNABC stimulated platelet activation and platelet-leukocyte interactions through a process that was dependent on LOX-1, neutrophil P2X7 and platelet P2Y1, P2Y12 and P2X1 receptors. Abacavir induced the expression of LOX-1 on neutrophils and of the soluble form of LOX-1 (sLOX-1) in SNABC . Neutrophils, LOX-1, P2X7, P2Y1, P2Y12 and P2X1 receptors were required for the pro-thrombotic actions of abacavir in vivo. CONCLUSION AND IMPLICATIONS Neutrophils are target cells in abacavir-induced thrombosis. Abacavir released sLOX-1 from neutrophils via activation of their P2X7 receptors, which in turn activated platelets. Hence, sLOX-1 could be the missing link in the cardiovascular risk associated with abacavir.
Collapse
Affiliation(s)
| | - Ainhoa Sánchez-López
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - César Ríos-Navarro
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Víctor Collado-Díaz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Samuel Orden
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.,FISABIO-Fundación Hospital Universitario Dr. Peset, Valencia, Spain
| | - María Angeles Martínez-Cuesta
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.,CIBERehd, Valencia, Spain
| | - Juan V Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.,FISABIO-Fundación Hospital Universitario Dr. Peset, Valencia, Spain.,CIBERehd, Valencia, Spain
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.,CIBERehd, Valencia, Spain
| |
Collapse
|
42
|
Colicchia M, Schrottmaier WC, Perrella G, Reyat JS, Begum J, Slater A, Price J, Clark JC, Zhi Z, Simpson MJ, Bourne JH, Poulter NS, Khan AO, Nicolson PLR, Pugh M, Harrison P, Iqbal AJ, Rainger GE, Watson SP, Thomas MR, Mutch NJ, Assinger A, Rayes J. S100A8/A9 drives the formation of procoagulant platelets through GPIbα. Blood 2022; 140:2626-2643. [PMID: 36026606 PMCID: PMC10653093 DOI: 10.1182/blood.2021014966] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022] Open
Abstract
S100A8/A9, also known as "calprotectin" or "MRP8/14," is an alarmin primarily secreted by activated myeloid cells with antimicrobial, proinflammatory, and prothrombotic properties. Increased plasma levels of S100A8/A9 in thrombo-inflammatory diseases are associated with thrombotic complications. We assessed the presence of S100A8/A9 in the plasma and lung autopsies from patients with COVID-19 and investigated the molecular mechanism by which S100A8/A9 affects platelet function and thrombosis. S100A8/A9 plasma levels were increased in patients with COVID-19 and sustained high levels during hospitalization correlated with poor outcomes. Heterodimeric S100A8/A9 was mainly detected in neutrophils and deposited on the vessel wall in COVID-19 lung autopsies. Immobilization of S100A8/A9 with collagen accelerated the formation of a fibrin-rich network after perfusion of recalcified blood at venous shear. In vitro, platelets adhered and partially spread on S100A8/A9, leading to the formation of distinct populations of either P-selectin or phosphatidylserine (PS)-positive platelets. By using washed platelets, soluble S100A8/A9 induced PS exposure but failed to induce platelet aggregation, despite GPIIb/IIIa activation and alpha-granule secretion. We identified GPIbα as the receptor for S100A8/A9 on platelets inducing the formation of procoagulant platelets with a supporting role for CD36. The effect of S100A8/A9 on platelets was abolished by recombinant GPIbα ectodomain, platelets from a patient with Bernard-Soulier syndrome with GPIb-IX-V deficiency, and platelets from mice deficient in the extracellular domain of GPIbα. We identified the S100A8/A9-GPIbα axis as a novel targetable prothrombotic pathway inducing procoagulant platelets and fibrin formation, in particular in diseases associated with high levels of S100A8/A9, such as COVID-19.
Collapse
Affiliation(s)
- Martina Colicchia
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Jasmeet S. Reyat
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jenefa Begum
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Joshua Price
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Joanne C. Clark
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Zhaogong Zhi
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Megan J. Simpson
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Joshua H. Bourne
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Natalie S. Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Abdullah O. Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Phillip L. R. Nicolson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Matthew Pugh
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Paul Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Asif J. Iqbal
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - George E. Rainger
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Mark R. Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Nicola J. Mutch
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Alice Assinger
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| |
Collapse
|
43
|
Qian W, Cao Y. An overview of the effects and mechanisms of m6 A methylation on innate immune cells in sepsis. Front Immunol 2022; 13:1041990. [PMID: 36505499 PMCID: PMC9731128 DOI: 10.3389/fimmu.2022.1041990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction Sepsis is a severe clinical syndrome caused by dysregulated systemic inflammatory responses to infection. Methylation modification, as a crucial mechanism of RNA functional modification, can manipulate the immunophenotype and functional activity of immune cells to participate in sepsis progression. This study aims to explore the mechanism of N6-methyladenosine (m6A) methylation modification in immune cell-mediated sepsis through keyword search. Methods Literature retrieval. Results and Discussion Literature retrieval reveals that m6A methylation is implicated in sepsis-induced lung injury and myocardial injury,as well as sepsis-related encephalopathy. Furthermore, it is found that m6A methylation can regulate sepsis by inhibiting the chemotaxis of neutrophils and the formation of neutrophil extracellular traps and suppressing macrophage phagocytosis, thereby playing a role in sepsis.
Collapse
Affiliation(s)
- Weiwei Qian
- Emergency Department, Shangjinnanfu Hospital, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Cao
- Emergency Department of West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Yu Cao,
| |
Collapse
|
44
|
Liu Y, Yan P, Bin Y, Qin X, Wu Z. Neutrophil extracellular traps and complications of liver transplantation. Front Immunol 2022; 13:1054753. [PMID: 36466888 PMCID: PMC9712194 DOI: 10.3389/fimmu.2022.1054753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/26/2022] [Indexed: 08/29/2023] Open
Abstract
Many end-stage liver disease etiologies are attributed to robust inflammatory cell recruitment. Neutrophils play an important role in inflammatory infiltration and neutrophil phagocytosis, oxidative burst, and degranulation. It has also been suggested that neutrophils may release neutrophil extracellular traps (NETs) to kill pathogens. It has been proven that neutrophil infiltration within the liver contributes to an inflammatory microenvironment and immune cell activation. Growing evidence implies that NETs are involved in the progression of numerous complications of liver transplantation, including ischemia-reperfusion injury, acute rejection, thrombosis, and hepatocellular carcinoma recurrence. NETs are discussed in this comprehensive review, focusing on their effects on liver transplantation complications. Furthermore, we discuss NETs as potential targets for liver transplantation therapy.
Collapse
Affiliation(s)
- Yanyao Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Yan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Bin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan Qin
- Department of General Surgery and Trauma Surgery, Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
45
|
Cha MJ, Ha J, Lee H, Kwon I, Kim S, Kim YD, Nam HS, Lee HS, Song TJ, Choi HJ, Heo JH. Neutrophil Recruitment in Arterial Thrombus and Characteristics of Stroke Patients with Neutrophil-Rich Thrombus. Yonsei Med J 2022; 63:1016-1026. [PMID: 36303310 PMCID: PMC9629897 DOI: 10.3349/ymj.2022.0328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Neutrophils contribute to thrombosis. However, there is limited information on the temporal course of neutrophil recruitment in thrombosis, the contribution of neutrophils to thrombus growth, and the characteristics of stroke patients with neutrophil-rich thrombi. MATERIALS AND METHODS After inducing carotid artery thrombosis in Institute of Cancer Research mice using ferric chloride, aged thrombi were produced by ligating the distal portion of the carotid artery in mice for 0.5, 1, 2, 3, 6, or 24 h. For thrombus analysis in stroke patients, we used registry data and thrombi that were obtained during intra-arterial thrombectomy. Immunohistochemistry was performed to determine thrombus composition. RESULTS In the thrombi of 70 mice, Ly6G positive cell counts (neutrophils) and histone H3-positive cell counts increased in a time-dependent manner (both p<0.001). Ly6G-positive cell count was strongly correlated with histone H3-positive cell counts (r=0.910, p<0.001), but not with thrombus size (p=0.320). In 75 stroke patients, atrial fibrillation and cardioembolism were more frequent in the higher neutrophil group (32/37, 86.5%) than in the lower neutrophil group (19/38, 50%) (p=0.002). The median erythrocyte fraction was higher [52.0 (interquartile range 39.9-57.8)] in the higher neutrophil group than in the lower neutrophil group [40.3 (interquartile range 23.5-53.2)]. The fraction of neutrophils was positively correlated with that of erythrocytes (R=0.35, p=0.002). CONCLUSION Neutrophils were recruited and increased in arterial thrombosis in a time-dependent manner; however, they were not associated with the growth of formed thrombi. Neutrophil fractions in the thrombi of stroke patients appeared to be associated with atrial fibrillation and erythrocyte fraction.
Collapse
Affiliation(s)
- Myoung-Jin Cha
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, National Police Hospital, Seoul, Korea
| | - Jimin Ha
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Hyungwoo Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Il Kwon
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Sungeun Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Young Dae Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo Suk Nam
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Tae-Jin Song
- Department of Neurology, Ewha Womans University School of Medicine, Seoul, Korea
| | - Hyun-Jung Choi
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hoe Heo
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
46
|
Immunothrombosis and the Role of Platelets in Venous Thromboembolic Diseases. Int J Mol Sci 2022; 23:ijms232113176. [PMID: 36361963 PMCID: PMC9656618 DOI: 10.3390/ijms232113176] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 12/05/2022] Open
Abstract
Venous thromboembolism (VTE) is the third leading cardiovascular cause of death and is conventionally treated with anticoagulants that directly antagonize coagulation. However, recent data have demonstrated that also platelets play a crucial role in VTE pathophysiology. In the current review, we outline how platelets are involved during all stages of experimental venous thrombosis. Platelets mediate initiation of the disease by attaching to the vessel wall upon which they mediate leukocyte recruitment. This process is referred to as immunothrombosis, and within this novel concept inflammatory cells such as leukocytes and platelets directly drive the progression of VTE. In addition to their involvement in immunothrombosis, activated platelets can directly drive venous thrombosis by supporting coagulation and secreting procoagulant factors. Furthermore, fibrinolysis and vessel resolution are (partly) mediated by platelets. Finally, we summarize how conventional antiplatelet therapy can prevent experimental venous thrombosis and impacts (recurrent) VTE in humans.
Collapse
|
47
|
Xu X, Wu Y, Xu S, Yin Y, Ageno W, De Stefano V, Zhao Q, Qi X. Clinical significance of neutrophil extracellular traps biomarkers in thrombosis. Thromb J 2022; 20:63. [PMID: 36224604 PMCID: PMC9555260 DOI: 10.1186/s12959-022-00421-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/25/2022] [Indexed: 11/10/2022] Open
Abstract
Neutrophil extracellular traps (NETs) may be associated with the development of thrombosis. Experimental studies have confirmed the presence of NETs in thrombi specimens and potential role of NETs in the mechanisms of thrombosis. Clinical studies also have demonstrated significant changes in the levels of serum or plasma NETs biomarkers, such as citrullinated histones, myeloperoxidase, neutrophil elastase, nucleosomes, DNA, and their complexes in patients with thrombosis. This paper aims to comprehensively review the currently available evidence regarding the change in the levels of NETs biomarkers in patients with thrombosis, summarize the role of NETs and its biomarkers in the development and prognostic assessment of venous thromboembolism, coronary artery diseases, ischemic stroke, cancer-associated thromboembolism, and coronavirus disease 2019-associated thromboembolism, explore the potential therapeutic implications of NETs, and further discuss the shortcomings of existing NETs biomarkers in serum and plasma and their detection methods.
Collapse
Affiliation(s)
- Xiangbo Xu
- Department of Gastroenterology, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China.,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China.,Department of Pharmacy, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
| | - Yuting Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China.,Department of Pharmacy, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
| | - Shixue Xu
- Department of Gastroenterology, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
| | - Yue Yin
- Department of Gastroenterology, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
| | - Walter Ageno
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Valerio De Stefano
- Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli IRCCS, Section of Hematology, Rome, Italy
| | - Qingchun Zhao
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China. .,Department of Pharmacy, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China.
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China. .,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
48
|
Neutrophil extracellular traps accelerate vascular smooth muscle cell proliferation via Akt/CDKN1b/TK1 accompanying with the occurrence of hypertension. J Hypertens 2022; 40:2045-2057. [PMID: 35950975 PMCID: PMC9451946 DOI: 10.1097/hjh.0000000000003231] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) can trigger pathological changes in vascular cells or vessel wall components, which are vascular pathological changes of hypertension. Therefore, we hypothesized that NETs would be associated with the occurrence of hypertension. METHODS To evaluate the relationship between NETs and hypertension, we evaluated both the NETs formation in spontaneously hypertensive rats (SHRs) and the blood pressure of mice injected phorbol-12-myristate-13-acetate (PMA) via the tail vein to induce NETs formation in arterial wall. Meanwhile, proliferation and cell cycle of vascular smooth muscle cells (VSMCs), which were co-cultured with NETs were assessed. In addition, the role of exosomes from VSMCs co-cultured with NETs on proliferation signaling delivery was assessed. RESULTS Formation of NETs increased in the arteries of SHR. PMA resulted in up-regulation expression of citrullinated Histone H3 (cit Histone H3, a NETs marker) in the arteries of mice accompanied with increasing of blood pressure. NET treatment significantly increased VSMCs count and accelerated G1/S transition in vitro . Cyclin-dependent kinase inhibitor 1b (CDKN1b) was down-regulated and Thymidine kinase 1 (TK1) was up-regulated in VSMCs. Exosomes from VSMCs co-cultured with NETs significantly accelerated the proliferation of VSMCs. TK1 was up-regulated in the exosomes from VSMCs co-cultured with NETs and in both the arterial wall and serum of mice with PMA. CONCLUSION NETs promote VSMCs proliferation via Akt/CDKN1b/TK1 and is related to hypertension development. Exosomes from VSMCs co-cultured with NETs participate in transferring the proliferation signal. These results support the role of NETs in the development of hypertension.
Collapse
|
49
|
Zhang H, Lao Q, Zhang J, Zhu J. Coagulopathy in COVID-19 and anticoagulation clinical trials. Best Pract Res Clin Haematol 2022; 35:101377. [PMID: 36494146 PMCID: PMC9395291 DOI: 10.1016/j.beha.2022.101377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/02/2022] [Accepted: 08/16/2022] [Indexed: 12/15/2022]
Abstract
Severe acute respiratory disease coronavirus 2 (SARS-COV-2) first emerged in Wuhan, China, in December 2019 and has caused a global pandemic of a scale unprecedented in the modern era. People infected with SARS-CoV-2 can be asymptomatic, moderate symptomatic or develop severe COVID-19. Other than the typical acute respiratory distress syndrome (ARDS), patients with moderate or severe COVID-19 also develop a distinctive systemic coagulopathy, known as COVID-19-associated coagulopathy (CAC), which is different from sepsis-related forms of disseminated intravascular coagulation (DIC). Endotheliopathy or endotheliitis are other unique features of CAC. The endothelial cell perturbation can further increase the risk of thrombotic events in COVID-19 patients. In this review, we will summarize the current knowledge on COVID-19 coagulopathy and the possible mechanisms for the condition. We also discuss the results of clinical trials testing methods for mitigating thrombosis events in COVID-19 patients.
Collapse
Affiliation(s)
- Heng Zhang
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA
| | - Qifang Lao
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA; Department of Critical Care Medicine, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Jue Zhang
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA
| | - Jieqing Zhu
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
50
|
Nayak L, Sweet DR, Thomas A, Lapping SD, Kalikasingh K, Madera A, Vinayachandran V, Padmanabhan R, Vasudevan NT, Myers JT, Huang AY, Schmaier A, Mackman N, Liao X, Maiseyeu A, Jain MK. A targetable pathway in neutrophils mitigates both arterial and venous thrombosis. Sci Transl Med 2022; 14:eabj7465. [PMID: 36044595 DOI: 10.1126/scitranslmed.abj7465] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Arterial and venous thrombosis constitutes a major source of morbidity and mortality worldwide. Long considered as distinct entities, accumulating evidence indicates that arterial and venous thrombosis can occur in the same populations, suggesting that common mechanisms are likely operative. Although hyperactivation of the immune system is a common forerunner to the genesis of thrombotic events in both vascular systems, the key molecular control points remain poorly understood. Consequently, antithrombotic therapies targeting the immune system for therapeutics gain are lacking. Here, we show that neutrophils are key effectors of both arterial and venous thrombosis and can be targeted through immunoregulatory nanoparticles. Using antiphospholipid antibody syndrome (APS) as a model for arterial and venous thrombosis, we identified the transcription factor Krüppel-like factor 2 (KLF2) as a key regulator of neutrophil activation. Upon activation through genetic loss of KLF2 or administration of antiphospholipid antibodies, neutrophils clustered P-selectin glycoprotein ligand 1 (PSGL-1) by cortical actin remodeling, thereby increasing adhesion potential at sites of thrombosis. Targeting clustered PSGL-1 using nanoparticles attenuated neutrophil-mediated thrombosis in APS and KLF2 knockout models, illustrating the importance and feasibility of targeting activated neutrophils to prevent pathological thrombosis. Together, our results demonstrate a role for activated neutrophils in both arterial and venous thrombosis and identify key molecular events that serve as potential targets for therapeutics against diverse causes of immunothrombosis.
Collapse
Affiliation(s)
- Lalitha Nayak
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - David R Sweet
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.,Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Asha Thomas
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Stephanie D Lapping
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Kenneth Kalikasingh
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Annmarie Madera
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Vinesh Vinayachandran
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Roshan Padmanabhan
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Neelakantan T Vasudevan
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Jay T Myers
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alex Y Huang
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alvin Schmaier
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Nigel Mackman
- Division of Hematology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xudong Liao
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Andrei Maiseyeu
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mukesh K Jain
- Warren Alpert Medical School of Brown University, Providence, R1 02903
| |
Collapse
|