1
|
Lu X, Chen Q, Wang L, Zhu H, Huang L, Zhou J, Guo Y. Association between platelet to lymphocyte ratio and febrile urinary tract infection after double-J stent removal in children underwent laparoscopic pyeloplasty. BMC Urol 2025; 25:122. [PMID: 40361034 PMCID: PMC12070569 DOI: 10.1186/s12894-025-01808-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE To evaluate the association between the inflammatory biomarkers and the prevalence of febrile urinary tract infection (fUTI) after double-J (DJ) stent removal in pediatrics following laparoscopic pyeloplasty (LP). METHODS A retrospective study was conducted in pediatrics underwent DJ stent removal following LP owing to primary ureteropelvic junction obstruction (UPJO) between September 2021 and November 2024. Baseline characteristics, preoperative data and the incidence of fUTI were documented. The inflammatory index including neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR) and systemic immune-inflammation index (SII) were calculated. The results of cultured pathogens were also identified. The univariate and multivariate logistic analysis were conducted to determine the potential risk factors of fUTI after DJ stent removal. The predictive value of potential risk factors were determined by receiver operating characteristic curve (ROC). RESULTS Overall, 295 patients were included in the study. fUTI occurred in 22 patients (7.5%) after DJ stent removal. Patients in the fUTI group were younger (P = 0.008) and had lower body weight (P = 0.003) compared to non-fUTI group. Additionally, the fUTI group showed higher levels of platelets and neutrophils, associated with lower levels of lymphocytes. The most commonly identified pathogens were Enterococcus and Escherichia coli in fUTI patients. Multivariate logistic analysis revealed that age (OR = 0.978, 95% CI: 0.956-0.999, P = 0.047), toilet training status (OR = 0.297, 95% CI: 0.109-0.807, P = 0.017) and higher levels of PLR (OR = 1.101, 95% CI: 1.005-1.022, P = 0.002) were predictive factors for fUTI after DJ stent removal. PLR had a high predictive value with an AUC of 0.827 with the sensitivity of 90.91% and the specificity of 69.23%. CONCLUSION PLR is a promising predictor for diagnosing fUTI after DJ stent removal. Patients with higher levels of PLR before DJ stent removal should be closely monitored. Further well-designed and prospective cohorts are required in future to explore the cause-and-effect relationship between PLR and fUTI after DJ removal.
Collapse
Affiliation(s)
- Xun Lu
- Department of Urology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lixia Wang
- Department of Urology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Haobo Zhu
- Department of Urology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Liqu Huang
- Department of Urology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jincai Zhou
- Department of Urology, Affiliated Jianhu Hospital of Xinglin College, Nantong University, Nantong, China.
| | - Yunfei Guo
- Department of Urology, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Kaiser R, Gold C, Stark K. Recent Advances in Immunothrombosis and Thromboinflammation. Thromb Haemost 2025. [PMID: 40311639 DOI: 10.1055/a-2523-1821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Inflammation and thrombosis are traditionally considered two separate entities of acute host responses to barrier breaks. While inciting inflammatory responses is a prerequisite to fighting invading pathogens and subsequent restoration of tissue homeostasis, thrombus formation is a crucial step of the hemostatic response to prevent blood loss following vascular injury. Though originally designed to protect the host, excessive induction of either inflammatory signaling or thrombus formation and their reciprocal activation contribute to a plethora of disorders, including cardiovascular, autoimmune, and malignant diseases. In this state-of-the-art review, we summarize recent insights into the intricate interplay of inflammation and thrombosis. We focus on the protective aspects of immunothrombosis as well as evidence of detrimental sequelae of thromboinflammation, specifically regarding recent studies that elucidate its pathophysiology beyond coronavirus disease 2019 (COVID-19). We introduce recently identified molecular aspects of key cellular players like neutrophils, monocytes, and platelets that contribute to both immunothrombosis and thromboinflammation. Further, we describe the underlying mechanisms of activation involving circulating plasma proteins and immune complexes. We then illustrate how these factors skew the inflammatory state toward detrimental thromboinflammation across cardiovascular as well as septic and autoimmune inflammatory diseases. Finally, we discuss how the advent of new technologies and the integration with clinical data have been used to investigate the mechanisms and signaling cascades underlying immunothrombosis and thromboinflammation. This review highlights open questions that will need to be addressed by the field to translate our mechanistic understanding into clinically meaningful therapeutic targeting.
Collapse
Affiliation(s)
- Rainer Kaiser
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christoph Gold
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
3
|
Chen X, Shi T, Chen F, Xie X, Fang H, Wu Z, Liu Y, Huang Y, Wang Q, Nie G, Xu J, Shao D. Orally Antigen-Engineered Yeast Vaccine Elicits Robust Intestinal Mucosal Immunity. ACS NANO 2025; 19:10841-10853. [PMID: 40082064 DOI: 10.1021/acsnano.4c14690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Mucosal immunity plays a pivotal role in safeguarding against significant global infectious diseases caused by mucosal pathogens. The development of mucosal vaccines has been limited by the poor efficiency of antigen display and the risk of adjuvants. Here, we report an engineered yeast vaccine integrating a well-displayed antigen with an intrinsic adjuvant for the development of innate and adaptive immunity to the intestinal mucosa. Compared with antigen-secretory yeast, antigen-anchored yeast significantly activated gut dendritic cells (DCs) and promoted follicular helper T (Tfh) cell differentiation, thereby amplifying the immune response by the interaction with Tfh-B cells. Consequently, oral vaccination of SARS-CoV-2 receptor-binding domain (RBD)-anchored yeast triggered stronger RBD-specific IgA-neutralizing effects, providing potential adaptive protections. Given its corresponding impact on the functionality of both innate and adaptive mucosal responses, the proposed RBD-anchored yeast outperformed RBD-anchored bacteria and biomimetic nanovaccine in the production of RBD-specific IgA and IgG. Together, these results revealed how antigen-displaying patterns could be modulated to elicit intestinal mucosal immunity and demonstrated the translational potential of antigen-displayed yeast for effective mucosal protection.
Collapse
Affiliation(s)
- Xuenian Chen
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P.R. China
| | - Tongfei Shi
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P.R. China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P.R. China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, P.R. China
| | - Xiaochun Xie
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P.R. China
| | - Hui Fang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P.R. China
| | - Ziping Wu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P.R. China
| | - Yang Liu
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 101408, P.R. China
| | - Yubiao Huang
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 101408, P.R. China
| | - Qin Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Guangjun Nie
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 101408, P.R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Jiaqi Xu
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 101408, P.R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Dan Shao
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P.R. China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P.R. China
| |
Collapse
|
4
|
De Chiara S, De Simone Carone L, Cirella R, Andretta E, Silipo A, Molinaro A, Mercogliano M, Di Lorenzo F. Beyond the Toll-Like Receptor 4. Structure-Dependent Lipopolysaccharide Recognition Systems: How far are we? ChemMedChem 2025; 20:e202400780. [PMID: 39752323 PMCID: PMC11911305 DOI: 10.1002/cmdc.202400780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
With an enormous potential in immunology and vaccinology, lipopolysaccharides (LPSs) are among the most extensively studied bacteria-derived molecules. LPS centered studies are countless, and their results reverberate in all areas of the life sciences, including chemistry, biology, genetics, biophysics, and medicine. Most of these research activities are focused on the LPS-induced immune response activation by means of Myeloid Differentiation protein-2/Toll Like Receptor 4 (MD-2/TLR4) complex, which currently is the most largely explored LPS sensing pathway. However, the enormous structural variability of LPS allows interactions with numerous other receptors involved in a wide range of equally important immunological scenarios. In this review, we explore these additional LPS recognition systems, which operate within interconnected signaling cascades, highlighting their role in maintaining physiological homeostasis and their involvement in the development of severe human diseases. Understanding these pathways, their interconnections, and the crosstalk between them and TLR4/MD-2 is essential for guiding the development of pharmacologically active molecules that could specifically modulate the inflammatory response, paving the way to new strategies for combating immune-mediated diseases and resistant infections.
Collapse
Affiliation(s)
- Stefania De Chiara
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Luca De Simone Carone
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Roberta Cirella
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Emanuela Andretta
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Alba Silipo
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
- CEINGE, Istituto di Biotecnologie avanzateVia Gaetano Salvatore, 48680131NaplesItaly
| | - Antonio Molinaro
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
- CEINGE, Istituto di Biotecnologie avanzateVia Gaetano Salvatore, 48680131NaplesItaly
- Department of ChemistrySchool of ScienceOsaka University1-1 Osaka University MachikaneyamaToyonakaOsaka560-0043Japan
| | - Marcello Mercogliano
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Flaviana Di Lorenzo
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
- CEINGE, Istituto di Biotecnologie avanzateVia Gaetano Salvatore, 48680131NaplesItaly
| |
Collapse
|
5
|
Tokarz-Deptuła B, Baraniecki Ł, Palma J, Stosik M, Deptuła W. Characterization of Platelet Receptors and Their Involvement in Immune Activation of These Cells. Int J Mol Sci 2024; 25:12611. [PMID: 39684330 DOI: 10.3390/ijms252312611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The article characterises platelets, pointing out the role and contribution of their numerous receptors determining their specific and broad immune activity. Three types of platelet receptors are described, that is, extracellular and intracellular receptors-TLR (toll-like receptors), NLR (NOD-like receptor), and RLR (RIG-I-like receptor); extracellular receptors-selectins and integrins; and their other extracellular receptors-CLR (C-type lectin receptor), CD (cluster of differentiation), TNF (tumour necrosis factor), among others. Outlining the contribution of these numerous platelet receptors to the intravascular immunity, it has been shown that they are formed by their fusion with pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), and lifestyle-associated molecular patterns (LAMPs). They are initiating and effector components of signal transduction of these cells, and their expression and quantity determine the specific and broad functions of platelets towards influencing vascular endothelial cells, but mainly PRRs (pattern recognition receptors) of blood immune cells. These facts make platelets the fundamental elements that shape not only intravascular homeostasis, as previously indicated, but they become the determinants of immunity in blood vessels. Describing the reactions of the characterised three groups of platelet receptors with PAMP, DAMP and LAMP molecules, the pathways and participation of platelets in the formation and construction of intravascular immune status, in physiological states, but mainly in pathological states, including bacterial and viral infections, are presented, making these cells essential elements in the health and disease of mammals, including humans.
Collapse
Affiliation(s)
| | - Łukasz Baraniecki
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
- Doctoral School, University of Szczecin, 70-384 Szczecin, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Michał Stosik
- Institute of Biological Science, Faculty of Biological Sciences, University of Zielona Góra, 65-516 Zielona Góra, Poland
| | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
| |
Collapse
|
6
|
Yan Y, Xu N, Wang X, Shi L, Huang Q, Wang J, Li X, Ni T, Yang Z, Guo W. Mesoporous polydopamine/copper sulfide hybrid nanocomposite for highly efficient NIR-triggered bacterial inactivation. Int J Biol Macromol 2024; 277:134238. [PMID: 39084434 DOI: 10.1016/j.ijbiomac.2024.134238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/08/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Polydopamine has gained considerable attention in the biomaterial domain owing to its excellent biocompatibility, antioxidant activity, photothermal effect and adhesion property. Herein, copper sulfide (Cu2-xS) wrapped in mesoporous polydopamine (MPDA) was synthesized through in-situ polymerization, followed by the surface modification with cationic polyethyleneimine (PEI). The mussel-inspired MPDA matrix successfully prevented the oxidation and agglomeration of Cu2-xS nanoparticles, and regulated the release of copper ions and reactive oxygen species (ROS) levels. Surface-modified PEI endow MPDA@Cu2-xS with positive charges, facilitating their rapid contact with negatively charged bacteria through electrostatic interactions. The pH-dependent Cu+/Cu2+ release and NIR-responsive ROS generation were confirmed using molecular probes and electron spin resonance (ESR). The MPDA@Cu2-xS/PEI showed significantly enhanced antibacterial activity and reduced cytotoxicity for NIH3T3 cells. Under NIR irradiation (1.0 W/cm2, 10 min), germicidal efficiency against Escherichia coli (E. coli) and Staphyloccocus aureus (S. aureus) could reach 100 % and 99.94 %, respectively. The exceptional antibacterial activities of MPDA@Cu2-xS/PEI was mainly attributed to the synergistic photothermal effect, controlled release of copper ions and ROS generation, as well as electrostatic interaction. More importantly, the MPDA@Cu2-xS/PEI composite exhibited excellent biocompatibility and biosafety. Overall, this organic/inorganic hybrid holds great potential as a promising candidate for wound treatment.
Collapse
Affiliation(s)
- Yunhui Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China.
| | - Na Xu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Xian Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Li Shi
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Qianqian Huang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Jia Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Xiangrong Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Tianjun Ni
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhijun Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Wei Guo
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
7
|
Vymazal O, Papatheodorou I, Andrejčinová I, Bosáková V, Vascelli G, Bendíčková K, Zelante T, Hortová-Kohoutková M, Frič J. Calcineurin-NFAT signaling controls neutrophils' ability of chemoattraction upon fungal infection. J Leukoc Biol 2024; 116:816-829. [PMID: 38648505 DOI: 10.1093/jleuko/qiae091] [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: 12/01/2023] [Revised: 03/03/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Calcineurin-nuclear factor of activated T cells (CN-NFAT) inhibitors are widely clinically used drugs for immunosuppression, but besides their required T cell response inhibition, they also undesirably affect innate immune cells. Disruption of innate immune cell function can explain the observed susceptibility of CN-NFAT inhibitor-treated patients to opportunistic fungal infections. Neutrophils play an essential role in innate immunity as a defense against pathogens; however, the effect of CN-NFAT inhibitors on neutrophil function was poorly described. Thus, we tested the response of human neutrophils to opportunistic fungal pathogens, namely Candida albicans and Aspergillus fumigatus, in the presence of CN-NFAT inhibitors. Here, we report that the NFAT pathway members were expressed in neutrophils and mediated part of the neutrophil response to pathogens. Upon pathogen exposure, neutrophils underwent profound transcriptomic changes with subsequent production of effector molecules. Importantly, genes and proteins involved in the regulation of the immune response and chemotaxis, including the chemokines CCL2, CCL3, and CCL4 were significantly upregulated. The presence of CN-NFAT inhibitors attenuated the expression of these chemokines and impaired the ability of neutrophils to chemoattract other immune cells. Our results amend knowledge about the impact of CN-NFAT inhibition in human neutrophils.
Collapse
Affiliation(s)
- Ondrej Vymazal
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Ioanna Papatheodorou
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Ivana Andrejčinová
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Veronika Bosáková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Gianluca Vascelli
- Section of Immunology and General Pathology, Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi 1/8, Perugia, 06132, Italy
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Teresa Zelante
- Section of Immunology and General Pathology, Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi 1/8, Perugia, 06132, Italy
| | - Marcela Hortová-Kohoutková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
- Institute of Hematology and Blood Transfusion, U Nemocnice 2094/1, Prague 2, 128 00, Czech Republic
| |
Collapse
|
8
|
Shi R, Bai C, Sun S, Wang F, Li C, Wang C, Hu L, Zhao Z, Guo Q, Du G, Xu D, Chen AF, Yang W. Identification of ferroptosis-related key genes associated with immune infiltration in sepsis by bioinformatics analysis and in vivo validation. Gene 2024; 918:148482. [PMID: 38649061 DOI: 10.1016/j.gene.2024.148482] [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: 02/07/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVES Sepsis is a life-threatening infectious disease in which an immune inflammatory response is triggered. The potential effect of ferroptosis-related genes (FRGs) in inflammation of sepsis remained unclear. We focused on identifying and validating core FRGs and their association with immune infiltration in blood from currently all patients with sepsis. METHODS All current raw data of septic blood were obtained from Gene Expression Omnibus. After removing the batch effect merging into a complete dataset and obtaining Diferentially expressed genes (DEGs). Common cross-talk genes were identified from DEGs and FRGs. WGCNA, GO, KEGG, PPI, GESA, ROC curves, and LASSO regression analysis were performed to indentify and validate key genes based on external septic datasets. Infiltrated immune cells in 2 hub genes (MAPK14 and ACSL4) were conducted using CIBERSORT algorithm and Spearman correlation analysis. Further, the expressions of 2 core FRGs were verified in the LPS-induced ALI and cardiac injury sepsis mice. RESULTS MAPK14 and ACSL4 were identified, mostly enriched in T cell infiltration through NOD-like receptor signaling pathway according to the high or low 2 hub genes expression. The upregulated 2 ferroptosis-related genes were validated in LPS-induced ALI and cardiac injury mice, accompanied by upregulation of the NLRP3 pathway. CONCLUSION MAPK14 and ACSL4 could become robustly reliable and promising biomarkers for sepsis by regulating ferroptosis through the NLRP3 pathway, which is mainly associated with T-cell infiltration.
Collapse
Affiliation(s)
- Rui Shi
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China.
| | - Chunyun Bai
- Yunnan Institute for Food and Drug Control, Kunming, China.
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China.
| | - Fang Wang
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Chaozhong Li
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Chongyu Wang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China.
| | - Lidan Hu
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Ziwen Zhao
- Department of Cardiac Surgery, Yunnan Fuwai Cardiovascular Hospital, Kunming Medical University, Kunming, China.
| | - Qiuzhe Guo
- Department of Cardiac Surgery, Yunnan Fuwai Cardiovascular Hospital, Kunming Medical University, Kunming, China.
| | - Guanhua Du
- Institute of Materia Medica Chinese Academy of Medical Sciences, Beijing, China.
| | - Dan Xu
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Weimin Yang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China.
| |
Collapse
|
9
|
Hawwari I, Rossnagel L, Rosero N, Maasewerd S, Vasconcelos MB, Jentzsch M, Demczuk A, Teichmann LL, Meffert L, Bertheloot D, Ribeiro LS, Kallabis S, Meissner F, Arditi M, Atici AE, Noval Rivas M, Franklin BS. Platelet transcription factors license the pro-inflammatory cytokine response of human monocytes. EMBO Mol Med 2024; 16:1901-1929. [PMID: 38977927 PMCID: PMC11319489 DOI: 10.1038/s44321-024-00093-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
In humans, blood Classical CD14+ monocytes contribute to host defense by secreting large amounts of pro-inflammatory cytokines. Their aberrant activity causes hyper-inflammation and life-threatening cytokine storms, while dysfunctional monocytes are associated with 'immunoparalysis', a state of immune hypo responsiveness and reduced pro-inflammatory gene expression, predisposing individuals to opportunistic infections. Understanding how monocyte functions are regulated is critical to prevent these harmful outcomes. We reveal platelets' vital role in the pro-inflammatory cytokine responses of human monocytes. Naturally low platelet counts in patients with immune thrombocytopenia or removal of platelets from healthy monocytes result in monocyte immunoparalysis, marked by impaired cytokine response to immune challenge and weakened host defense transcriptional programs. Remarkably, supplementing monocytes with fresh platelets reverses these conditions. We discovered that platelets serve as reservoirs of key cytokine transcription regulators, such as NF-κB and MAPK p38, and pinpointed the enrichment of platelet NF-κB2 in human monocytes by proteomics. Platelets proportionally restore impaired cytokine production in human monocytes lacking MAPK p38α, NF-κB p65, and NF-κB2. We uncovered a vesicle-mediated platelet-monocyte-propagation of inflammatory transcription regulators, positioning platelets as central checkpoints in monocyte inflammation.
Collapse
Affiliation(s)
- Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
| | - Lukas Rossnagel
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Marius Jentzsch
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Agnieszka Demczuk
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lino L Teichmann
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Lisa Meffert
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lucas S Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sebastian Kallabis
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Felix Meissner
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asli E Atici
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
10
|
Jiang K, Hwa J, Xiang Y. Novel strategies for targeting neutrophil against myocardial infarction. Pharmacol Res 2024; 205:107256. [PMID: 38866263 DOI: 10.1016/j.phrs.2024.107256] [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: 05/09/2024] [Revised: 06/08/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Inflammation is a crucial factor in cardiac remodeling after acute myocardial infarction (MI). Neutrophils, as the first wave of leukocytes to infiltrate the injured myocardium, exacerbate inflammation and cardiac injury. However, therapies that deplete neutrophils to manage cardiac remodeling after MI have not consistently produced promising outcomes. Recent studies have revealed that neutrophils at different time points and locations may have distinct functions. Thus, transferring neutrophil phenotypes, rather than simply blocking their activities, potentially meet the needs of cardiac repair. In this review, we focus on discussing the fate, heterogeneity, functions of neutrophils, and attempt to provide a more comprehensive understanding of their roles and targeting strategies in MI. We highlight the strategies and translational potential of targeting neutrophils to limit cardiac injury to reduce morbidity and mortality from MI.
Collapse
Affiliation(s)
- Kai Jiang
- State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Yaozu Xiang
- State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
11
|
Du CH, Wu YD, Yang K, Liao WN, Ran L, Liu CN, Zhang SZ, Yu K, Chen J, Quan Y, Chen M, Shen MQ, Tang H, Chen SL, Wang S, Zhao JH, Cheng TM, Wang JP. Apoptosis-resistant megakaryocytes produce large and hyperreactive platelets in response to radiation injury. Mil Med Res 2023; 10:66. [PMID: 38111039 PMCID: PMC10729570 DOI: 10.1186/s40779-023-00499-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND The essential roles of platelets in thrombosis have been well recognized. Unexpectedly, thrombosis is prevalent during thrombocytopenia induced by cytotoxicity of biological, physical and chemical origins, which could be suffered by military personnel and civilians during chemical, biological, radioactive, and nuclear events. Especially, thrombosis is considered a major cause of mortality from radiation injury-induced thrombocytopenia, while the underlying pathogenic mechanism remains elusive. METHODS A mouse model of radiation injury-induced thrombocytopenia was built by exposing mice to a sublethal dose of ionizing radiation (IR). The phenotypic and functional changes of platelets and megakaryocytes (MKs) were determined by a comprehensive set of in vitro and in vivo assays, including flow cytometry, flow chamber, histopathology, Western blotting, and chromatin immunoprecipitation, in combination with transcriptomic analysis. The molecular mechanism was investigated both in vitro and in vivo, and was consolidated using MK-specific knockout mice. The translational potential was evaluated using a human MK cell line and several pharmacological inhibitors. RESULTS In contrast to primitive MKs, mature MKs (mMKs) are intrinsically programmed to be apoptosis-resistant through reprogramming the Bcl-xL-BAX/BAK axis. Interestingly, mMKs undergo minority mitochondrial outer membrane permeabilization (MOMP) post IR, resulting in the activation of the cyclic GMP-AMP synthase-stimulator of IFN genes (cGAS-STING) pathway via the release of mitochondrial DNA. The subsequent interferon-β (IFN-β) response in mMKs upregulates a GTPase guanylate-binding protein 2 (GBP2) to produce large and hyperreactive platelets that favor thrombosis. Further, we unmask that autophagy restrains minority MOMP in mMKs post IR. CONCLUSIONS Our study identifies that megakaryocytic mitochondria-cGAS/STING-IFN-β-GBP2 axis serves as a fundamental checkpoint that instructs the size and function of platelets upon radiation injury and can be harnessed to treat platelet pathologies.
Collapse
Affiliation(s)
- Chang-Hong Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| | - Yi-Ding Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
- Frontier Medical Training Brigade, Army Medical University, Xinjiang, 831200, China
| | - Ke Yang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Wei-Nian Liao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Li Ran
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Chao-Nan Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Shu-Zhen Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Kuan Yu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Jun Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Yong Quan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Mo Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Ming-Qiang Shen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Hong Tang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Shi-Lei Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Song Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Jing-Hong Zhao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Tian-Min Cheng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Jun-Ping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
12
|
Poli V, Di Gioia M, Zanoni I. Quantitative cytofluorimetric analysis of mouse neutrophil extracellular traps. STAR Protoc 2023; 4:102372. [PMID: 37352106 PMCID: PMC10331305 DOI: 10.1016/j.xpro.2023.102372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/21/2023] [Indexed: 06/25/2023] Open
Abstract
The release of neutrophil extracellular traps (NETs) has been involved in numerous infectious and non-infectious diseases. Nevertheless, quantitative analysis of NETs in vivo has been challenging. Here, we present a protocol for NET quantification by flow cytometry in the bronchoalveolar lavage fluid (BALF) of mice upon pulmonary infection with S. aureus. We describe steps for bacteria growth and instillation and BALF recovery. We then detail staining to quantify the release of NETs and neutrophils recruited to the site of infection. For complete information on the generation and use of this protocol, please refer to Poli et al. (2021)1 and Poli et al. (2022).2.
Collapse
Affiliation(s)
- Valentina Poli
- Harvard Medical School, Boston, MA 02115, USA; Boston Children's Hospital, Division of Immunology, Boston, MA 02115, USA
| | - Marco Di Gioia
- Harvard Medical School, Boston, MA 02115, USA; Boston Children's Hospital, Division of Immunology, Boston, MA 02115, USA
| | - Ivan Zanoni
- Harvard Medical School, Boston, MA 02115, USA; Boston Children's Hospital, Division of Immunology, Boston, MA 02115, USA; Boston Children's Hospital, Division of Gastroenterology, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Gullotta GS, De Feo D, Friebel E, Semerano A, Scotti GM, Bergamaschi A, Butti E, Brambilla E, Genchi A, Capotondo A, Gallizioli M, Coviello S, Piccoli M, Vigo T, Della Valle P, Ronchi P, Comi G, D'Angelo A, Maugeri N, Roveri L, Uccelli A, Becher B, Martino G, Bacigaluppi M. Age-induced alterations of granulopoiesis generate atypical neutrophils that aggravate stroke pathology. Nat Immunol 2023; 24:925-940. [PMID: 37188941 DOI: 10.1038/s41590-023-01505-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023]
Abstract
Aging accounts for increased risk and dismal outcome of ischemic stroke. Here, we investigated the impact of age-related changes in the immune system on stroke. Upon experimental stroke, compared with young mice, aged mice had increased neutrophil clogging of the ischemic brain microcirculation, leading to worse no-reflow and outcomes. Aged mice showed an enhanced granulopoietic response to stroke that led to the accumulation of CD101+CD62Llo mature and CD177hiCD101loCD62Llo and CD177loCD101loCD62Lhi immature atypical neutrophils in the blood, endowed with increased oxidative stress, phagocytosis and procoagulant features. Production of CXCL3 by CD62Llo neutrophils of the aged had a key role in the development and pathogenicity of aging-associated neutrophils. Hematopoietic stem cell rejuvenation reverted aging-associated neutropoiesis and improved stroke outcome. In elderly patients with ischemic stroke, single-cell proteome profile of blood leukocytes identified CD62Llo neutrophil subsets associated with worse reperfusion and outcome. Our results unveil how stroke in aging leads to a dysregulated emergency granulopoiesis impacting neurological outcome.
Collapse
Affiliation(s)
- Giorgia Serena Gullotta
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Ekaterina Friebel
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Aurora Semerano
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
- Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Andrea Bergamaschi
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Erica Butti
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Brambilla
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Angela Genchi
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
- Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Alessia Capotondo
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Mattia Gallizioli
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | | | - Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS, Policlinico San Donato, Milan, Italy
| | - Tiziana Vigo
- IRCCS, Ospedale Policlinico San Martino, Genova, Italy
| | - Patrizia Della Valle
- Coagulation Service and Thrombosis Research Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Paola Ronchi
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, Telethon Institute for Gene Therapy (HSR-TIGET), IRCCS San Raffaele Hospital, Milan, Italy
| | - Giancarlo Comi
- Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Armando D'Angelo
- Coagulation Service and Thrombosis Research Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Norma Maugeri
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Luisa Roveri
- Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Antonio Uccelli
- IRCCS, Ospedale Policlinico San Martino, Genova, Italy
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genoa, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Gianvito Martino
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
- Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Marco Bacigaluppi
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy.
- Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy.
| |
Collapse
|
14
|
Rolling CC, Barrett TJ, Berger JS. Platelet-monocyte aggregates: molecular mediators of thromboinflammation. Front Cardiovasc Med 2023; 10:960398. [PMID: 37255704 PMCID: PMC10225702 DOI: 10.3389/fcvm.2023.960398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023] Open
Abstract
Platelets, key facilitators of primary hemostasis and thrombosis, have emerged as crucial cellular mediators of innate immunity and inflammation. Exemplified by their ability to alter the phenotype and function of monocytes, activated platelets bind to circulating monocytes to form monocyte-platelet aggregates (MPA). The platelet-monocyte axis has emerged as a key mechanism connecting thrombosis and inflammation. MPA are elevated across the spectrum of inflammatory and autoimmune disorders, including cardiovascular disease, systemic lupus erythematosus (SLE), and COVID-19, and are positively associated with disease severity. These clinical disorders are all characterized by an increased risk of thromboembolic complications. Intriguingly, monocytes in contact with platelets become proinflammatory and procoagulant, highlighting that this interaction is a central element of thromboinflammation.
Collapse
Affiliation(s)
- Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
15
|
Yan C, Wu H, Fang X, He J, Zhu F. Platelet, a key regulator of innate and adaptive immunity. Front Med (Lausanne) 2023; 10:1074878. [PMID: 36968817 PMCID: PMC10038213 DOI: 10.3389/fmed.2023.1074878] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/14/2023] [Indexed: 03/12/2023] Open
Abstract
Platelets, anucleate blood components, represent the major cell type involved in the regulation of hemostasis and thrombosis. In addition to performing haemostatic roles, platelets can influence both innate and adaptive immune responses. In this review, we summarize the development of platelets and their functions in hemostasis. We also discuss the interactions between platelet products and innate or adaptive immune cells, including neutrophils, monocytes, macrophages, T cells, B cells and dendritic cells. Activated platelets and released molecules regulate the differentiation and function of these cells via platelet-derived receptors or secreting molecules. Platelets have dual effects on nearly all immune cells. Understanding the exact mechanisms underlying these effects will enable further application of platelet transfusion.
Collapse
Affiliation(s)
- Cheng Yan
- Department of Blood Transfusion, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haojie Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xianchun Fang
- Department of Blood Transfusion, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junji He
- Department of Blood Transfusion, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Zhu
- Department of Blood Transfusion, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- *Correspondence: Feng Zhu,
| |
Collapse
|
16
|
Poli V, Zanoni I. Neutrophil intrinsic and extrinsic regulation of NETosis in health and disease. Trends Microbiol 2023; 31:280-293. [PMID: 36344311 PMCID: PMC9974585 DOI: 10.1016/j.tim.2022.10.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
Neutrophil extracellular traps (NETs) evolved to protect the host against microbial infections and are formed by a web-like structure of DNA that is decorated with antimicrobial effectors. Due to their potent inflammatory functions, NETs also cause tissue damage and can favor and/or aggravate inflammatory diseases. This multipronged activity of NETs requires that the induction, release, and degradation of NETs are tightly regulated. Here we describe the key pathways that are intrinsic to neutrophils and regulate NETosis, and we review the most recent findings on how neutrophil extrinsic factors participate in the formation of NETs. In particular, we emphasize how bystander cells contribute to modifying the capacity of neutrophils to undergo NETosis. Finally, we discuss how these neutrophil extrinsic processes can be harnessed to protect the host against the excessive inflammation elicited by uncontrolled NET release.
Collapse
Affiliation(s)
- Valentina Poli
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Division of Gastroenterology, Boston, MA, USA
| | - Ivan Zanoni
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Division of Gastroenterology, Boston, MA, USA.
| |
Collapse
|
17
|
Macrophage NFATC2 mediates angiogenic signaling during mycobacterial infection. Cell Rep 2022; 41:111817. [PMID: 36516756 PMCID: PMC9880963 DOI: 10.1016/j.celrep.2022.111817] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
During mycobacterial infections, pathogenic mycobacteria manipulate both host immune and stromal cells to establish and maintain a productive infection. In humans, non-human primates, and zebrafish models of infection, pathogenic mycobacteria produce and modify the specialized lipid trehalose 6,6'-dimycolate (TDM) in the bacterial cell envelope to drive host angiogenesis toward the site of forming granulomas, leading to enhanced bacterial growth. Here, we use the zebrafish-Mycobacterium marinum infection model to define the signaling basis of the host angiogenic response. Through intravital imaging and cell-restricted peptide-mediated inhibition, we identify macrophage-specific activation of NFAT signaling as essential to TDM-mediated angiogenesis in vivo. Exposure of cultured human cells to Mycobacterium tuberculosis results in robust induction of VEGFA, which is dependent on a signaling pathway downstream of host TDM detection and culminates in NFATC2 activation. As granuloma-associated angiogenesis is known to serve bacterial-beneficial roles, these findings identify potential host targets to improve tuberculosis disease outcomes.
Collapse
|
18
|
Zhang F, Li Y, Wu J, Zhang J, Cao P, Sun Z, Wang W. The role of extracellular traps in ischemia reperfusion injury. Front Immunol 2022; 13:1022380. [PMID: 36211432 PMCID: PMC9533173 DOI: 10.3389/fimmu.2022.1022380] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
In response to strong signals, several types of immune cells release extracellular traps (ETs), which are web-like structures consisting of DNA decorated with various protein substances. This process is most commonly observed in neutrophils. Over the past two decades, ET formation has been recognized as a unique mechanism of host defense and pathogen destruction. However, the role of ETs in sterile inflammation has only been studied extensively in recent years. Ischemia reperfusion injury (IRI) is a type of sterile inflammatory injury. Several studies have reported that ETs have an important role in IRI in various organs. In this review, we describe the release of ETs by various types of immune cells and focus on the mechanism underlying the formation of neutrophil ETs (NETs). In addition, we summarize the role of ETs in IRI in different organs and their effects on tumors. Finally, we discuss the value of ETs as a potential therapeutic target for organ IRI and present possible challenges in conducting studies on IRI-related ETs as well as future research directions and prospects.
Collapse
Affiliation(s)
- Feilong Zhang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Yuqing Li
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Peng Cao
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
- *Correspondence: Wei Wang,
| |
Collapse
|
19
|
Immunothrombosis and the molecular control of tissue factor by pyroptosis: prospects for new anticoagulants. Biochem J 2022; 479:731-750. [PMID: 35344028 DOI: 10.1042/bcj20210522] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023]
Abstract
The interplay between innate immunity and coagulation after infection or injury, termed immunothrombosis, is the primary cause of disseminated intravascular coagulation (DIC), a condition that occurs in sepsis. Thrombosis associated with DIC is the leading cause of death worldwide. Interest in immunothrombosis has grown because of COVID-19, the respiratory disease caused by SARS-CoV-2, which has been termed a syndrome of dysregulated immunothrombosis. As the relatively new field of immunothrombosis expands at a rapid pace, the focus of academic and pharmacological research has shifted from generating treatments targeted at the traditional 'waterfall' model of coagulation to therapies better directed towards immune components that drive coagulopathies. Immunothrombosis can be initiated in macrophages by cleavage of the non-canonical inflammasome which contains caspase-11. This leads to release of tissue factor (TF), a membrane glycoprotein receptor that forms a high-affinity complex with coagulation factor VII/VIIa to proteolytically activate factors IX to IXa and X to Xa, generating thrombin and leading to fibrin formation and platelet activation. The mechanism involves the post-translational activation of TF, termed decryption, and release of decrypted TF via caspase-11-mediated pyroptosis. During aberrant immunothrombosis, decryption of TF leads to thromboinflammation, sepsis, and DIC. Therefore, developing therapies to target pyroptosis have emerged as an attractive concept to counteract dysregulated immunothrombosis. In this review, we detail the three mechanisms of TF control: concurrent induction of TF, caspase-11, and NLRP3 (signal 1); TF decryption, which increases its procoagulant activity (signal 2); and accelerated release of TF into the intravascular space via pyroptosis (signal 3). In this way, decryption of TF is analogous to the two signals of NLRP3 inflammasome activation, whereby induction of pro-IL-1β and NLRP3 (signal 1) is followed by activation of NLRP3 (signal 2). We describe in detail TF decryption, which involves pathogen-induced alterations in the composition of the plasma membrane and modification of key cysteines on TF, particularly at the location of the critical, allosterically regulated disulfide bond of TF in its 219-residue extracellular domain. In addition, we speculate towards the importance of identifying new therapeutics to block immunothrombotic triggering of TF, which can involve inhibition of pyroptosis to limit TF release, or the direct targeting of TF decryption using cysteine-modifying therapeutics.
Collapse
|
20
|
|