1
|
Wu S, Hu Y, Tao Z, Yu Y, Zhu P, Li T, Jin Y, Wang Y, Qian H, Wang H, Ma Y. Comprehensive Management of Ulcerative Colitis and its Associated Intra-Extra Intestinal Complications with a Multifunctional Inulin Hydrogel Complex. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500726. [PMID: 40263912 DOI: 10.1002/smll.202500726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Excessive accumulation of reactive oxygen species (ROS) and dysbiosis of gut microbiota are pivotal etiological factors in the pathogenesis of ulcerative colitis (UC) and its associated intestinal and extraintestinal manifestations (e.g., intestinal microthrombosis, anxiety, and depression symptoms). This investigation presents a multifunctional inulin complex (PB/NKase@Inulin gel) incorporating Prussian blue nanozymes (PB NZs) and the thrombolytic agent nattokinase (NKase) for the therapeutic management of dextran sulfate sodium (DSS)-induced UC and its associated intestinal and extraintestinal complications. Following oral administration, the PB/NKase@Inulin gel, characterized by prolonged retention of PB NZs and NKase at inflamed colonic sites, can facilitate continuously ROS scavenging, attenuate oxidative stress damage, effectively reduce pro-inflammatory cytokine levels. Importantly, PB/NKase@Inulin gel can not only robustly inhibit inflammatory microthrombosis formation but also effectively lyses thrombi due to the potent thrombolytic properties of NKase both in vitro and in vivo. Furthermore, the PB/NKase@Inulin gel is able to modulate gut microbiota homeostasis and alleviate multiple stresses responses (including anxiety and depression) in a UC mouse model via microbiota-gut-brain (MGB) axis interactions. Overall, the PB/NKase@Inulin gel offers an innovative paradigm for comprehensive therapeutic interventions in DSS-induced UC and its multifaceted complications.
Collapse
Affiliation(s)
- Silong Wu
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Yaoyu Hu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Zhenchao Tao
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, China
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, Hefei, 230031, China
| | - Yi Yu
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Pengfei Zhu
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Tao Li
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Yu Jin
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Yujie Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
2
|
Li H, Chen S, Li Y, Sang Z, Chen Z, Mei X, Ren X. Preparation of gastrointestinal pH-responsive zein coated tea polyphenol-heparin hydrocolloids using for inflammatory bowel disease therapy. Int J Biol Macromol 2025; 309:143135. [PMID: 40233898 DOI: 10.1016/j.ijbiomac.2025.143135] [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: 12/17/2024] [Revised: 03/09/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025]
Abstract
In the pathogenesis of inflammatory bowel disease (IBD), overstimulation of inflammatory factors can trigger a coagulation cascade, increase the risk of intestinal micro-thrombosis and lead to microcirculation disorders. However, prevention of microcirculation pathways has not received enough attention. Heparin is commonly used in anticoagulant therapy, but oral delivery does not have an excellent anticoagulant effect. To improve the stability of heparin (HEP) in the gastrointestinal tract, zein/tea polyphenol nanospheres with a core-shell structure (EGNs@Z) were developed for oral administration of heparin (HEGNs@Z). The Zein shell has pH-responsive properties and is effective in preventing premature dissolution of heparin. At the same time, EGCG nanospheres (EGNs) play an anti-inflammatory role, jointly improve the vicious cycle between inflammation and microthrombosis. The results of SEM, TEM and FTIR showed that EGNs successfully encapsulated heparin and formed zein shells on the surface of microspheres with a thickness of 50-100 nm. In vitro simulated digestion experiments showed that zein shells prevented the breakdown of microspheres and heparin in a simulated gastric environment, whereas EGNs and HEP were slowly degraded and released in a simulated intestinal environment. Coagulation analysis showed that HEGNs@Z was effective in delaying clotting time. A mouse model of acute colitis has also shown that HEGNs@Z robust promotes colonic epithelial regeneration, inhibits malignant microcirculation, and reduces bleeding risk. These findings reveal that this orally bioavailable multifunctional material may provide a novel, effective and convenient treatment for inflammatory bowel disease.
Collapse
Affiliation(s)
- Hui Li
- Pharmaceutical Sciences School, Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Shuangshuang Chen
- Pharmaceutical Sciences School, Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yumei Li
- Pharmaceutical Sciences School, Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zelin Sang
- Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zhenhua Chen
- Pharmaceutical Sciences School, Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, China.
| | - Xifan Mei
- Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, China; The Third Affiliated Hospital of Jinzhou Medical University, 121000 Jinzhou, China.
| | - Xiuli Ren
- Pharmaceutical Sciences School, Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou Medical University, Jinzhou, Liaoning, China.
| |
Collapse
|
3
|
Lai Y, Zhu Y, Zhang X, Ding S, Wang F, Hao J, Wang Z, Shi C, Xu Y, Zheng L, Huang W. Gut microbiota-derived metabolites: Potential targets for cardiorenal syndrome. Pharmacol Res 2025; 214:107672. [PMID: 40010448 DOI: 10.1016/j.phrs.2025.107672] [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: 11/06/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
The characteristic of cardiorenal syndrome (CRS) is simultaneous damage to both the heart and kidneys. CRS has caused a heavy burden of mortality and incidence rates worldwide. The regulation of host microbiota metabolism that triggers heart and kidney damage is an emerging research field that promotes a new perspective on cardiovascular risk. We summarize current studies from bench to bedside of gut microbiota-derived metabolites to better understand CRS in the context of gut microbiota-derived metabolites. We focused on the involvement of gut microbiota-derived metabolites in the pathophysiology of CRS, including lipid and cholesterol metabolism disorders, coagulation abnormalities and platelet aggregation, oxidative stress, endothelial dysfunction, inflammation, mitochondrial damage and energy metabolism disorders, vascular calcification and renal fibrosis, as well as emerging therapeutic approaches targeting CRS metabolism in gut microbiota-derived metabolites which provides an innovative treatment approach for CRS to improve patient prognosis and overall quality of life.
Collapse
Affiliation(s)
- Yuchen Lai
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yujie Zhu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Xihui Zhang
- Department of Blood Purification, General Hospital of Central Theater Command(Hankou Campus), No.68, Huangpu Avenue, Wuhan, 430010, China
| | - Shifang Ding
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, China
| | - Fang Wang
- Department of Blood Purification, General Hospital of Central Theater Command(Hankou Campus), No.68, Huangpu Avenue, Wuhan, 430010, China
| | - Jincen Hao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Zhaomeng Wang
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Institute of Brain Disorders, The Capital Medical University, Beijing 100050, China
| | - Congqi Shi
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yongjin Xu
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China; Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Institute of Brain Disorders, The Capital Medical University, Beijing 100050, China.
| | - Wei Huang
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, China.
| |
Collapse
|
4
|
Zhou C, Liang C, Zhang R, Wang Y, Luo S, Pan J. TREM2 improves coagulopathy and lung inflammation in sepsis through the AKT-mTOR pathway. Int Immunopharmacol 2025; 150:114330. [PMID: 39970715 DOI: 10.1016/j.intimp.2025.114330] [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: 11/15/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Sepsis is a systemic inflammatory response syndrome triggered by infection, often accompanied by severe coagulopathy, leading to high mortality. Tissue factor (TF) plays a pivotal role in sepsis by promoting both coagulation and inflammation. Recently, TREM2 (Triggering Receptor Expressed on Myeloid cells 2) has emerged as a key regulator of macrophage function, but its specific role in sepsis remains unclear. METHODS An in vitro sepsis model was established by stimulating RAW264.7 cells with 10 μg/mL lipopolysaccharide (LPS) for 6 h, with four groups: Negative Control (NC), NC + LPS, TREM2, and TREM2 + LPS. Inflammatory cytokines and coagulation factors were measured in each group. Cells in the TREM2 and TREM2 + LPS groups were pretreated with TREM2 overexpression plasmid for 48 h. In vivo, mice were assigned to Sham, TREM2, Cecal Ligation and Puncture (CLP), CLP + NC, and CLP + TREM2 groups. Mice in the NC group received macrophages via tail vein injection, while those in the TREM2 and CLP + TREM2 groups received TREM2-overexpressing macrophages. Lung tissue and plasma samples were collected to assess inflammatory cytokines, coagulation factors, and signaling pathway activity. RESULTS TREM2 overexpression significantly improved survival, reduced lung inflammation, and alleviated coagulopathy in mice. It increased platelet counts and reduced fibrin deposition. Furthermore, TREM2 inhibited TF release from macrophages by suppressing aberrant activation of the AKT-mTOR signaling pathway, thereby modulating the macrophage inflammatory response. CONCLUSIONS TREM2 plays a crucial protective role in sepsis-associated coagulopathy, suggesting that it could serve as a potential therapeutic target, providing novel strategies to improve clinical outcomes in sepsis patients.
Collapse
Affiliation(s)
- Chen Zhou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Wenzhou Medical University, Wenzhou 325000, China
| | - Chenglong Liang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Wenzhou Medical University, Wenzhou 325000, China
| | - Rongrong Zhang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315300, China
| | - Ying Wang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315300, China
| | - Shuang Luo
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jingye Pan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Wenzhou Medical University, Wenzhou 325000, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315300, China; Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, 325000, China; Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, 325000, China.
| |
Collapse
|
5
|
Bonisoli GL, Argentino G, Friso S, Tinazzi E. Extracellular Vesicles Analysis as Possible Signatures of Antiphospholipid Syndrome Clinical Features. Int J Mol Sci 2025; 26:2834. [PMID: 40243411 PMCID: PMC11989148 DOI: 10.3390/ijms26072834] [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: 02/03/2025] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 04/18/2025] Open
Abstract
Antiphospholipid syndrome (APS) is a rare autoimmune disease characterized by thrombosis and obstetric complications. Extracellular vesicles (EVs) of either platelet and endothelial origin are recognized to be involved in the pathophysiology of the disease. This study aimed to evaluate the potential role of endothelial- and platelet-derived extracellular vesicles and the clinical features or progression of APS. We enrolled 22 patients diagnosed with APS and 18 age and sex-matched healthy controls. We determined APS-specific antibody positivity and clinical manifestations in APS affected patients, with a focus on neurological, cardiovascular, dermatological, hematological manifestations, and pregnancy-related complications. Platelet-poor plasma was collected from either patients and controls for the analysis of EVs by flow cytometry technology using monoclonal antibodies to specifically identify those derived from either platelets and/or endothelial cells. EVs of endothelial and platelet origins were overall significantly increased in patients as compared to healthy controls. Furthermore, a significant association was also observed between the number of extracellular vesicles and specific organ involvement, particularly central nervous system manifestations, hematological abnormalities, and obstetric complications. An elevated proportion of endothelial-derived EVs in APS and a reduction of resting endothelial cell-derived EVs were observed in APS-affected women with obstetric complications. Our findings highlight the involvement of endothelial cells and platelets in mirroring the activities of endothelial cells and platelets in APS. Additionally, extracellular vesicles may serve as potential predictors of organ involvement and disease-related damage.
Collapse
Affiliation(s)
| | | | | | - Elisa Tinazzi
- Department of Medicine, University of Verona, 37134 Verona, Italy
| |
Collapse
|
6
|
Xia X, Tie X, Hong M, Yin W. Exploration of the causal relationship and mechanisms between serum albumin and venous thrombosis: a bidirectional mendelian randomization analysis and bioinformatics study. Thromb J 2025; 23:17. [PMID: 40033322 DOI: 10.1186/s12959-025-00700-4] [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: 10/08/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND To explore the causal relationship between serum albumin and venous thromboembolism (VTE) comprises deep vein thrombosis (DVT) and its consequential condition, pulmonary embolism (PE), through Mendelian randomization (MR) design, seeking to clarify the protective roles of albumin in the development of venous thrombosis. METHODS We performed a bidirectional two-sample Mendelian randomization analysis utilizing albumin genome-wide association study (GWAS) data alongside VTE datasets from various sources. Additionally, to minimize heterogeneity across different datasets, a meta-analysis of the Mendelian randomization results was conducted. Furthermore, genes associated with such exposures were identified to unravel how exposure impacts outcomes. This was followed by applying bioinformatics techniques for gene enrichment analysis and employing the Cytoscape software to pinpoint the hub genes. RESULTS The findings from the meta-analysis of the Mendelian randomization indicate that reduced levels of albumin are associated with an elevated risk of VTE (OR = 0.739, 95% CI: 0.695 to 0.787, P = 1.82e-9), DVT (OR = 0.700, 95% CI: 0.646 to 0.772, P = 2.96e-15), and PE (OR = 0.717, 95% CI: 0.647 to 0.793, P = 1.74e-10). Bioinformatics analysis revealed that serum albumin primarily protects against VTE by influencing inflammation and cytokines. CONCLUSIONS Our bidirectional MR analysis confirmed a substantial causal association linking serum albumin to VTE. Bioinformatics analysis revealed that this causal link is mediated by the immune response through inflammation and cytokines.
Collapse
Affiliation(s)
- Xuemei Xia
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37, Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, China
| | - Xin Tie
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37, Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, China
| | - Maolin Hong
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37, Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, China
| | - Wanhong Yin
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37, Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, China.
| |
Collapse
|
7
|
Gong F, Zheng X, Zhao S, Liu H, Chen E, Xie R, Li R, Chen Y. Disseminated intravascular coagulation: cause, molecular mechanism, diagnosis, and therapy. MedComm (Beijing) 2025; 6:e70058. [PMID: 39822757 PMCID: PMC11733103 DOI: 10.1002/mco2.70058] [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: 08/30/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/19/2025] Open
Abstract
Disseminated intravascular coagulation (DIC) is a complex and serious condition characterized by widespread activation of the coagulation cascade, resulting in both thrombosis and bleeding. This review aims to provide a comprehensive overview of DIC, emphasizing its clinical significance and the need for improved management strategies. We explore the primary causes of DIC, including sepsis, trauma, malignancies, and obstetric complications, which trigger an overactive coagulation response. At the molecular level, DIC is marked by excessive thrombin generation, leading to platelet and fibrinogen activation while simultaneously depleting clotting factors, creating a paradoxical bleeding tendency. Diagnosing DIC is challenging and relies on a combination of existing diagnostic criteria and laboratory tests. Treatment strategies focus on addressing the underlying causes and may involve supportive care, anticoagulation therapy, and other supportive measures. Recent advances in understanding the pathophysiology of DIC are paving the way for more targeted therapeutic approaches. This review highlights the critical need for ongoing research to enhance diagnostic accuracy and treatment efficacy, ultimately improving patient outcomes in those affected by DIC.
Collapse
Affiliation(s)
- Fangchen Gong
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiangtao Zheng
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shanzhi Zhao
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Huan Liu
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Erzhen Chen
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of Aviation Medicine, Shanghai Jiao Tong University Medical School Affiliated Ruijin HospitalShanghaiChina
| | - Rongli Xie
- Department of General SurgeryRuijin Hospital Lu Wan Branch, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Ranran Li
- Department of Critical Care MedicineRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Chen
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Emergency and Critical Care MedicineRuijin Hospital Wuxi Branch, Shanghai Jiao Tong University School of MedicineWuxiChina
| |
Collapse
|
8
|
Abdulrahim AO, Doddapaneni NSP, Salman N, Giridharan A, Thomas J, Sharma K, Abboud E, Rochill K, Shreelakshmi B, Gupta V, Lakkimsetti M, Mowo-Wale A, Ali N. The gut-heart axis: a review of gut microbiota, dysbiosis, and cardiovascular disease development. Ann Med Surg (Lond) 2025; 87:177-191. [PMID: 40109640 PMCID: PMC11918638 DOI: 10.1097/ms9.0000000000002789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/20/2024] [Indexed: 03/22/2025] Open
Abstract
Background Cardiovascular diseases (CVDs) are a major cause of morbidity and mortality worldwide and there are strong links existing between gut health and cardiovascular health. Gut microbial diversity determines gut health. Dysbiosis, described as altered gut microbiota, causes bacterial translocations and abnormal gut byproducts resulting in systemic inflammation. Objective To review the current literature on the relationships between gut microbiota, dysbiosis, and CVD development, and explore therapeutic methods to prevent dysbiosis and support cardiovascular health. Summary Dysbiosis increases levels of pro-inflammatory substances while reducing those of anti-inflammatory substances. This accumulative inflammatory effect negatively modulates the immune system and promotes vascular dysfunction and atherosclerosis. High Firmicutes to Bacteroidetes ratios, high trimethylamine-n-oxide to short-chain fatty acid ratios, high indole sulfate levels, low cardiac output, and polypharmacy are all associated with worse cardiovascular outcomes. Supplementation with prebiotics and probiotics potentially alleviates some CVD risk. Blood and stool samples may be used in clinical practice to quantify and qualify gut bacterial ratios and byproducts, assess patients' risk for adverse cardiovascular outcomes, and track their gut health progress. Further research is required to set population-based cutoffs for normal and abnormal gut microbiota and byproduct ratios.
Collapse
Affiliation(s)
| | | | - Nadhra Salman
- Department of Internal Medicine, Baqai Medical University, Karachi, Pakistan
| | | | | | - Kavya Sharma
- Maharishi Markandeshwar Medical College and Hospital, Himachal Pradesh, India
| | - Elias Abboud
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | | | - B Shreelakshmi
- Navodaya Medical College Hospital & Research Centre, Karnataka, India
| | | | | | | | - Noor Ali
- Dubai Medical College, Dubai, United Arab Emirates
| |
Collapse
|
9
|
Nandakumar M, Das P, Sathyapalan T, Butler AE, Atkin SL. Cardiovascular Risk Biomarkers in Women with and Without Polycystic Ovary Syndrome. Biomolecules 2024; 15:4. [PMID: 39858399 PMCID: PMC11763313 DOI: 10.3390/biom15010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a prevalent metabolic disorder with an increased risk for cardiovascular disease (CVD) that is enhanced by obesity. This study sought to determine whether a panel of cardiovascular risk proteins (CVRPs) would be dysregulated in overweight/obese PCOS patients, highlighting potential biomarkers for CVD in PCOS. METHODS In this exploratory cross-sectional study, plasma levels of 54 CVRPs were analyzed in women with PCOS (n = 147) and controls (n = 97). CVRPs were measured using the SOMAscan proteomic platform (version 3.1), with significant proteins identified through linear models, regression analysis, and receiver operating characteristic (ROC) analysis. Analysis on BMI-matched subsets of the cohort were undertaken. Functional enrichment and protein-protein interaction analyses elucidated the pathways involved. RESULTS Eleven CVRPs were dysregulated in PCOS (whole set, without matching for body mass index (BMI) or insulin resistance (IR)): leptin, Interleukin-1 receptor antagonist protein (IL-1Ra), polymeric immunoglobulin receptor (PIGR), interleukin-18 receptor (IL-18Ra), C-C motif chemokine 3 (MIP-1a), and angiopoietin-1 (ANGPT1) were upregulated whilst advanced glycosylation end product-specific receptor, soluble (sRAGE), bone morphogenetic protein 6 (BMP6); growth/differentiation factor 2 (GDF2), superoxide dismutase [Mn] mitochondrial (MnSOD), and SLAM family member 5 (SLAF5) were downregulated versus the controls. In BMI-matched (overweight/obese, BMI ≥ 26 kg/m2) subset analysis, six CVRPs were common to the whole set: ANGPT1 and IL-1Ra were upregulated; and sRAGE, BMP6, GDF2, and Mn-SOD were downregulated. In addition, lymphotactin (XCL1) was upregulated and placenta growth factor (PIGF), alpha-L-iduronidase (IDUA), angiopoietin-1 receptor, and soluble (sTie-2) and macrophage metalloelastase (MMP12) were downregulated. A subset analysis of BMI-matched plus insulin resistance (IR)-matched women revealed only upregulation of tissue factor (TF) and renin in PCOS, potentially serving as biomarkers for cardiovascular risk in overweight/obese women with PCOS. CONCLUSIONS A combination of upregulated obesity-related CVRPs (ANGPT1/IL/1Ra/XCL1) and downregulated cardioprotective proteins (sRAGE/BMP6/Mn-SOD/GDF2) in overweight/obese PCOS women may contribute to the increased risk for CVD. TF and renin upregulation observed in the BMI- and IR-matched limited sample PCOS subgroup indicates their potential risk of CVD.
Collapse
Affiliation(s)
- Manjula Nandakumar
- Research Department, Royal College of Surgeons of Ireland, Adliya, Busaiteen 15503, Bahrain; (M.N.); (P.D.); (S.L.A.)
| | - Priya Das
- Research Department, Royal College of Surgeons of Ireland, Adliya, Busaiteen 15503, Bahrain; (M.N.); (P.D.); (S.L.A.)
| | - Thozhukat Sathyapalan
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull HU6 7RU, UK;
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons of Ireland, Adliya, Busaiteen 15503, Bahrain; (M.N.); (P.D.); (S.L.A.)
| | - Stephen L. Atkin
- Research Department, Royal College of Surgeons of Ireland, Adliya, Busaiteen 15503, Bahrain; (M.N.); (P.D.); (S.L.A.)
| |
Collapse
|
10
|
Heidari Z, Fallahi J, Sisakht M, Safari F, Hosseini K, Bahmanimehr A, Savardashtaki A, Khajeh S, Tabei SMB, Razban V. Impact of Tissue Factor Gene Knockout on Coagulation Properties of Umbilical Cord-Derived Multipotent Mesenchymal Stromal/Stem Cells. Cell Biochem Funct 2024; 42:e70021. [PMID: 39660566 DOI: 10.1002/cbf.70021] [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: 08/14/2024] [Revised: 10/18/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024]
Abstract
Multipotent mesenchymal stromal/stem cells (MSCs) refer to a population of stem cells that exhibit distinct progenitor cell characteristics including the potential for differentiation into a wide range of cell types. MSCs have become a promising candidate for cell therapy and tissue regeneration due to their unique properties, such as their ability to differentiate into multiple cell types, their capacity for expansion, self-renewal, and immune-regulatory effects. However, reports have brought attention to thrombosis-related complications associated with MSCs therapy in the last decade. As tissue factor (TF) is a powerful coagulation activator expressed by MSCs that stimulates the extrinsic coagulation pathway, we investigated the thrombotic properties of human umbilical cord MSCs (HUCMSCs) after knocking out the TF gene. MSCs populations that obtained from umbilical cord were cultured and expanded in the appropriate medium cell culture. The identity of the MSCs was verified through flow cytometry, and their ability to differentiate into osteogenic and adipogenic lineages. Two gRNAs for Exons 1 and 2 of the TF gene have been designed and cloned into px458 vector's backbone (pSpCas9 (BB)-2A-GFP). Following transfecting of gRNAs into HUCMSCs and successfully knocking out the TF gene using GAP-PCR, the impact of normal and knockout HUCMSCs on coagulation was assessed through prothrombin time (PT), D-dimer level, clotting time (CT), and turbidity assay. Furthermore, the impact of TF knockout (TFKO) on MMP19 expression was assessed. Our results revealed that the PT was prolonged and D-dimer level was decreased in TFKO group compared to normal HUCMSCs. These findings suggest that TF gene plays a crucial role in regulating coagulation in HUCMSCs. Also, a significant reduction in MMP19 expression was observed within the TFKO group.
Collapse
Affiliation(s)
- Zahra Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Sisakht
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamran Hosseini
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ardeshir Bahmanimehr
- Thalassemia and Hemophilia Genetic, PND Research Center, Dastgheib Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Orthopedic & Rehabilitation Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
George Pryzdial EL, Perrier JR, Rashid MU, West HE, Sutherland MR. Viral coagulation: pushing the envelope. J Thromb Haemost 2024; 22:3366-3382. [PMID: 39260743 DOI: 10.1016/j.jtha.2024.08.014] [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: 02/27/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Many virus types affect the blood clotting system with correlations to pathology that range widely from thrombosis to hemorrhage linking to inflammation. Here we overview the intricate crosstalk induced by infection between proteins on the virus encoded by either the host or virus genomes, coagulation proteins, platelets, leukocytes, and endothelial cells. For blood-borne viruses with an outer covering acquired from the host cell, the envelope, a key player may be the cell-derived trigger of coagulation on the virus surface, tissue factor (TF). TF is a multifunctional transmembrane cofactor that accelerates factor (F)VIIa-dependent activation of FX to FXa, leading to clot formation. However, the nascent TF/FVIIa/FXa complex also facilitates G protein-coupled modulation of cells via protease-activated receptor 2. As a viral envelope constituent, TF can bypass the physiological modes of regulation, thereby initiating the activation of neighboring platelets, leukocytes, and endothelial cells. A thromboinflammatory environment is predicted due to feedback amplification in response to cellular release of cytokines, procoagulant proteins, neutrophil extracellular traps, and stimulus-induced accessibility of adhesive receptors, resulting in cellular aggregates. The pathobiological effects of thromboinflammation ultimately contribute to innate and adaptive immunity for viral clearance. In contrast, the preceding stages of viral infection may be enhanced via the TF-protease axis.
Collapse
Affiliation(s)
- Edward Louis George Pryzdial
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada.
| | - John Ruggles Perrier
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Mahamud-Ur Rashid
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Henry Euan West
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Michael Ross Sutherland
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| |
Collapse
|
12
|
Hansildaar R, Raadsen R, Gerritsen M, Nagy M, Dijkshoorn B, Spronk HMH, Ten Cate H, Nurmohamed MT. Comparative Analysis of Coagulation Activation in Rheumatoid Arthritis Patients Treated With TNF Inhibitors Versus JAK Inhibitors: A Longitudinal Study. J Clin Rheumatol 2024; 30:e166-e171. [PMID: 39342416 DOI: 10.1097/rhu.0000000000002136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
OBJECTIVES This study aims to investigate the activation of the coagulation system of RA patients and assess changes during anti-inflammatory treatment with tumor necrosis factor blockers (anti-TNF) and Janus kinase inhibitors (JAKi). METHODS Biomarkers for the coagulation system, including D-dimer, fibrinogen, prothrombin time, activated partial thrombin time, prothrombin fragment 1 + 2, thrombin-antithrombin complex (TAT), activated factor IX, antithrombin complex, and von Willebrand factor (vWF), were longitudinally measured in 83 RA patients treated with anti-TNF and 38 RA patients with JAKi. Data were collected at baseline, after 1, 3, and 6 months. RESULTS The mean age was 57 (±14) years; 76% was female. The mean DAS28-CRP was 3.6 (±1.3) for anti-TNF users and 4.1 (±1.4) for JAKi users at baseline and declined in both groups. Baseline coagulation markers levels were comparable between groups. In anti-TNF users, D-dimer and fibrinogen levels significantly declined (-0.31 mg/L, p = 0.01 and -0.71 g/L, p < 0.001, respectively), whereas TAT significantly increased after 6 months follow-up (1.46 μg/L, p = 0.03) and no effect on vWF ( p = 0.98). In JAKi users, vWF declined significantly during the 6 months follow-up (-37.41%, p < 0.001); additionally, there were reductions of D-dimer, fibrinogen, and TAT that did not reach significance (-0.17 mg/L, p = 0.59; -0.49 g/L, p = 0.12; and 0.68 μg/L, p = 0.27, respectively). CONCLUSIONS The prothrombotic tendency in active RA declined during effective treatment with both anti-TNF and JAKi. Altogether, the biomarkers used in this study suggest that an increased VTE risk in the first 6 months due to either treatment with anti-TNF or JAKi is unlikely.
Collapse
Affiliation(s)
- Romy Hansildaar
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Location Reade, Amsterdam, the Netherlands
| | - Reinder Raadsen
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Location Reade, Amsterdam, the Netherlands
| | - Martijn Gerritsen
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Location Reade, Amsterdam, the Netherlands
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Bas Dijkshoorn
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Location Reade, Amsterdam, the Netherlands
| | | | | | | |
Collapse
|
13
|
Bokel J, Martins-Gonçalves R, Grinsztejn E, Mendes-de-Almeida DP, Hoagland B, Cardoso SW, Geraldo KM, Coutinho SN, Georg I, Oliveira MH, Dos Santos Souza F, Sacramento CQ, Rozini SV, Vizzoni AG, Veloso V, Bozza PT, Grinsztejn B. Anti-PF4 positivity and platelet activation after Ad26.COV2·S vaccination in Brazil. Vaccine 2024; 42:126175. [PMID: 39107160 DOI: 10.1016/j.vaccine.2024.126175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/13/2024] [Accepted: 07/22/2024] [Indexed: 08/09/2024]
Abstract
INTRODUCTION The Ad26.COV2·S (Janssen/Johnson & Johnson) COVID-19 vaccine, has been rarely associated with vaccine-induced immune thrombocytopenia and thrombosis (VITT). We investigated the prevalence of anti-PF4 antibody positivity, thrombocytopenia, D-dimer elevation, plasmatic thromboinflammatory markers, and platelet functional assays following Ad26.COV2·S vaccination in Rio de Janeiro, Brazil. METHODS From July to September 2021, participants were assessed prior, 1, and 3 weeks post-vaccination. Platelet count and D-dimer were measured at each visit and anti-PF4 at week 3. A positive anti-PF4 prompted retrospective testing of the sample from week 0. Individuals with new thrombocytopenia or elevated D-dimer, positive anti-PF4, and 38 matched controls without laboratory abnormalities were evaluated for plasmatic p-selectin, tissue factor, and functional platelet activation assays. RESULTS 630 individuals were included; 306 (48.57%) females, median age 28 years. Forty-two (6.67%) presented ≥1 laboratory abnormality in week 1 or 3. Five (0.79%) had thrombocytopenia, 31 (4.91%) elevated D-dimer, and 9 (1.57%) had positive anti-PF4 at week 3. Individuals with laboratory abnormalities and controls showed a slight increase in plasmatic p-selectin and tissue factor. Ten individuals with laboratory abnormalities yielded increased surface expression of p-selectin, and their ability to activate platelets in a FcγRIIa dependent manner was further evaluated. Two were partially inhibited by high concentrations of heparin and blockage of FcγRII with IV.3 antibody. Plasma obtained before vaccination produced similar results, suggesting a lack of association with vaccination. CONCLUSIONS Vaccination with Ad26.COV2·S vaccine led to a very low frequency of low-titer positive anti-PF4 antibodies, elevation of D-dimer, and mild thrombocytopenia, with no associated clinically relevant increase in thromboinflammatory markers and platelet activation.
Collapse
Affiliation(s)
- Joanna Bokel
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.
| | | | | | - Daniela P Mendes-de-Almeida
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Brenda Hoagland
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Sandra Wagner Cardoso
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Kim Mattos Geraldo
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Sandro Nazer Coutinho
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Ingebourg Georg
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Maria Helena Oliveira
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Flávia Dos Santos Souza
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Carolina Q Sacramento
- Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Stephane V Rozini
- Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Alexandre G Vizzoni
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Valdiléa Veloso
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Patrícia T Bozza
- Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Beatriz Grinsztejn
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Chea M, Bouvier S, Gris JC. The hemostatic system in chronic brain diseases: A new challenging frontier? Thromb Res 2024; 243:109154. [PMID: 39305718 DOI: 10.1016/j.thromres.2024.109154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/19/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Neurological diseases (ND), including neurodegenerative diseases (NDD) and psychiatric disorders (PD), present a significant public health challenge, ranking third in Europe for disability and premature death, following cardiovascular diseases and cancers. In 2017, approximately 540 million cases of ND were reported among Europe's 925 million people, with strokes, dementia, and headaches being most prevalent. Nowadays, more and more evidence highlight the hemostasis critical role in cerebral homeostasis and vascular events. Indeed, hemostasis, thrombosis, and brain abnormalities contributing to ND form a complex and poorly understood equilibrium. Alterations in vascular biology, particularly involving the blood-brain barrier, are implicated in ND, especially dementia, and PD. While the roles of key coagulation players such as thrombin and fibrinogen are established, the roles of other hemostasis components are less clear. Moreover, the involvement of these elements in psychiatric disease pathogenesis is virtually unstudied, except in specific pathological models such as antiphospholipid syndrome. Advanced imaging techniques, primarily functional magnetic resonance imaging and its derivatives like diffusion tensor imaging, have been developed to study brain areas affected by ND and to improve our understanding of the pathophysiology of these diseases. This literature review aims to clarify the current understanding of the connections between hemostasis, thrombosis, and neurological diseases, as well as explore potential future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mathias Chea
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France.
| | - Sylvie Bouvier
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Jean-Christophe Gris
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France; I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
15
|
De Luca G, Goette NP, Lev PR, Baroni Pietto MC, Marin Oyarzún CP, Castro Ríos MA, Moiraghi B, Sackmann F, Kamiya LJ, Verri V, Caula V, Fernandez V, Vicente A, Pose Cabarcos J, Caruso V, Camacho MF, Larripa IB, Khoury M, Marta RF, Glembotsky AC, Heller PG. Elevated levels of damage-associated molecular patterns HMGB1 and S100A8/A9 coupled with toll-like receptor-triggered monocyte activation are associated with inflammation in patients with myelofibrosis. Front Immunol 2024; 15:1365015. [PMID: 39391311 PMCID: PMC11465240 DOI: 10.3389/fimmu.2024.1365015] [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: 01/03/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Inflammation plays a pivotal role in the pathogenesis of primary and post-essential thrombocythemia or post-polycythemia vera myelofibrosis (MF) in close cooperation with the underlying molecular drivers. This inflammatory state is induced by a dynamic spectrum of inflammatory cytokines, although recent evidence points to the participation of additional soluble inflammatory mediators. Damage-associated molecular patterns (DAMPs) represent endogenous signals released upon cell death or damage which trigger a potent innate immune response. We assessed the contribution of two prototypical DAMPs, HMGB1 and S100A8/A9, to MF inflammation. Circulating HMGB1 and S100A8/A9 were elevated in MF patients in parallel to the degree of systemic inflammation and levels increased progressively during advanced disease stages. Patients with elevated DAMPs had higher frequency of adverse clinical features, such as anemia, and inferior survival, suggesting their contribution to disease progression. Monocytes, which are key players in MF inflammation, were identified as a source of S100A8/A9 but not HMGB1 release, while both DAMPs correlated with cell death parameters, such as serum LDH and cell-free DNA, indicating that passive release is an additional mechanism leading to increased DAMPs. HMGB1 and S100A8/A9 promote inflammation through binding to Toll-like receptor (TLR) 4, whereas the former also binds TLR2. Monocytes from MF patients were shown to be hyperactivated at baseline, as reflected by higher CD11b and tissue factor exposure and increased expression levels of proinflammatory cytokines IL-1β and IL-6. Patient monocytes showed preserved TLR4 and TLR2 expression and were able to mount normal or even exacerbated functional responses and cytokine upregulation following stimulation of TLR4 and TLR2. Elevated levels of endogenous TLR ligands HMGB1 and S100A8/A9 coupled to the finding of preserved or hyperreactive TLR-triggered responses indicate that DAMPs may promote monocyte activation and cytokine production in MF, fueling inflammation. Plasma IL-1β and IL-6 were elevated in MF and correlated with DAMPs levels, raising the possibility that DAMPs could contribute to cytokine generation in vivo. In conclusion, this study highlights that, in cooperation with classic proinflammatory cytokines, DAMPs represent additional inflammatory mediators that may participate in the generation of MF inflammatory state, potentially providing novel biomarkers of disease progression and new therapeutic targets.
Collapse
Affiliation(s)
- Geraldine De Luca
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Nora P. Goette
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Paola R. Lev
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Maria C. Baroni Pietto
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cecilia P. Marin Oyarzún
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | | | | | - Laureano J. Kamiya
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Veronica Verri
- División Hematología Clínica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Victoria Caula
- División Hematología Clínica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vanina Fernandez
- Departamento de Hematología, Hospital Posadas, Buenos Aires, Argentina
| | - Angeles Vicente
- Departamento de Hematología, Hospital Alemán, Buenos Aires, Argentina
| | - Julio Pose Cabarcos
- Departamento de Hematología, Sanatorio Otamendi Miroli, Buenos Aires, Argentina
| | - Vanesa Caruso
- Departamento de Hematología, Hospital Piñero, Buenos Aires, Argentina
| | - Maria F. Camacho
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Irene B. Larripa
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Marina Khoury
- Departamento de Docencia e Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosana F. Marta
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana C. Glembotsky
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paula G. Heller
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
16
|
Triantafyllis AS, Sfantou D, Karapedi E, Peteinaki K, Kotoulas SC, Saad R, Fountoulakis PN, Tsamakis K, Tsiptsios D, Rallidis L, Tsoporis JN, Varvarousis D, Hamodraka E, Giannakopoulos A, Poulimenos LE, Ikonomidis I. Coronary Implications of COVID-19. Med Princ Pract 2024; 34:1-12. [PMID: 39307131 PMCID: PMC11805551 DOI: 10.1159/000541553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Patients with SARS-CoV-2 infection carry an increased risk of cardiovascular disease encompassing various implications, including acute myocardial injury or infarction, myocarditis, heart failure, and arrhythmias. A growing volume of evidence correlates SARS-CoV-2 infection with myocardial injury, exposing patients to higher mortality risk. SARS-CoV-2 attacks the coronary arterial bed with various mechanisms including thrombosis/rupture of preexisting atherosclerotic plaque, de novo coronary thrombosis, endotheliitis, microvascular dysfunction, vasculitis, vasospasm, and ectasia/aneurysm formation. The angiotensin-converting enzyme 2 receptor plays pivotal role on the cardiovascular homeostasis and the unfolding of COVID-19. The activation of immune system, mediated by proinflammatory cytokines along with the dysregulation of the coagulation system, can pose an insult on the coronary artery, which usually manifests as an acute coronary syndrome (ACS). Electrocardiogram, echocardiography, cardiac biomarkers, and coronary angiography are essential tools to set the diagnosis. Revascularization is the first-line treatment in all patients with ACS and obstructed coronary arteries, whereas in type 2 myocardial infarction treatment of hypoxia, anemia and systemic inflammation are indicated. In patients presenting with coronary vasospasm, nitrates and calcium channel blockers are preferred, while treatment of coronary ectasia/aneurysm mandates the use of antiplatelets/anticoagulants, corticosteroids, immunoglobulin, and biologic agents. It is crucial to untangle the exact mechanisms of coronary involvement in COVID-19 in order to ensure timely diagnosis and appropriate treatment. We have reviewed the current literature and provide a detailed overview of the pathophysiology and clinical spectrum associated with coronary implications of SARS-COV-2 infection. Patients with SARS-CoV-2 infection carry an increased risk of cardiovascular disease encompassing various implications, including acute myocardial injury or infarction, myocarditis, heart failure, and arrhythmias. A growing volume of evidence correlates SARS-CoV-2 infection with myocardial injury, exposing patients to higher mortality risk. SARS-CoV-2 attacks the coronary arterial bed with various mechanisms including thrombosis/rupture of preexisting atherosclerotic plaque, de novo coronary thrombosis, endotheliitis, microvascular dysfunction, vasculitis, vasospasm, and ectasia/aneurysm formation. The angiotensin-converting enzyme 2 receptor plays pivotal role on the cardiovascular homeostasis and the unfolding of COVID-19. The activation of immune system, mediated by proinflammatory cytokines along with the dysregulation of the coagulation system, can pose an insult on the coronary artery, which usually manifests as an acute coronary syndrome (ACS). Electrocardiogram, echocardiography, cardiac biomarkers, and coronary angiography are essential tools to set the diagnosis. Revascularization is the first-line treatment in all patients with ACS and obstructed coronary arteries, whereas in type 2 myocardial infarction treatment of hypoxia, anemia and systemic inflammation are indicated. In patients presenting with coronary vasospasm, nitrates and calcium channel blockers are preferred, while treatment of coronary ectasia/aneurysm mandates the use of antiplatelets/anticoagulants, corticosteroids, immunoglobulin, and biologic agents. It is crucial to untangle the exact mechanisms of coronary involvement in COVID-19 in order to ensure timely diagnosis and appropriate treatment. We have reviewed the current literature and provide a detailed overview of the pathophysiology and clinical spectrum associated with coronary implications of SARS-COV-2 infection.
Collapse
Affiliation(s)
| | - Danai Sfantou
- Department of Cardiology, Asklepeion General Hospital, Athens, Greece
| | - Eleni Karapedi
- Department of Cardiology, Asklepeion General Hospital, Athens, Greece
| | | | | | - Richard Saad
- Department of Cardiology, Asklepeion General Hospital, Athens, Greece
| | | | | | - Dimitrios Tsiptsios
- Department of Neurology, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Loukianos Rallidis
- Second Department of Cardiology, Attikon University Hospital, Athens, Greece
| | - James N. Tsoporis
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | - Ignatios Ikonomidis
- Second Department of Cardiology, Attikon University Hospital, Athens, Greece
| |
Collapse
|
17
|
Pharoun J, Berro J, Sobh J, Abou-Younes MM, Nasr L, Majed A, Khalil A, Joseph, Stephan, Faour WH. Mesenchymal stem cells biological and biotechnological advances: Implications for clinical applications. Eur J Pharmacol 2024; 977:176719. [PMID: 38849038 DOI: 10.1016/j.ejphar.2024.176719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stem cells that are able to differentiate into multiple lineages including bone, cartilage, muscle and fat. They hold immunomodulatory properties and therapeutic ability to treat multiple diseases, including autoimmune and chronic degenerative diseases. In this article, we reviewed the different biological properties, applications and clinical trials of MSCs. Also, we discussed the basics of manufacturing conditions, quality control, and challenges facing MSCs in the clinical setting. METHODS Extensive review of the literature was conducted through the databases PubMed, Google Scholar, and Cochrane. Papers published since 2015 and covering the clinical applications and research of MSC therapy were considered. Furthermore, older papers were considered when referring to pioneering studies in the field. RESULTS The most widely studied stem cells in cell therapy and tissue repair are bone marrow-derived mesenchymal stem cells. Adipose tissue-derived stem cells became more common and to a lesser extent other stem cell sources e.g., foreskin derived MSCs. MSCs therapy were also studied in the setting of COVID-19 infections, ischemic strokes, autoimmune diseases, tumor development and graft rejection. Multiple obstacles, still face the standardization and optimization of MSC therapy such as the survival and the immunophenotype and the efficiency of transplanted cells. MSCs used in clinical settings displayed heterogeneity in their function despite their extraction from healthy donors and expression of similar surface markers. CONCLUSION Mesenchymal stem cells offer a rising therapeutic promise in various diseases. However, their potential use in clinical applications requires further investigation.
Collapse
Affiliation(s)
- Jana Pharoun
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Jana Berro
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Jeanine Sobh
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | | | - Leah Nasr
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Ali Majed
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Alia Khalil
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Joseph
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Stephan
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36.
| |
Collapse
|
18
|
Peng X, Zhang X, Zhao M, Chang D, Yang L, Mei H, Zhang R. Coagulation abnormalities associated with CAR-T-cell therapy in haematological malignancies: A review. Br J Haematol 2024; 205:420-428. [PMID: 38887101 DOI: 10.1111/bjh.19583] [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: 12/30/2023] [Accepted: 05/27/2024] [Indexed: 06/20/2024]
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has demonstrated considerable efficacy and safety in the treatment of patients with relapsed/refractory haematological malignancies. Owing to significant advances, CAR-T-cell therapeutic modality has undergone substantial shifts in its clinical application. Coagulation abnormalities, which are prevalent complications in CAR-T-cell therapy, can range in severity from simple abnormalities in coagulation parameters to serious haemorrhage or disseminated intravascular coagulation associated with life-threatening multiorgan dysfunction. Nonetheless, there is a lack of a comprehensive overview concerning the coagulation abnormalities associated with CAR-T-cell therapy. With an aim to attract heightened clinical focus and to enhance the safety of CAR-T-cell therapy, this review presents the characteristics of the coagulation abnormalities associated with CAR-T-cell therapy, including clinical manifestations, coagulation parameters, pathogenesis, risk factors and their influence on treatment efficacy in patients receiving CAR-T-cell infusion. Due to limited data, these conclusions may undergo changes as more experience accumulates.
Collapse
Affiliation(s)
- Xiaojuan Peng
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, ShanXi, China
| | - Xialin Zhang
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, ShanXi, China
| | - Meiling Zhao
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, ShanXi, China
| | - Doudou Chang
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, ShanXi, China
| | - Linhua Yang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, ShanXi, China
| | - Heng Mei
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruijuan Zhang
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, ShanXi, China
| |
Collapse
|
19
|
刘 萌, 侯 改, 杨 晓, 张 秋, 梅 晓, 丁 樱, 宋 兰, 黄 岩. [Exploring the mechanism of IgA vasculitis pathogenesis through the interaction of thrombin and inflammatory factors using urinary proteomics]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:683-689. [PMID: 39014943 PMCID: PMC11562050 DOI: 10.7499/j.issn.1008-8830.2311151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/30/2024] [Indexed: 07/18/2024]
Abstract
OBJECTIVES To explore the evidence, urinary biomarkers, and partial mechanisms of hypercoagulability in the pathogenesis of IgA vasculitis (IgAV). METHODS Differential expression of proteins in the urine of 10 healthy children and 10 children with IgAV was screened using high-performance liquid chromatography-tandem mass spectrometry, followed by Reactome pathway analysis. Protein-protein interaction (PPI) network analysis was conducted using STRING and Cytoscape software. In the validation cohort, 15 healthy children and 25 children with IgAV were included, and the expression levels of differential urinary proteins were verified using enzyme-linked immunosorbent assay. RESULTS A total of 772 differential proteins were identified between the IgAV group and the control group, with 768 upregulated and 4 downregulated. Reactome pathway enrichment results showed that neutrophil degranulation, platelet activation, and hemostasis pathways were involved in the pathogenesis of IgAV. Among the differential proteins, macrophage migration inhibitory factor (MIF) played a significant role in neutrophil degranulation and hemostasis, while thrombin was a key protein in platelet activation and hemostasis pathways. PPI analysis indicated that thrombin directly interacted with several proteins involved in inflammatory responses, and these interactions involved MIF. Validation results showed that compared to healthy children, children with IgAV had significantly higher urine thrombin/creatinine and urine MIF/creatinine levels (P<0.05). CONCLUSIONS Thrombin contributes to the pathogenesis of IgAV through interactions with inflammatory factors. Urinary thrombin and MIF can serve as biomarkers reflecting the hypercoagulable and inflammatory states in children with IgAV.
Collapse
Affiliation(s)
- 萌萌 刘
- 河南中医药大学第一附属医院儿科医院,河南郑州450046
| | - 改灵 侯
- 河南中医药大学第一附属医院儿科医院,河南郑州450046
| | - 晓青 杨
- 河南中医药大学第一附属医院儿科医院,河南郑州450046
| | - 秋爽 张
- 河南中医药大学第一附属医院儿科医院,河南郑州450046
| | - 晓峰 梅
- 河南中医药大学第一附属医院儿科医院,河南郑州450046
| | - 樱 丁
- 河南中医药大学第一附属医院儿科医院,河南郑州450046
| | | | - 岩杰 黄
- 上海市儿童医院/上海交通大学医学院附属儿童医院;中医科上海200062
| |
Collapse
|
20
|
Wang L, Ling J, Zhu X, Zhang Y, Li R, Huang J, Huang D, Wu C, Zhou H. The coagulation status in women of endometriosis with stage IV. BMC Womens Health 2024; 24:386. [PMID: 38961373 PMCID: PMC11223421 DOI: 10.1186/s12905-024-03227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Endometriosis is considered as a systemic disease with the presence of proinflammatory cytokines in the circulation, which drives hypercoagulable state of endometriosis. Currently, endometriosis is classified into four stages: I (minimal), II (mild), III (moderate) and IV (severe). The aim of this study is to investigate the correlations between inflammatory markers and coagulation factors in patients diagnosed of endometriosis with stage IV. METHODS This retrospective case-control study included 171 endometriosis patients with stage IV and 184 controls. Continuous data were expressed by mean ± standard deviation. Mann-Whitney U and χ2 tests were used to compare the medians and frequencies among the groups. Spearman analysis was conducted to determine the correlation among the measured parameters. The diagnostic values of the parameters differentiating endometriomas were tested by receiver operating characteristic (ROC) curve. RESULTS The time of activated partial thromboplastin time (APTT) was decreased and the concentration of fibrinogen (FIB) and neutrophil-to-lymphocyte ratio (NLR) were increased in women of endometriosis with stage IV. The APTT were negatively correlated with NLR while the concentrations of FIB were positively correlated with NLR. The ROC analysis showed that the Area under the curve (AUC) of FIB was 0.766 (95% confidence interval:0.717-0.814) with sensitivity and specificity reaching 86.5 and 60.9%, respectively. The AUC of CA125 and CA199 was 0.638 (95% confidence interval: 0.578-0.697), 0.71 (95% confidence interval: 0.656-0.763) with sensitivity and specificity reaching 40.9 and 91.8%, 80.7 and 56.5% respectively. The combination of these factors showed the highest AUC of 0.895 (0.862-0.927) with sensitivity of 88.9% and specificity of 77.7%. CONCLUSION In the present study, we found that inflammatory factors showed significant correlation with APTT or FIB in endometriosis with stage IV. Moreover, the coagulation factors combined with CA125 and CA199 were more reliable for identifying the endometriosis with stage IV.
Collapse
Affiliation(s)
- Lu Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Jingxian Ling
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Xianghong Zhu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Rong Li
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Jingjing Huang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Doudou Huang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Chan Wu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
| | - Huaijun Zhou
- Department of Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
| |
Collapse
|
21
|
Friebel J, Wegner M, Blöbaum L, Schencke PA, Jakobs K, Puccini M, Ghanbari E, Lammel S, Thevathasan T, Moos V, Witkowski M, Landmesser U, Rauch-Kröhnert U. Characterization of Biomarkers of Thrombo-Inflammation in Patients with First-Diagnosed Atrial Fibrillation. Int J Mol Sci 2024; 25:4109. [PMID: 38612918 PMCID: PMC11012942 DOI: 10.3390/ijms25074109] [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: 01/26/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Patients with first-diagnosed atrial fibrillation (FDAF) exhibit major adverse cardiovascular events (MACEs) during follow-up. Preclinical models have demonstrated that thrombo-inflammation mediates adverse cardiac remodeling and atherothrombotic events. We have hypothesized that thrombin activity (FIIa) links coagulation with inflammation and cardiac fibrosis/dysfunction. Surrogate markers of the thrombo-inflammatory response in plasma have not been characterized in FDAF. In this prospective longitudinal study, patients presenting with FDAF (n = 80), and 20 matched controls, were included. FIIa generation and activity in plasma were increased in the patients with early AF compared to the patients with chronic cardiovascular disease without AF (controls; p < 0.0001). This increase was accompanied by elevated biomarkers (ELISA) of platelet and endothelial activation in plasma. Pro-inflammatory peripheral immune cells (TNF-α+ or IL-6+) that expressed FIIa-activated protease-activated receptor 1 (PAR1) (flow cytometry) circulated more frequently in patients with FDAF compared to the controls (p < 0.0001). FIIa activity correlated with cardiac fibrosis (collagen turnover) and cardiac dysfunction (NT-pro ANP/NT-pro BNP) surrogate markers. FIIa activity in plasma was higher in patients with FDAF who experienced MACE. Signaling via FIIa might be a presumed link between the coagulation system (tissue factor-FXa/FIIa-PAR1 axis), inflammation, and pro-fibrotic pathways (thrombo-inflammation) in FDAF.
Collapse
Affiliation(s)
- Julian Friebel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Max Wegner
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Leon Blöbaum
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Philipp-Alexander Schencke
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Kai Jakobs
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Marianna Puccini
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Emily Ghanbari
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Stella Lammel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Tharusan Thevathasan
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Verena Moos
- Medical Department I, Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Marco Witkowski
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- Friede Springer Cardiovascular Prevention Center at Charité, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Ursula Rauch-Kröhnert
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| |
Collapse
|
22
|
Li X, Hu L, Naeem A, Xiao S, Yang M, Shang H, Zhang J. Neutrophil Extracellular Traps in Tumors and Potential Use of Traditional Herbal Medicine Formulations for Its Regulation. Int J Nanomedicine 2024; 19:2851-2877. [PMID: 38529365 PMCID: PMC10961241 DOI: 10.2147/ijn.s449181] [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: 11/23/2023] [Accepted: 02/28/2024] [Indexed: 03/27/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are extracellular fibers composed of deoxyribonucleic acid (DNA) and decorated proteins produced by neutrophils. Recently, NETs have been associated with the development of many diseases, including tumors. Herein, we reviewed the correlation between NETs and tumors. In addition, we detailed active compounds from traditional herbal medicine formulations that inhibit NETs, related nanodrug delivery systems, and antibodies that serve as "guiding moieties" to ensure targeted delivery to NETs. Furthermore, we discussed the strategies used by pathogenic microorganisms to evade NETs.
Collapse
Affiliation(s)
- Xiang Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Lei Hu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Abid Naeem
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, People’s Republic of China
| | - Shanghua Xiao
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
| | - Ming Yang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
| | - Hongming Shang
- Department of Biochemistry & Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Jing Zhang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
| |
Collapse
|
23
|
Costantini TW, Kornblith LZ, Pritts T, Coimbra R. The intersection of coagulation activation and inflammation after injury: What you need to know. J Trauma Acute Care Surg 2024; 96:347-356. [PMID: 37962222 PMCID: PMC11001294 DOI: 10.1097/ta.0000000000004190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Affiliation(s)
- Todd W Costantini
- From the Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery (T.W.C.), UC San Diego School of Medicine, San Diego; Department of Surgery (L.Z.K.), Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, California; Department of Surgery (T.P.), University of Cincinnati College of Medicine, Cincinnati, Ohio; and Comparative Effectiveness and Clinical Outcomes Research Center (R.C.), Riverside University Health System, Loma Linda University School of Medicine, Riverside, California
| | | | | | | |
Collapse
|
24
|
Catherine Prater M, Polley KR, Cooper JA. Improvements in markers of inflammation and coagulation potential following a 5-day high-fat diet rich in cottonseed oil vs. Olive oil in healthy males. Cytokine 2024; 175:156494. [PMID: 38171039 DOI: 10.1016/j.cyto.2023.156494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/14/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
Low-grade inflammation is believed to be a risk factor for chronic diseases and is nutritionally responsive. Cottonseed oil (CSO), which is rich in n-6 polyunsaturated fats, has been shown to lower cholesterol and other chronic disease risk factors. The purpose of this secondary analysis was to determine the comparative responses of markers of inflammation and coagulation potential of healthy adult males consuming diets rich in CSO vs. olive oil (OO). METHODS Fifteen normal-weight males, ages 21.7 ± 2.58y, completed a randomized crossover trial. Each intervention consisted of a 3-day lead-in diet and a 5-day outpatient, controlled feeding intervention (CSO or OO). There was a 2 to 4-week washout period between interventions. The 5-day intervention diets were 35 % carbohydrate, 15 % protein, and 50 % fat, enriched with either CSO or OO (44 % of total energy from oil). At pre- and post- diet intervention visits, a fasting blood draw was collected for analysis of markers of inflammation (Tumor Necrosis Factor Alpha (TNF-α), Interleukin-6 (IL-6), C-Reactive Protein (CRP)) and coagulation potential (Tissue Factor (TF), Plasminogen Activator Inhibitor-1 (PAI-1)). RESULTS The CSO-enriched diets reduced TNF-α (CSO: -0.12 ± 0.02 pg/ml, OO: -0.01 ± 0.05 pg/ml; p < 0.01) and TF (CSO: -0.59 ± 0.68 pg/ml, OO: 1.13 ± 0.83 pg/ml; p = 0.02) compared to OO diets. There were no differences in IL-6, CRP, or PAI-1 between diets. CONCLUSION A 5-day, CSO-enriched diet may be sufficient to reduce inflammation and coagulation potential compared to OO-enriched diets in a healthy male population which could have implications in chronic disease prevention.
Collapse
Affiliation(s)
- M Catherine Prater
- Department of Nutritional Sciences, University of Georgia, 305 Sanford Drive, Athens, GA 30602, USA
| | - Kristine R Polley
- Department of Nutritional Sciences, University of Georgia, 305 Sanford Drive, Athens, GA 30602, USA
| | - Jamie A Cooper
- Department of Kinesiology, University of Georgia, 330 River Road, Athens, GA 30602, USA.
| |
Collapse
|
25
|
Prater MC, Scheurell AR, Paton CM, Cooper JA. No Observed Difference in Inflammatory and Coagulation Markers Following Diets Rich in n-6 Polyunsaturated Fat vs Monounsaturated Fat in Adults With Untreated Hypercholesterolemia: A Randomized Trial. J Acad Nutr Diet 2024; 124:205-214.e1. [PMID: 37619782 DOI: 10.1016/j.jand.2023.08.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Inflammatory and prothrombotic responses are hallmark to the progression of cardiovascular disease and may be influenced by the type of dietary fat. Cottonseed oil (CSO) is rich in n-6 polyunsaturated fats and improves traditional cardiovascular disease risk factors such as cholesterol profiles. However, some clinicians are still hesitant to promote n-6 polyunsaturated fats consumption despite growing evidence suggesting they may not be independently pro-inflammatory. OBJECTIVE To investigate the inflammatory and coagulation marker responses to an 8-week diet intervention rich in either CSO or olive oil (OO) (OO is rich in monounsaturated fat) in adults with untreated hypercholesterolemia. DESIGN This was a secondary analysis of a parallel-arm randomized clinical trial with the main outcome of cholesterol measures. PARTICIPANTS/SETTING Participants included in this analysis were 42 sedentary adults aged 30 to 75 years (62% women) in the Athens, GA, area, between May 2018 and June 2021, with untreated hypercholesterolemia or elevated blood lipids and body mass index >18.5. Hypercholesterolemia was defined as at least two blood lipid levels in a borderline undesirable/at risk range (total cholesterol level ≥180 mg/dL, low-density lipoprotein cholesterol level ≥110 mg/dL, high-density lipoprotein cholesterol level <50 mg/dL, or triglyceride level ≥130 mg/dL), or at least one in an undesirable range (total cholesterol level ≥240 mg/dL, low-density lipoprotein cholesterol level ≥160 mg/dL, high-density lipoprotein cholesterol level <40 mg/dL, or triglyceride level ≥200 mg/dL). INTERVENTION Participants were randomly assigned to either the CSO or OO group in a partial outpatient feeding trial. Meals from the study provided approximately 60% of their energy needs with 30% of energy needs from either CSO or OO for 8 weeks. Participants fulfilled their remaining energy needs with meals of their choosing. MAIN OUTCOME MEASURES Fasting plasma concentrations of inflammatory markers, including C-reactive protein, tumor necrosis factor-α, interleukin-6, and interleukin-1β were measured at baseline and 8 weeks. Markers of coagulation potential, including plasminogen activator inhibitor-1, and tissue factor were measured at the same time points. STATISTICAL ANALYSES PERFORMED Repeated measures linear mixed models were used with treatment and visit in the model for analyses of all biochemical markers. RESULTS There were no significant differences in fasting C-reactive protein (P = 0.70), tumor necrosis factor-α (P = 0.98), interleukin-6 (P = 0.21), interleukin-1β (P = 0.13), plasminogen activator inhibitor-1 (P = 0.29), or tissue factor (P = 0.29) between groups across the intervention. CONCLUSIONS Inflammation and coagulation marker responses to diets rich in CSO vs OO were not significantly different between groups, and neither group showed changes in these markers in adults with untreated hypercholesterolemia. This provides additional evidence suggesting that dietary n-6 polyunsaturated fats may not promote inflammation compared with monounsaturated fatty acids, even in adults at increased risk for cardiovascular disease.
Collapse
Affiliation(s)
- M Catherine Prater
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia
| | - Alexis R Scheurell
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia
| | - Chad M Paton
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia; Department of Food Science and Technology, University of Georgia, Athens, Georgia
| | - Jamie A Cooper
- Department of Kinesiology, University of Georgia, Athens, Georgia.
| |
Collapse
|
26
|
Meng S, Xu B, Yang W, Zhao M. Microparticle-associated tissue factor activity correlates with the inflammatory response in septic disseminated intravascular coagulation patients. PeerJ 2024; 12:e16636. [PMID: 38213768 PMCID: PMC10782946 DOI: 10.7717/peerj.16636] [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: 03/07/2023] [Accepted: 11/18/2023] [Indexed: 01/13/2024] Open
Abstract
Background Sepsis is often accompanied by the formation of disseminated intravascular coagulation (DIC). Microparticles can exert their procoagulant and proinflammatory properties in a variety of ways. The purpose of this study was to investigate the relationship between microparticle-associated tissue factor activity (TF+-MP activity) and the inflammatory response. Methods Data from a total of 31 DIC patients with sepsis and 31 non-DIC patients with sepsis admitted to the ICU of the First Affiliated Hospital of Harbin Medical University from December 2017 to March 2019 were collected. Blood samples were collected and DIC scores were calculated on the day of enrollment. The hospital's clinical laboratory completed routine blood, procalcitonin, and C-reactive protein tests. TF+-MP activity was measured using a tissue factor-dependent FXa generation assay. Interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) levels were determined using ELISA kits. Results Compared with the non-DIC group, the DIC group had higher levels of leukocytes, neutrophils, procalcitonin, C-reactive protein, IL-1β, and TNF-α, and more severe inflammatory reactions. TF+-MP activity in the DIC group was higher than that in the non-DIC group. In sepsis patients, TF+-MP activity was strongly correlated with inflammatory response indices and DIC scores. Conclusion TF+-MP activity may play a major role in promoting inflammatory response in septic DIC.
Collapse
Affiliation(s)
- Shishuai Meng
- Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Bin Xu
- Department of Cardiology, Cardiology, Harbin First Hospital, Harbin, Heilongjiang, China
| | - Wei Yang
- Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Mingyan Zhao
- Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
27
|
Martinelli N, Moruzzi S, Udali S, Castagna A, Di Santo L, Ambrosani F, Baroni M, Pattini P, Pizzolo F, Ruzzenente A, Conci S, Grusse M, Campagnaro T, Van Dreden P, Guglielmi A, Bernardi F, Olivieri O, Friso S. Tissue factor pathway-related biomarkers in liver cancer: activated factor VII-antithrombin complex and tissue factor mRNA levels are associated with mortality. Res Pract Thromb Haemost 2024; 8:102310. [PMID: 38282902 PMCID: PMC10818084 DOI: 10.1016/j.rpth.2023.102310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024] Open
Abstract
Background Tissue factor (TF), the main initiator of the coagulation cascade, plays a role in cancer progression and prognosis. Activated factor VII-antithrombin complex (FVIIa-AT) is considered an indirect marker of TF exposure by reflecting TF-FVIIa interaction. Objectives To assess the link between FVIIa-AT plasma levels, TF messenger RNA (mRNA) expression, and survival in cancer. Methods TF pathway-related coagulation biomarkers were assessed in 136 patients with cancer (52 with hepatocellular carcinoma, 41 with cholangiocarcinoma, and 43 with colon cancer) undergoing surgical intervention with curative intent. TF mRNA expression analysis in neoplastic vs nonneoplastic liver tissues was evaluated in a subgroup of 91 patients with primary liver cancer. Results FVIIa-AT levels were higher in patients with cancer than in 136 sex- and age-matched cancer-free controls. In patients with cancer, high levels of FVIIa-AT and total TF pathway inhibitor were associated with an increased mortality risk after adjustment for confounders, but only FVIIa-AT remained a predictor of mortality by including both FVIIa-AT and total TF pathway inhibitor in Cox regression (hazard ratio, 2.80; 95% CI, 1.23-6.39; the highest vs the lowest quartile). This association remained significant even after adjustment for extracellular vesicle-associated TF-dependent procoagulant activity. In the subgroup of patients with primary liver cancer, patients with high TF mRNA levels had an increased mortality risk compared with that for those with low TF mRNA levels (hazard ratio, 1.92; 95% CI, 1.03-3.57), and there was a consistent correlation among high FVIIa-AT levels, high TF mRNA levels, and increased risk of mortality. Conclusion High FVIIa-AT levels may allow the identification of patients with cancer involving high TF expression and predict a higher mortality risk in liver cancer.
Collapse
Affiliation(s)
| | - Sara Moruzzi
- Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Udali
- Department of Medicine, University of Verona, Verona, Italy
| | | | - Laura Di Santo
- Department of Medicine, University of Verona, Verona, Italy
| | | | - Marcello Baroni
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | | | | | - Simone Conci
- Department of Surgery, University of Verona, Verona, Italy
| | - Matthieu Grusse
- Clinical Research Department, Diagnostica Stago, Gennevilliers, France
| | | | | | | | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | | |
Collapse
|
28
|
Heidari Z, Naeimzadeh Y, Fallahi J, Savardashtaki A, Razban V, Khajeh S. The Role of Tissue Factor In Signaling Pathways of Pathological Conditions and Angiogenesis. Curr Mol Med 2024; 24:1135-1151. [PMID: 37817529 DOI: 10.2174/0115665240258746230919165935] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/10/2023] [Accepted: 07/27/2023] [Indexed: 10/12/2023]
Abstract
Tissue factor (TF) is an integral transmembrane protein associated with the extrinsic coagulation pathway. TF gene expression is regulated in response to inflammatory cytokines, bacterial lipopolysaccharides, and mechanical injuries. TF activity may be affected by phosphorylation of its cytoplasmic domain and alternative splicing. TF acts as the primary initiator of physiological hemostasis, which prevents local bleeding at the injury site. However, aberrant expression of TF, accompanied by the severity of diseases and infections under various pathological conditions, triggers multiple signaling pathways that support thrombosis, angiogenesis, inflammation, and metastasis. Protease-activated receptors (PARs) are central in the downstream signaling pathways of TF. In this study, we have reviewed the TF signaling pathways in different pathological conditions, such as wound injury, asthma, cardiovascular diseases (CVDs), viral infections, cancer and pathological angiogenesis. Angiogenic activities of TF are critical in the repair of wound injuries and aggressive behavior of tumors, which are mainly performed by the actions of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1 (HIF1-α). Pro-inflammatory effects of TF have been reported in asthma, CVDs and viral infections, including COVID-19, which result in tissue hypertrophy, inflammation, and thrombosis. TF-FVII induces angiogenesis via clotting-dependent and -independent mechanisms. Clottingdependent angiogenesis is induced via the generation of thrombin and cross-linked fibrin network, which facilitate vessel infiltration and also act as a reservoir for endothelial cells (ECs) growth factors. Expression of TF in tumor cells and ECs triggers clotting-independent angiogenesis through induction of VEGF, urokinase-type plasminogen activator (uPAR), early growth response 1 (EGR1), IL8, and cysteine-rich angiogenic inducer 61 (Cyr61).
Collapse
Affiliation(s)
- Zahra Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
29
|
Udovenko A, Makogonenko Y, Korolova D, Druzhyna N, Chernyshenko V, Komisarenko S. Formation and elimination of soluble fibrin and D-dimer in the bloodstream. Croat Med J 2023; 64:421-429. [PMID: 38168523 PMCID: PMC10797241 DOI: 10.3325/cmj.2023.64.421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/06/2023] [Indexed: 04/20/2025] Open
Abstract
Soluble fibrin is composed mainly of desA fibrin and fibrinogen oligomers consisting of fewer than 16 monomers partially cross-linked by factor XIIIa. Soluble fibrin cannot stimulate Glu-plasminogen activation by tissue plasminogen activator (t-PA); therefore, it may not be a direct predecessor of D-dimer. However, within the microcirculatory system, soluble fibrin oligomers may form microclots. Fibrin microclots stimulate Glu-plasminogen activation by t-PA, a process resulting in the formation of Glu-plasmin. Glu-plasmin dissolves the microclots, forming D-dimer. In normal and pathological blood plasma samples, soluble fibrin levels are substantially higher than those of D-dimer. Their concentrations in the plasma are also regulated by transendothelial transfer, absorption by blood macrophages, and binding and internalization with low-density lipoprotein receptors of the cells of the reticuloendothelial system. Therefore, the exact mechanisms of fibrin clots formation and elimination in normal and pathological conditions remain unclear. In this study, we reviewed findings on the molecular mechanisms of the formation and dissolution of fibrin clots, fibrin-dependent activation of Glu-plasminogen by t-PA, and blood plasma behavior in the microcirculatory system. Finally, we proposed a model that explains the relations of D-dimer and soluble fibrin underlying the common and separate mechanisms of their formation and elimination.
Collapse
|
30
|
Cartland SP, Stanley CP, Bursill C, Passam F, Figtree GA, Patel S, Loa J, Golledge J, Robinson DA, Aitken SJ, Kavurma MM. Sex, Endothelial Cell Functions, and Peripheral Artery Disease. Int J Mol Sci 2023; 24:17439. [PMID: 38139267 PMCID: PMC10744086 DOI: 10.3390/ijms242417439] [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: 10/03/2023] [Revised: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
Peripheral artery disease (PAD) is caused by blocked arteries due to atherosclerosis and/or thrombosis which reduce blood flow to the lower limbs. It results in major morbidity, including ischemic limb, claudication, and amputation, with patients also suffering a heightened risk of heart attack, stroke, and death. Recent studies suggest women have a higher prevalence of PAD than men, and with worse outcomes after intervention. In addition to a potential unconscious bias faced by women with PAD in the health system, with underdiagnosis, and lower rates of guideline-based therapy, fundamental biological differences between men and women may be important. In this review, we highlight sexual dimorphisms in endothelial cell functions and how they may impact PAD pathophysiology in women. Understanding sex-specific mechanisms in PAD is essential for the development of new therapies and personalized care for patients with PAD.
Collapse
Affiliation(s)
- Siân P. Cartland
- Heart Research Institute, The University of Sydney, Sydney, NSW 2042, Australia; (S.P.C.); (C.P.S.); (S.P.)
| | - Christopher P. Stanley
- Heart Research Institute, The University of Sydney, Sydney, NSW 2042, Australia; (S.P.C.); (C.P.S.); (S.P.)
| | - Christina Bursill
- South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia;
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Freda Passam
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2050, Australia; (F.P.); (G.A.F.); (S.J.A.)
| | - Gemma A. Figtree
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2050, Australia; (F.P.); (G.A.F.); (S.J.A.)
- Kolling Institute of Medical Research, Sydney, NSW 2064, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, NSW 2042, Australia; (S.P.C.); (C.P.S.); (S.P.)
- Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia (D.A.R.)
| | - Jacky Loa
- Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia (D.A.R.)
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia;
- Department of Vascular and Endovascular Surgery, The Townsville University Hospital, Townsville, QLD 4814, Australia
| | | | - Sarah J. Aitken
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2050, Australia; (F.P.); (G.A.F.); (S.J.A.)
- Concord Institute of Academic Surgery, Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Mary M. Kavurma
- Heart Research Institute, The University of Sydney, Sydney, NSW 2042, Australia; (S.P.C.); (C.P.S.); (S.P.)
| |
Collapse
|
31
|
Harasawa A, Ishiyama S, Mochizuki K. Fructo-oligosaccharide-mediated alteration in claudin expression in small intestinal absorptive Caco-2 cells is positively associated with the induction of inflammatory genes and the glucan receptor gene CLEC7A. Nutrition 2023; 115:112140. [PMID: 37481839 DOI: 10.1016/j.nut.2023.112140] [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: 03/24/2023] [Revised: 06/02/2023] [Accepted: 06/18/2023] [Indexed: 07/25/2023]
Abstract
OBJECTIVES Indigestible carbohydrates may strengthen tight junctions (TJs) independently of intestinal bacteria. This study investigated whether indigestible carbohydrates (i.e., fructo-oligosaccharides [FOS]) promote TJs directly to intestinal absorptive Caco-2 cells and examined the association between the expression of genes constructing TJs and other genes using mRNA microarray analysis. METHODS Caco-2 cells at 1.0 × 105/mL were seeded in a type I collagen plate and cultured in high-glucose Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal calf serum (FCS); the cells reached confluence at 7 d after seeding. Ten days after the cells reached confluency, they were cultured for 24 h in 10% FCS-containing DMEM medium supplemented with 0%, 5%, or 10% FOS. We performed mRNA microarray to identify the genes whose expression was altered by FOS. Subsequently, quantitative reverse transcription polymerase chain reaction was performed for these altered genes, including CLEC7A encoding the glucan receptor, and for the claudin (CLDN) family genes. The expression of CLDN2, CLDN4, and CLEC7A proteins was assessed using western blot analysis. RESULTS FOS decreased the mRNA and protein expression of CLDN2, which weakens TJs, and increased those of CLDN4, which strengthens TJs, in Caco-2 cells. FOS treatment (10%) reduced the mRNA expression of antioxidative genes and induced the expression of immune response-related genes, including CLEC7A, CCL2, and ITGA2. Furthermore, the expression of CLEC7A protein was enhanced by FOS. CONCLUSIONS Induction of TJ-strengthening CLDN4 and reduction of TJ-weakening CLDN2 by FOS treatment in small intestinal absorptive Caco-2 cells is positively associated with the induction of inflammatory genes, including CLEC7A.
Collapse
Affiliation(s)
- Aya Harasawa
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Japan
| | - Shiori Ishiyama
- Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Japan
| | - Kazuki Mochizuki
- Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Japan.
| |
Collapse
|
32
|
Dehnavi S, Sadeghi M, Tavakol Afshari J, Mohammadi M. Interactions of mesenchymal stromal/stem cells and immune cells following MSC-based therapeutic approaches in rheumatoid arthritis. Cell Immunol 2023; 393-394:104771. [PMID: 37783061 DOI: 10.1016/j.cellimm.2023.104771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
Rheumatoid arthritis (RA) is considered to be a degenerative and progressive autoimmune disorder. Although several medicinal regimens are used to treat RA, potential adverse events such as metabolic disorders and increased risk of infection, as well as drug resistance in some patients, make it essential to find an effective and safe therapeutic approach. Mesenchymal stromal/stem cells (MSCs) are a group of non-hematopoietic stromal cells with immunomodulatory and inhibitory potential. These cells exert their regulatory properties through direct cell-to-cell interactions and paracrine effects on various immune and non-immune cells. As conventional therapeutic approaches for RA are limited due to their side effects, and some patients became refractory to the treatment, MSCs are considered as a promising alternative treatment for RA. In this review, we introduced various experimental and clinical studies conducted to evaluate the therapeutic effects of MSCs on animal models of arthritis and RA patients. Then, possible modulatory and suppressive effects of MSCs on different innate and adaptive immune cells, including dendritic cells, neutrophils, macrophages, natural killer cells, B lymphocytes, and various subtypes of T cells, were categorized and summarized. Finally, limitations and future considerations for the efficient application of MSCs as a therapeutic approach in RA patients were presented.
Collapse
Affiliation(s)
- Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Mojgan Mohammadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
33
|
He S, He X, Pan S, Jiang W. Exploring the Mechanism of Chuanxiong Rhizoma against Thrombosis Based on Network Pharmacology, Molecular Docking and Experimental Verification. Molecules 2023; 28:6702. [PMID: 37764479 PMCID: PMC10535320 DOI: 10.3390/molecules28186702] [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: 08/29/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Chuanxiong rhizoma (CX) has been utilized for centuries as a traditional herb to treat blood stasis syndromes. However, the pharmacological mechanisms are still not completely revealed. This research was aimed at exploring the molecular mechanisms of CX treatment for thrombosis. Network pharmacology was used to predict the potential anti-thrombosis mechanism after correlating the targets of active components with targets of thrombosis. Furthermore, we verified the mechanism of using CX to treat thrombosis via molecular docking and in vitro experiments. Network pharmacology results showed that a total of 18 active ingredients and 65 targets of CX treatment for thrombosis were collected, including 8 core compounds and 6 core targets. We revealed for the first time that tissue factor (TF) had a close relationship with most core targets of CX in the treatment of thrombosis. TF is a primary coagulation factor in physiological hemostasis and pathological thrombosis. Furthermore, core components of CX have strong affinity for core targets and TF according to molecular docking analysis. The in vitro experiments indicated that Ligustilide (LIG), the representative component of CX, could inhibit TF procoagulant activity, TF mRNA and protein over-expression in a dose-dependent manner in EA.hy926 cells through the PI3K/Akt/NF-κB signaling pathway. This work demonstrated that hemostasis or blood coagulation was one of the important biological processes in the treatment of thrombosis with CX, and TF also might be a central target of CX when used for treating thrombosis. The inhibition of TF might be a novel mechanism of CX in the treatment of thrombosis.
Collapse
Affiliation(s)
- Shasha He
- School of Pharmacy, Guizhou University, Guiyang 550025, China; (S.H.); (X.H.); (S.P.)
| | - Xuhua He
- School of Pharmacy, Guizhou University, Guiyang 550025, China; (S.H.); (X.H.); (S.P.)
| | - Shujuan Pan
- School of Pharmacy, Guizhou University, Guiyang 550025, China; (S.H.); (X.H.); (S.P.)
- Engineering Research Center of the Utilization for Characteristic Bio-Pharmaceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Wenwen Jiang
- School of Pharmacy, Guizhou University, Guiyang 550025, China; (S.H.); (X.H.); (S.P.)
| |
Collapse
|
34
|
Jing-Lun Z, Shuang C, Li-Mei Z, Xiao-Dong L. YKL-40 promotes chemokine expression following drug-induced liver injury via TF-PAR1 pathway in mice. Front Pharmacol 2023; 14:1205062. [PMID: 37693903 PMCID: PMC10484592 DOI: 10.3389/fphar.2023.1205062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Background: The inflammatory factor YKL-40 is associated with various inflammatory diseases and is key to remodeling inflammatory cells and tissues. YKL-40 (Chi3l1) promotes the activation of tissue factor (TF), leading to intrahepatic vascular coagulation (IAOC) and liver injury. TF is a key promoter of the exogenous coagulation cascade and is also involved in several signaling involving cell proliferation, apoptosis, charring, migration and inflammatory diseases pathways. However, the effect of YKL-40-induced TF-PAR1 pathway on the expression of downstream chemokines remains unknown. Methods: We established a liver injury model using Concanavalin A (ConA) in C57 BL/6 mice. By adopting various experimental techniques, the effect of YKL-40 induced TF-PAR1 pathway on the expression of downstream chemokine ligand 2 (CCL2) and IP-10 was verified. Results: We found that overexpression of YKL-40 increased the expression of TF, protease-activated receptor 1 (PAR1), CCL2 and IP-10 in mice and exacerbated the severity of liver injury. However, blocking the expression of TF significantly reversed the extent of liver injury. Conclusion: We found that YKL-40 promotes the expression of downstream chemokines ligand 2 (CCL2) and IP-10 by activating the TF-PAR1 pathway, leading to increased recruitment of inflammatory cells and exacerbating the progression of liver injury. This provides a new approach for the clinical treatment of drug-induced liver injury.
Collapse
Affiliation(s)
- Zhan Jing-Lun
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyan, China
- Department of the Second Clinical Pharmacy, School of Pharmacy, China Medical University, Shenyang, China
| | - Chai Shuang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyan, China
- Department of the Second Clinical Pharmacy, School of Pharmacy, China Medical University, Shenyang, China
| | - Zhao Li-Mei
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyan, China
- Department of the Second Clinical Pharmacy, School of Pharmacy, China Medical University, Shenyang, China
| | - Liu Xiao-Dong
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyan, China
- Department of the Second Clinical Pharmacy, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
35
|
Costa CHN, Chang KP, Costa DL, Cunha FVM. From Infection to Death: An Overview of the Pathogenesis of Visceral Leishmaniasis. Pathogens 2023; 12:969. [PMID: 37513817 PMCID: PMC10384967 DOI: 10.3390/pathogens12070969] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/02/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Kala-azar, also known as visceral leishmaniasis (VL), is a disease caused by Leishmania infantum and L. donovani. Patients experience symptoms such as fever, weight loss, paleness, and enlarged liver and spleen. The disease also affects immunosuppressed individuals and has an overall mortality rate of up to 10%. This overview explores the literature on the pathogenesis of preclinical and clinical stages, including studies in vitro and in animal models, as well as complications and death. Asymptomatic infection can result in long-lasting immunity. VL develops in a minority of infected individuals when parasites overcome host defenses and multiply in tissues such as the spleen, liver, and bone marrow. Hepatosplenomegaly occurs due to hyperplasia, resulting from parasite proliferation. A systemic inflammation mediated by cytokines develops, triggering acute phase reactants from the liver. These cytokines can reach the brain, causing fever, cachexia and vomiting. Similar to sepsis, disseminated intravascular coagulation (DIC) occurs due to tissue factor overexpression. Anemia, hypergammaglobulinemia, and edema result from the acute phase response. A regulatory response and lymphocyte depletion increase the risk of bacterial superinfections, which, combined with DIC, are thought to cause death. Our understanding of VL's pathogenesis is limited, and further research is needed to elucidate the preclinical events and clinical manifestations in humans.
Collapse
Affiliation(s)
- Carlos H N Costa
- Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151-Sul, Teresina 64002-510, PI, Brazil
| | - Kwang-Poo Chang
- Department of Microbiology/Immunology, Center for Cancer Cell Biology, Immunology & Infection, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Dorcas L Costa
- Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151-Sul, Teresina 64002-510, PI, Brazil
| | - Francisco Valmor M Cunha
- Departament of Physiotherapy, Centro Universitário Uninovafapi, Rua Vitorino Orthiges Fernandes, 6123-Uruguai, Teresina 64073-505, PI, Brazil
| |
Collapse
|
36
|
Danckwardt S, Trégouët DA, Castoldi E. Post-transcriptional control of haemostatic genes: mechanisms and emerging therapeutic concepts in thrombo-inflammatory disorders. Cardiovasc Res 2023; 119:1624-1640. [PMID: 36943786 PMCID: PMC10325701 DOI: 10.1093/cvr/cvad046] [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: 09/26/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 03/23/2023] Open
Abstract
The haemostatic system is pivotal to maintaining vascular integrity. Multiple components involved in blood coagulation have central functions in inflammation and immunity. A derailed haemostasis is common in prevalent pathologies such as sepsis, cardiovascular disorders, and lately, COVID-19. Physiological mechanisms limit the deleterious consequences of a hyperactivated haemostatic system through adaptive changes in gene expression. While this is mainly regulated at the level of transcription, co- and posttranscriptional mechanisms are increasingly perceived as central hubs governing multiple facets of the haemostatic system. This layer of regulation modulates the biogenesis of haemostatic components, for example in situations of increased turnover and demand. However, they can also be 'hijacked' in disease processes, thereby perpetuating and even causally entertaining associated pathologies. This review summarizes examples and emerging concepts that illustrate the importance of posttranscriptional mechanisms in haemostatic control and crosstalk with the immune system. It also discusses how such regulatory principles can be used to usher in new therapeutic concepts to combat global medical threats such as sepsis or cardiovascular disorders.
Collapse
Affiliation(s)
- Sven Danckwardt
- Centre for Thrombosis and Hemostasis (CTH), University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Centre for Cardiovascular Research (DZHK),
Berlin, Germany
- Posttranscriptional Gene Regulation, University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University
Medical Centre Mainz, Langenbeckstr. 1, 55131
Mainz, Germany
- Center for Healthy Aging (CHA), Mainz,
Germany
| | - David-Alexandre Trégouët
- INSERM, Bordeaux Population Health Research Center, UMR 1219, Department of
Molecular Epidemiology of Vascular and Brain Disorders (ELEANOR), University of
Bordeaux, Bordeaux, France
| | - Elisabetta Castoldi
- Department of Biochemistry, Cardiovascular Research Institute Maastricht
(CARIM), Maastricht University, Universiteitsingel 50, 6229
ER Maastricht, The Netherlands
| |
Collapse
|
37
|
Ghanbari EP, Jakobs K, Puccini M, Reinshagen L, Friebel J, Haghikia A, Kränkel N, Landmesser U, Rauch-Kröhnert U. The Role of NETosis and Complement Activation in COVID-19-Associated Coagulopathies. Biomedicines 2023; 11:biomedicines11051371. [PMID: 37239041 DOI: 10.3390/biomedicines11051371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Inflammation-induced coagulopathy is a common complication associated with coronavirus disease 2019 (COVID-19). We aim to evaluate the association of NETosis and complement markers with each other as well as their association with thrombogenicity and disease severity in COVID-19. The study included hospitalized patients with an acute respiratory infection: patients with SARS-CoV2 infection (COVpos, n = 47) or either pneumonia or infection-triggered acute exacerbated COPD (COVneg, n = 36). Our results show that NETosis, coagulation, and platelets, as well as complement markers, were significantly increased in COVpos patients, especially in severely ill COVpos patients. NETosis marker MPO/DNA complexes correlated with coagulation, platelet, and complement markers only in COVpos. Severely ill COVpos patients showed an association between complement C3 and SOFA (R = 0.48; p ≤ 0.028), C5 and SOFA (R = 0.46; p ≤ 0.038), and C5b-9 and SOFA (R = 0.44; p ≤ 0.046). This study provides further evidence that NETosis and the complement system are key players in COVID-19 inflammation and clinical severity. Unlike previous studies that found NETosis and complement markers to be elevated in COVID-19 patients compared to healthy controls, our findings show that this characteristic distinguishes COVID-19 from other pulmonary infectious diseases. Based on our results, we propose that COVID-19 patients at high risk for immunothrombosis could be identified via elevated complement markers such as C5.
Collapse
Affiliation(s)
- Emily Parissa Ghanbari
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Kai Jakobs
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Marianna Puccini
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Leander Reinshagen
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Julian Friebel
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Arash Haghikia
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicolle Kränkel
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ursula Rauch-Kröhnert
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| |
Collapse
|
38
|
Iannucci J, Grammas P. Thrombin, a Key Driver of Pathological Inflammation in the Brain. Cells 2023; 12:cells12091222. [PMID: 37174621 PMCID: PMC10177239 DOI: 10.3390/cells12091222] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/21/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), are major contributors to death and disability worldwide. A multitude of evidence suggests that neuroinflammation is critical in neurodegenerative disease processes. Exploring the key mediators of neuroinflammation in AD, a prototypical neurodegenerative disease, could help identify pathologic inflammatory mediators and mechanisms in other neurodegenerative diseases. Elevated levels of the multifunctional inflammatory protein thrombin are commonly found in conditions that increase AD risk, including diabetes, atherosclerosis, and traumatic brain injury. Thrombin, a main driver of the coagulation cascade, has been identified as important to pathological events in AD and other neurodegenerative diseases. Furthermore, recent evidence suggests that coagulation cascade-associated proteins act as drivers of inflammation in the AD brain, and studies in both human populations and animal models support the view that abnormalities in thrombin generation promote AD pathology. Thrombin drives neuroinflammation through its pro-inflammatory activation of microglia, astrocytes, and endothelial cells. Due to the wide-ranging pro-inflammatory effects of thrombin in the brain, inhibiting thrombin could be an effective strategy for interrupting the inflammatory cascade which contributes to neurodegenerative disease progression and, as such, may be a potential therapeutic target for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | | |
Collapse
|
39
|
Yan S, Campos de Souza S, Xie Z, Bao Y. Research progress in clinical trials of stem cell therapy for stroke and neurodegenerative diseases. IBRAIN 2023; 9:214-230. [PMID: 37786546 PMCID: PMC10529019 DOI: 10.1002/ibra.12095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 10/04/2023]
Abstract
The incidence of stroke and neurodegenerative diseases is gradually increasing in modern society, but there is still no treatment that is effective enough. Stem cells are cells that can reproduce (self-renew) and differentiate into the body, which have shown significance in basic research, while doctors have also taken them into clinical trials to determine their efficacy and safety. Existing clinical trials mainly include middle-aged and elderly patients with stroke or Parkinson's disease (mostly 40-80 years old), mainly involving injection of mesenchymal stem cells and bone marrow mesenchymal stem cells through the veins and the putamen, with a dosage of mostly 106-108 cells. The neural and motor functions of the patients were restored after stem cell therapy, and the safety was found to be good during the follow-up period of 3 months to 5 years. Here, we review all clinical trials and the latest advances in stroke, Alzheimer's disease, and Parkinson's disease, with the hope that stem cell therapy will be used in the clinic in the future to achieve effective treatment rates and benefit patients.
Collapse
Affiliation(s)
- Shan‐Shan Yan
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Senio Campos de Souza
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
| | - Zhen‐Dong Xie
- Institute for Bioengineering of CataloniaUniversity of BarcelonaCarrer de Baldiri ReixacBarcelonaSpain
| | - Yong‐Xin Bao
- Qingdao Women and Children's HospitalQingdao UniversityQingdaoChina
| |
Collapse
|
40
|
Zhu C, Liang Y, Luo Y, Ma X. Role of pyroptosis in hemostasis activation in sepsis. Front Immunol 2023; 14:1114917. [PMID: 36756123 PMCID: PMC9899792 DOI: 10.3389/fimmu.2023.1114917] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/10/2023] [Indexed: 01/24/2023] Open
Abstract
Sepsis is frequently associated with hemostasis activation and thrombus formation, and systematic hemostatic changes are associated with a higher risk of mortality. The key events underlying hemostasis activation during sepsis are the strong activation of innate immune pathways and the excessive inflammatory response triggered by invading pathogens. Pyroptosis is a proinflammatory form of programmed cell death, that defends against pathogens during sepsis. However, excessive pyroptosis can lead to a dysregulation of host immune responses and organ dysfunction. Recently, pyroptosis has been demonstrated to play a prominent role in hemostasis activation in sepsis. Several studies have demonstrated that pyroptosis participates in the release and coagulation activity of tissue factors. In addition, pyroptosis activates leukocytes, endothelial cells, platelets, which cooperate with the coagulation cascade, leading to hemostasis activation in sepsis. This review article attempts to interpret the molecular and cellular mechanisms of the hemostatic imbalance induced by pyroptosis during sepsis and discusses potential therapeutic strategies.
Collapse
Affiliation(s)
- Chengrui Zhu
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yingjian Liang
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yangtuo Luo
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning, China,*Correspondence: Yangtuo Luo, ; Xiaochun Ma,
| | - Xiaochun Ma
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China,*Correspondence: Yangtuo Luo, ; Xiaochun Ma,
| |
Collapse
|
41
|
CD14 +-Monocytes Exposed to Apolipoprotein CIII Express Tissue Factor. Int J Mol Sci 2023; 24:ijms24032223. [PMID: 36768547 PMCID: PMC9916694 DOI: 10.3390/ijms24032223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Apolipoprotein CIII (ApoCIII) represents a key regulator of plasma lipid metabolism and a recognized risk factor for atherosclerosis and cardiovascular diseases. Beyond the regulation of lipoprotein trafficking, ApoCIII is also involved in endothelial dysfunction and monocyte recruitment related to atherothrombosis. With tissue factor (TF) being the primary initiator of the blood coagulation cascade, we hypothesized that ApoCIII-treated monocytes could express it. Hence, human CD14+-monocytes and autologous neutrophils were incubated with ApoCIII and sera from human subjects containing previously measured ApoCIII amounts. By RT-qPCR and ELISA, CD14+-monocytes, but not neutrophils, were found to show increased mRNA expression and production of TNFα, IL-1β and IL-6 as well as TF mRNA once exposed to ultra-purified ApoCIII. By flow cytometry, CD14+-monocytes were found to rapidly express TF on their cell surface membrane when incubated with either ApoCIII or sera with known concentrations of ApoCIII. Finally, preincubation with specific ApoCIII-neutralizing antibodies significantly reduced the ability of most sera with known concentrations of ApoCIII to upregulate TF protein, other than partially inhibiting cytokine release, in CD14+-monocytes. In sum, herein we demonstrate that ApoCIII activates CD14+-monocytes to express TF. The data identify a potential mechanism which links circulating apolipoproteins with inflammation and atherothrombosis-related processes underlying cardiovascular risk.
Collapse
|
42
|
Takahashi K. Microglial heterogeneity in amyotrophic lateral sclerosis. J Neuropathol Exp Neurol 2023; 82:140-149. [PMID: 36440536 DOI: 10.1093/jnen/nlac110] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an intractable neurodegenerative disease of the central nervous system that is pathologically characterized by motor neuron loss. Although the cause of the disease is still unknown, its pathophysiology is considered heterogeneous. In recent years, there have been a series of reports on the existence of disease-associated microglia (DAM) in the lesions of various neurodegenerative diseases. DAM have also been reported in SOD1-deficient mice, a disease model of ALS. However, the role of DAM in sporadic ALS remains unclear. This study revealed that spinal cord lesions in ALS can be pathologically distinguished into 2 subgroups (TMEM119+ and TMEM119- microglia) according to the type of microglia. Expression of the microglial activation marker CD68 and endothelial activation were also observed in the TMEM119+ microglia group, suggesting the presence of inflammatory processes in ALS lesions. Since DAM suppress the expression of TMEM119, the TMEM119+ microglia group may indicate DAM-independent inflammatory neurodegeneration. These results may explain why, in some clinical trials of anti-inflammatory drugs for ALS, only some cases showed suppression of disease progression.
Collapse
Affiliation(s)
- Kazuya Takahashi
- Department of Medicine, National Hospital Organization Iou National Hospital, Kanazawa, Japan
| |
Collapse
|
43
|
Intestinal Barrier Dysfunction and Microbial Translocation in Patients with First-Diagnosed Atrial Fibrillation. Biomedicines 2023; 11:biomedicines11010176. [PMID: 36672684 PMCID: PMC9856173 DOI: 10.3390/biomedicines11010176] [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: 12/13/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND According to the leaky gut concept, microbial products (e.g., lipopolysaccharide, LPS) enter the circulation and mediate pro-inflammatory immunological responses. Higher plasma LPS levels have been reported in patients with various cardiovascular diseases, but not specifically during early atrial fibrillation (AF). METHODS We studied data and blood samples from patients presenting with first-diagnosed AF (FDAF) (n = 80) and 20 controls. RESULTS Circulating biomarkers that are suggestive of mucosal inflammation (zonulin, mucosal adhesion molecule MAdCAM-1) and intestinal epithelium damage (intestinal fatty acid binding protein, IFABP) were increased in the plasma of patients with FDAF when compared to patients with chronic cardiovascular diseases but without AF. Surrogate plasma markers of increased intestinal permeability (LPS, CD14, LPS-binding protein, gut-derived LPS-neutralising IgA antibodies, EndoCAbs) were detected during early AF. A reduced ratio of IgG/IgM EndoCAbs titres indicated chronic endotoxaemia. Collagen turnover biomarkers, which corresponded to the LPS values, suggested an association of gut-derived low-grade endotoxaemia with adverse structural remodelling. The LPS concentrations were higher in FDAF patients who experienced a major adverse cardiovascular event. CONCLUSIONS Intestinal barrier dysfunction and microbial translocation accompany FDAF. Improving gut permeability and low-grade endotoxaemia might be a potential therapeutic approach to reducing the disease progression and cardiovascular complications in FDAF.
Collapse
|
44
|
Lu Y, Li D, Huang Y, Sun Y, Zhou H, Ye F, Yang H, Xu T, Quan S, Pan J. Pretreatment with Eupatilin Attenuates Inflammation and Coagulation in Sepsis by Suppressing JAK2/STAT3 Signaling Pathway. J Inflamm Res 2023; 16:1027-1042. [PMID: 36926276 PMCID: PMC10013575 DOI: 10.2147/jir.s393850] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Purpose Sepsis is an aggressive and life-threatening organ dysfunction induced by infection. Excessive inflammation and coagulation contribute to the negative outcomes for sepsis, resulting in high morbidity and mortality. In this study, we explored whether Eupatilin could alleviate lung injury, reduce inflammation and coagulation during sepsis. Methods We constructed an in vitro sepsis model by stimulating RAW264.7 cells with 1 μg/mL lipopolysaccharide (LPS) for 6 hours. The cells were divided into control group, LPS group, LPS+ Eupatilin (Eup) group, and Eup group to detect their cell activity and inflammatory cytokines and coagulation factor levels. Cells in LPS+Eup and Eup group were pretreated with Eupatilin (10μM) for 2 hours. In vivo, mice were divided into sham operation group, cecal ligation and puncture (CLP) group and Eup group. Mice in the CLP and Eup groups were pretreated with Eupatilin (10mg/kg) for 2 hours by gavage. Lung tissue and plasma were collected and inflammatory cytokines, coagulation factors and signaling were measured. Results In vitro, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and tissue factor (TF) expression in LPS-stimulated RAW264.7 cells was downregulated by Eupatilin (10μM). Furthermore, Eupatilin inhibited phosphorylation of the JAK2/STAT3 signaling pathway and suppressed p-STAT3 nuclear translocation. In vivo, Eupatilin increased the survival rate of the mice. In septic mice, plasma concentrations of TNF-α, IL-1β and IL-6, as well as TF, plasminogen activator inhibitor 1 (PAI-1), D-dimer, thrombin-antithrombin complex (TAT) and fibrinogen were improved by Eupatilin. Moreover, Eupatilin alleviated lung injury by improving the expression of inflammatory cytokines and TF, fibrin deposition and macrophage infiltration in lung tissue. Conclusion Our results revealed that Eupatilin may modulate inflammation and coagulation indicators as well as improve lung injury in sepsis via the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yilun Lu
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China
| | - Ding Li
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China
| | - Yueyue Huang
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China
| | - Yuanyuan Sun
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China
| | - Hongmin Zhou
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China
| | - Fanrong Ye
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China
| | - Hongjing Yang
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China
| | - Tingting Xu
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China
| | - Shichao Quan
- Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China.,Collaborative Innovation Center for Intelligence Medical Education, Wenzhou, People's Republic of China.,Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, People's Republic of China.,Department of General Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jingye Pan
- Department of Intensive Care Unit, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, People's Republic of China.,Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, People's Republic of China.,Collaborative Innovation Center for Intelligence Medical Education, Wenzhou, People's Republic of China.,Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, People's Republic of China
| |
Collapse
|
45
|
Myllylahti L, Ropponen J, Lax M, Lassila R, Nykänen AI. Upregulation of Coagulation Factor VIII and Fibrinogen After Pulmonary Endarterectomy in Patients with Chronic Thromboembolic Pulmonary Hypertension. Clin Appl Thromb Hemost 2023; 29:10760296231158369. [PMID: 36890726 PMCID: PMC9998419 DOI: 10.1177/10760296231158369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023] Open
Abstract
OBJECTIVES Chronic thromboembolic pulmonary hypertension (CTEPH) is associated with thrombotic states including elevated coagulation factor VIII (FVIII). Pulmonary endarterectomy (PEA) is the main treatment for CTEPH, and efficient anticoagulation is essential to prevent thromboembolism recurrence after surgery. We aimed to characterize longitudinal changes in FVIII and other coagulation biomarkers after PEA. METHODS Coagulation biomarker levels were measured at baseline and up to 12 months after operation in 17 consecutive patients with PEA. Temporal patterns of coagulation biomarkers, and correlation of FVIII with other coagulation biomarkers, were analyzed. RESULTS Baseline FVIII levels were elevated in 71% of the patients (mean 216 ± 67 IU/dl). FVIII doubled 7 days after PEA, peaking at 471 ± 87 IU/dl, and gradually returned to respective baseline levels within 3 months. Postoperative fibrinogen levels were also elevated. Antithrombin decreased at 1 to 3 days, D-dimer increased at 1 to 4 weeks, and thrombocytosis was observed at 2 weeks. CONCLUSIONS FVIII is elevated in most patients with CTEPH. After PEA, early but transient elevation of FVIII and fibrinogen, and delayed reactive thrombocytosis, occurs, and warrants careful postoperative anticoagulation to prevent thromboembolism recurrence.
Collapse
Affiliation(s)
- Lasse Myllylahti
- Division of Internal Medicine, Department of Internal Medicine and Rehabilitation, Helsinki University Hospital, Helsinki, Finland
| | - Jussi Ropponen
- Department of Cardiac Surgery, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mikko Lax
- Division of Anesthesiology, Department of Anesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Riitta Lassila
- Unit of Coagulation Disorders, Department of Hematology, Comprehensive Cancer Center, Helsinki University Hospital, and Research Program Unit in Systems Oncology, 3835University of Helsinki, Helsinki, Finland
| | - Antti I Nykänen
- Department of Cardiac Surgery, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
46
|
Gawaz A, Gonser L, Strölin A, Kofler L, Häfner HM, Kofler K. Concomitant thrombosis in patients with cellulitis as incidental finding. Clin Hemorheol Microcirc 2022; 82:335-340. [PMID: 35938241 DOI: 10.3233/ch-221519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Although inflammation and thrombosis are tightly connected, only 45% of patients with lower leg cellulitis receive anticoagulant therapy. Available data about the prevalence of concomitant deep venous thrombosis (DVT) in patients with cellulitis of the lower extremity is scarce and general guidelines regarding diagnosis and prevention of venous thromboembolism are missing. OBJECTIVE We sought to determine how frequently DVT occurs as an incidental finding in patients with cellulitis and to provide recommendations for diagnostics and anticoagulant therapy. METHODS Patients' records were analysed and 192 consecutive patients with cellulitis were included in this study. The prevalence of concomitant DVT was examined by duplex ultrasound, as well as comorbidities and risk factors. RESULTS We detected thrombosis in 12.0% of the patients with lower leg cellulitis, of which 43.5% were located in a proximal vein and 52.2% in the veins of the calf. CONCLUSIONS Our results clearly indicate that cellulitis is not only a differential diagnosis, but should be considered a risk factor for venous thrombosis. Therefore, prophylactic anticoagulation should be considered in patients suffering from cellulitis and a systematic screening for venous thrombosis in patients with cellulitis should be performed.
Collapse
Affiliation(s)
- Andrea Gawaz
- Universitätshautklinik Tübingen, Liebermeisterstr, Tübingen, Germany
| | - Lena Gonser
- Universitätshautklinik Tübingen, Liebermeisterstr, Tübingen, Germany
| | - Anke Strölin
- Universitätshautklinik Tübingen, Liebermeisterstr, Tübingen, Germany
| | - Lukas Kofler
- Universitätshautklinik Tübingen, Liebermeisterstr, Tübingen, Germany
| | | | - Katrin Kofler
- Universitätshautklinik Tübingen, Liebermeisterstr, Tübingen, Germany
| |
Collapse
|
47
|
Silva AVBDA, Campanati JDEAG, Barcelos IDES, Santos ACL, Deus UPDE, Soares TDEJ, Amaral LSDEB. COVID-19 and Acute Kidney Injury - Direct and Indirect Pathophysiological Mechanisms Underlying Lesion Development. AN ACAD BRAS CIENC 2022; 94:e20211501. [PMID: 36477239 DOI: 10.1590/0001-3765202220211501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/08/2022] [Indexed: 11/29/2022] Open
Abstract
COVID-19 is a pandemic disease caused by the SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) responsible for millions of deaths worldwide. Although the respiratory system is the main target of COVID-19, the disease can affect other organs, including the kidneys. Acute Kidney Injury (AKI), commonly seen in patients infected with COVID-19, has a multifactorial cause. Several studies associate this injury with the direct involvement of the virus in renal cells and the indirect damage stimulated by the infection. The direct cytopathic effects of SARS-CoV-2 are due to the entry and replication of the virus in renal cells, changing several regulatory pathways, especially the renin-angiotensin-aldosterone system (RAAS), with repercussions on the kallikrein-kinin system (KKS). Furthermore, the virus can deregulate the immune system, leading to an exaggerated response of inflammatory cells, characterizing the state of hypercytokinemia. The such exaggerated inflammatory response is commonly associated with hemodynamic changes, reduced renal perfusion, tissue hypoxia, generation of reactive oxygen species (ROS), endothelial damage, and coagulopathies, which can result in severe damage to the renal parenchyma. Thereby, understanding the molecular mechanisms and pathophysiology of kidney injuries induced by SARS-COV-2 is of fundamental importance to obtaining new therapeutic insights for the prevention and management of AKI.
Collapse
Affiliation(s)
- Antônio V B DA Silva
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - João DE A G Campanati
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Isadora DE S Barcelos
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Alberto C L Santos
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Uildson P DE Deus
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Telma DE J Soares
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Liliany S DE B Amaral
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| |
Collapse
|
48
|
Oe Y, Takahashi N. Tissue Factor, Thrombosis, and Chronic Kidney Disease. Biomedicines 2022; 10:2737. [PMID: 36359257 PMCID: PMC9687479 DOI: 10.3390/biomedicines10112737] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/14/2023] Open
Abstract
Coagulation abnormalities are common in chronic kidney disease (CKD). Tissue factor (TF, factor III) is a master regulator of the extrinsic coagulation system, activating downstream coagulation proteases, such as factor Xa and thrombin, and promoting fibrin formation. TF and coagulation proteases also activate protease-activated receptors (PARs) and are implicated in various organ injuries. Recent studies have shown the mechanisms by which thrombotic tendency is increased under CKD-specific conditions. Uremic toxins, such as indoxyl sulfate and kynurenine, are accumulated in CKD and activate TF and coagulation; in addition, the TF-coagulation protease-PAR pathway enhances inflammation and fibrosis, thereby exacerbating renal injury. Herein, we review the recent research studies to understand the role of TF in increasing the thrombotic risk and CKD progression.
Collapse
Affiliation(s)
- Yuji Oe
- Division of Nephrology, Rheumatology, and Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA 92161, USA
- VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Nobuyuki Takahashi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Sendai 980-8578, Japan
| |
Collapse
|
49
|
Chen Y, Liu J, Su Y, Zhao H, Zhao Y, Wen M, Lu S, Cao X, Zhang W, Liu L, Wu J. Annexin V - and tissue factor + microparticles as biomarkers for predicting deep vein thrombosis in patients after joint arthroplasty. Clin Chim Acta 2022; 536:169-179. [PMID: 36191610 DOI: 10.1016/j.cca.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/29/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Venous thromboembolism (VTE) is a common and severe complication of joint arthroplasty. Microparticles (MPs) containing phosphatidylserine (PS) and tissue factor (TF) can trigger coagulation in VTE. This study aims to measure and compare MP levels in joint arthroplasty patients with and without VTE. METHODS This prospective cohort study enrolled 181 patients who underwent joint arthroplasty. Ultrasound examination was used to diagnose VTE on preoperative day 0 and postoperative day 6. MPs were analysed using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and flow cytometry. The levels of platelet-derived microparticles (PMPs), endothelial cell-derived microparticles (EMPs), granulocyte-derived microparticles (GMPs), red cell-derived microparticles (RMPs), monocyte-derived microparticles (MMPs), Annexin V+ MPs (AV+ MPs), and tissue factor+ MPs (TF+ MPs) derived from five kinds of MPs were measured on day 0 (before surgery), 1, 2, 3, 4, 5, and 6 after surgery. RESULTS The levels of AV-TF+ EMPs and AV-TF+ MMPs were significantly increased in patients with VTE on postoperative day 5 compared to those without VTE (P=0.031 and P=0.031, respectively). CONCLUSION AV-TF+ MPs may indicate the development of VTE and serve as predictive markers in joint arthroplasty patients.
Collapse
Affiliation(s)
- Yuying Chen
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Jian Liu
- Adult reconstruction department, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Yu Su
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Huiru Zhao
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Yujing Zhao
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Meng Wen
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Shan Lu
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Xiangyu Cao
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Wenjie Zhang
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Lei Liu
- Department of Clinical Laboratory, Liyuan Hospital of Tongji Medical College of Huazhong University of Science and Technology, Hubei, P.R.China
| | - Jun Wu
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China; Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China.
| |
Collapse
|
50
|
Wang Y, Wu H, Sheng H, Wang Y, Li X, Wang Y, Zhao L. Discovery of anti-stroke active substances in Guhong injection based on multi-phenotypic screening of zebrafish. Biomed Pharmacother 2022; 155:113744. [PMID: 36156365 DOI: 10.1016/j.biopha.2022.113744] [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: 08/19/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/02/2022] Open
Abstract
Ischemic stroke is a leading cause of death worldwide, and it remains an urgent task to develop novel and alternative therapeutic strategies for the disease. We previously reported the positive effects of Guhong injection (GHI), composed of safflower extract and aceglutamide, in promoting functional recovery in ischemic stroke mice. However, the active substances and pharmacological mechanism of GHI is still elusive. Aiming to identify the active anti-stroke components in GHI, here we conducted a multi-phenotypic screening in zebrafish models of phenylhydrazine-induced thrombosis and ponatinib-induced cerebral ischemia. Peripheral and cerebral blood flow was quantified endogenously in erythrocytes fluorescence-labeled thrombosis fish, and baicalein and rutin were identified as major anti-thrombotic substances in GHI. Moreover, using a high-throughput video-tracking system, the effects of locomotion promotion of GHI and its main compounds were analyzed in cerebral ischemia model. Chlorogenic acid and gallic acid showed significant effects in preventing locomotor dyfunctions. Finally, GHI treatment greatly decreased the expression levels of coagulation factors F7 and F2, NF-κB and its mediated proinflammatory cytokines in the fish models. Molecular docking suggested strong affinities between baicalein and F7, and between active substances (baicalein, chlorogenic acid, gallic acid, and rutin) and NF-κB p65. In summary, our findings established a novel drug discovery method based on multi-phenotypic screening of zebrafish, provided endogenous evidences of GHI in preventing thrombus formation and promoting behavioral recovery after cerebral ischemia, and identified baicalein, rutin, chlorogenic acid, and gallic acid as active compounds in the management of ischemic stroke.
Collapse
Affiliation(s)
- Yule Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Huimin Wu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Hongda Sheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Yingchao Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, 291 Fucheng Road, Qiantang District, Hangzhou 310020, China
| | - Xuecai Li
- Tonghua Guhong Pharmaceutical Co., Ltd., 5099 Jianguo Road, Meihekou 135099, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China; Jinan Microecological Biomedicine Shandong Laboratory, 3716 Qingdao Road, Huaiyin District, Jinan 250117, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, Jinghai District, Tianjin 301617, China.
| | - Lu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China.
| |
Collapse
|