Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
J Cell Mol Med ; 28(11): e18370, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38818568

RESUMEN

The Finkel-Biskis-Jinkins Osteosarcoma (c-Fos; encoded by FOS) plays an important role in several cardiovascular diseases, including atherosclerosis and stroke. However, the relationship between FOS and venous thromboembolism (VTE) remains unknown. We identified differentially expressed genes in Gene Expression Omnibus dataset, GSE48000, comprising VTE patients and healthy individuals, and analysed them using CIBERSORT and weighted co-expression network analysis (WGCNA). FOS and CD46 expressions were significantly downregulated (FOS p = 2.26E-05, CD64 p = 8.83E-05) and strongly linked to neutrophil activity in VTE. We used GSE19151 and performed PCR to confirm that FOS and CD46 had diagnostic potential for VTE; however, only FOS showed differential expression by PCR and ELISA in whole blood samples. Moreover, we found that hsa-miR-144 which regulates FOS expression was significantly upregulated in VTE. Furthermore, FOS expression was significantly downregulated in neutrophils of VTE patients (p = 0.03). RNA sequencing performed on whole blood samples of VTE patients showed that FOS exerted its effects in VTE via the leptin-mediated adipokine signalling pathway. Our results suggest that FOS and related genes or proteins can outperform traditional clinical markers and may be used as diagnostic biomarkers for VTE.


Asunto(s)
Biología Computacional , MicroARNs , Neutrófilos , Proteínas Proto-Oncogénicas c-fos , Tromboembolia Venosa , Humanos , MicroARNs/genética , MicroARNs/sangre , MicroARNs/metabolismo , Neutrófilos/metabolismo , Tromboembolia Venosa/genética , Tromboembolia Venosa/metabolismo , Tromboembolia Venosa/sangre , Biología Computacional/métodos , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Regulación de la Expresión Génica , Masculino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Femenino , Biomarcadores/sangre , Biomarcadores/metabolismo
2.
Proteomics Clin Appl ; 18(4): e202300014, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38193270

RESUMEN

BACKGROUND: Venous thromboembolism (VTE) remains a significant cause of morbidity and mortality worldwide. Rivaroxaban, a direct oral factor Xa inhibitor, mediates anti-inflammatory and cardiovascular-protective effects besides its well-established anticoagulant properties; yet, these remain poorly characterized. Extracellular vesicles (EVs) are considered proinflammatory messengers regulating a myriad of (patho)physiological processes and may be highly relevant to the pathophysiology of VTE. The effects of Rivaroxaban on circulating EVs in VTE patients remain unknown. We have established that differential EV biosignatures are found in patients with non-valvular atrial fibrillation anticoagulated with Rivaroxaban versus warfarin. Here, we investigated whether differential proteomic profiles of circulating EVs could also be found in patients with VTE. METHODS AND RESULTS: We performed comparative label-free quantitative proteomic profiling of enriched plasma EVs from VTE patients anticoagulated with either Rivaroxaban or warfarin using a tandem mass spectrometry approach. Of the 182 quantified proteins, six were found to be either exclusive to, or enriched in, Rivaroxaban-treated patients. Intriguingly, these proteins are involved in negative feedback regulation of inflammatory and coagulation pathways, suggesting that EV proteomic signatures may reflect both Rivaroxaban's anti-coagulatory and anti-inflammatory potential. CONCLUSIONS: These differences suggest Rivaroxaban may have pleiotropic effects, supporting the reports of its emerging anti-inflammatory and cardiovascular-protective characteristics relative to warfarin.


Asunto(s)
Vesículas Extracelulares , Proteómica , Rivaroxabán , Tromboembolia Venosa , Humanos , Rivaroxabán/farmacología , Rivaroxabán/uso terapéutico , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/metabolismo , Tromboembolia Venosa/sangre , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Masculino , Femenino , Persona de Mediana Edad , Inhibidores del Factor Xa/farmacología , Inhibidores del Factor Xa/uso terapéutico , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Anciano
3.
Thromb Haemost ; 124(3): 203-222, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37967855

RESUMEN

BACKGROUND: Platelet C-type lectin-like receptor 2 (CLEC-2) induces platelet activation and aggregation after clustering by its ligand podoplanin (PDPN). PDPN, which is not normally expressed in cells in contact with blood flow, is induced in inflammatory immune cells and some malignant tumor cells, thereby increasing the risk of venous thromboembolism (VTE) and tumor metastasis. Therefore, small-molecule compounds that can interfere with the PDPN-CLEC-2 axis have the potential to become selective antiplatelet agents. METHODS AND RESULTS: Using molecular docking analysis of CLEC-2 and a PDPN-CLEC-2 binding-inhibition assay, we identified a group of diphenyl-tetrazol-propanamide derivatives as novel CLEC-2 inhibitors. A total of 12 hit compounds also inhibited PDPN-induced platelet aggregation in humans and mice. Unexpectedly, these compounds also fit the collagen-binding pocket of the glycoprotein VI molecule, thereby inhibiting collagen interaction. These compounds also inhibited collagen-induced platelet aggregation, and one compound ameliorated collagen-induced thrombocytopenia in mice. For clinical use, these compounds will require a degree of chemical modification to decrease albumin binding. CONCLUSION: Nonetheless, as dual activation of platelets by collagen and PDPN-positive cells is expected to occur after the rupture of atherosclerotic plaques, these dual antagonists could represent a promising pharmacophore, particularly for arterial thrombosis, in addition to VTE and metastasis.


Asunto(s)
Compuestos de Bifenilo , Tromboembolia Venosa , Humanos , Ratones , Animales , Simulación del Acoplamiento Molecular , Tromboembolia Venosa/metabolismo , Glicoproteínas de Membrana/metabolismo , Plaquetas/metabolismo , Agregación Plaquetaria , Glicoproteínas , Lectinas Tipo C/metabolismo , Colágeno/metabolismo
4.
J Nanobiotechnology ; 21(1): 461, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-38037042

RESUMEN

Venous thromboembolism (VTE) is a multifactorial disease, and pulmonary hypertension (PH) is a serious condition characterized by pulmonary vascular remodeling leading with increased pulmonary vascular resistance, ultimately leading to right heart failure and death. Although VTE and PH have distinct primary etiologies, they share some pathophysiologic similarities such as dysfunctional vasculature and thrombosis. In both conditions there is solid evidence that EVs derived from a variety of cell types including platelets, monocytes, endothelial cells and smooth muscle cells contribute to vascular endothelial dysfunction, inflammation, thrombosis, cellular activation and communications. However, the roles and importance of EVs substantially differ between studies depending on experimental conditions and parent cell origins of EVs that modify the nature of their cargo. Numerous studies have confirmed that EVs contribute to the pathophysiology of VTE and PH and increased levels of various EVs in relation with the severity of VTE and PH, confirming its potential pathophysiological role and its utility as a biomarker of disease severity and as potential therapeutic targets.


Asunto(s)
Vesículas Extracelulares , Hipertensión Pulmonar , Trombosis , Tromboembolia Venosa , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/terapia , Tromboembolia Venosa/metabolismo , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo
5.
Front Immunol ; 14: 1240597, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37753073

RESUMEN

Venous thromboembolism (VTE) is a leading cause of preventable deaths in hospitals, and its incidence is not decreasing despite extensive efforts in clinical and laboratory research. Venous thrombi are primarily formed in the valve pockets of deep veins, where activated monocytes play a crucial role in bridging innate immune activation and hemostatic pathways through the production of inflammatory cytokines, chemokines, and tissue factor (TF) - a principal initiator of coagulation. In the valve pocket inflammation and hypoxia (sustained/intermittent) coexist, however their combined effects on immunothrombotic processes are poorly understood. Inflammation is strongly associated with VTE, while the additional contribution of hypoxia remains largely unexplored. To investigate this, we modelled the intricate conditions of the venous valve pocket using a state-of-the-art hypoxia chamber with software-controlled oxygen cycling. We comprehensively studied the effects of sustained and intermittent hypoxia alone, and in combination with VTE-associated inflammatory stimuli on primary monocytes. TF expression and activity was measured in monocytes subjected to sustained and intermittent hypoxia alone, or in combination with IL-1ß. Monocyte responses were further analyzed in detailed by RNA sequencing and validated by ELISA. Stimulation with IL-1ß alone promoted both transcription and activity of TF. Interestingly, the stimulatory effect of IL-1ß on TF was attenuated by sustained hypoxia, but not by intermittent hypoxia. Our transcriptome analysis further confirmed that sustained hypoxia limited the pro-inflammatory response induced by IL-1ß, and triggered a metabolic shift in monocytes. Intermittent hypoxia alone had a modest effect on monocyte transcript. However, in combination with IL-1ß intermittent hypoxia significantly altered the expression of 2207 genes and enhanced the IL-1ß-stimulatory effects on several chemokine and interleukin genes (e.g., IL-19, IL-24, IL-32, MIF), as well as genes involved in coagulation (thrombomodulin) and fibrinolysis (VEGFA, MMP9, MMP14 and PAI-1). Increased production of CCL2, IL-6 and TNF following stimulation with intermittent hypoxia and IL-1ß was confirmed by ELISA. Our findings provide valuable insights into how the different hypoxic profiles shape the immunothrombotic response of monocytes and shed new light on the early events in the pathogenesis of venous thrombosis.


Asunto(s)
Monocitos , Tromboembolia Venosa , Humanos , Tromboembolia Venosa/metabolismo , Citocinas/metabolismo , Hipoxia/metabolismo , Inflamación/metabolismo , Tromboplastina/metabolismo
6.
Science ; 380(6641): 178-187, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37053338

RESUMEN

Venous thromboembolism (VTE) comprising deep venous thrombosis and pulmonary embolism is a major cause of morbidity and mortality. Short-term immobility-related conditions are a major risk factor for the development of VTE. Paradoxically, long-term immobilized free-ranging hibernating brown bears and paralyzed spinal cord injury (SCI) patients are protected from VTE. We aimed to identify mechanisms of immobility-associated VTE protection in a cross-species approach. Mass spectrometry-based proteomics revealed an antithrombotic signature in platelets of hibernating brown bears with heat shock protein 47 (HSP47) as the most substantially reduced protein. HSP47 down-regulation or ablation attenuated immune cell activation and neutrophil extracellular trap formation, contributing to thromboprotection in bears, SCI patients, and mice. This cross-species conserved platelet signature may give rise to antithrombotic therapeutics and prognostic markers beyond immobility-associated VTE.


Asunto(s)
Plaquetas , Proteínas del Choque Térmico HSP47 , Hipocinesia , Traumatismos de la Médula Espinal , Ursidae , Tromboembolia Venosa , Animales , Humanos , Ratones , Fibrinolíticos/uso terapéutico , Embolia Pulmonar/tratamiento farmacológico , Embolia Pulmonar/etnología , Embolia Pulmonar/metabolismo , Factores de Riesgo , Traumatismos de la Médula Espinal/complicaciones , Ursidae/metabolismo , Tromboembolia Venosa/etiología , Tromboembolia Venosa/metabolismo , Hipocinesia/complicaciones , Proteínas del Choque Térmico HSP47/metabolismo , Plaquetas/metabolismo
7.
J Thromb Haemost ; 21(5): 1227-1237, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36736832

RESUMEN

BACKGROUND: von Willebrand factor (VWF) and its cleaving protease, ADAMTS-13, form a pivotal axis that regulates hemostasis. However, the role of the VWF-ADAMTS-13 axis in the risk of future venous thromboembolism (VTE) is unknown. OBJECTIVES: To investigate whether plasma ADAMTS-13 levels and an imbalance with VWF levels, assessed as the VWF/ADAMTS-13 ratio, are associated with the risk of future VTE. PATIENTS/METHODS: A population-based nested case-control study, comprising 383 incident VTE cases and 780 age- and sex-matched controls, was derived from the Tromsø study cohort (1994-2007). Antigen levels of ADAMTS-13 and VWF were measured in plasma samples obtained at cohort baseline. Odds ratios (ORs) with 95% CIs were estimated according to quartile cutoffs of ADAMTS-13 and VWF/ADAMTS-13 ratio determined in controls. RESULTS: In age- and sex-adjusted analysis, ADAMTS-13 levels were inversely associated with the VTE risk, with an OR of 1.40 (95% CI, 0.99-1.99) for the lowest vs highest quartiles. The VWF/ADAMTS-13 ratio was linearly associated with the VTE risk (P for trend = .001), with an OR of 1.70 (95% CI, 1.19-2.43) for the highest vs lowest quartiles, and the association was particularly pronounced for unprovoked VTE (OR, 2.81; 95% CI, 1.65-4.81). The ORs were only slightly attenuated after additional adjustments for body mass index and C-reactive protein. CONCLUSIONS: Lowered ADAMTS-13 levels and an imbalance between ADAMTS-13 and VWF levels, reflected by an increased VWF/ADAMTS-13 ratio, were associated with an increased risk of future VTE. Our findings suggest that the VWF-ADAMTS-13 axis is involved in the pathogenesis of VTE.


Asunto(s)
Proteína ADAMTS13 , Tromboembolia Venosa , Factor de von Willebrand , Humanos , Proteína ADAMTS13/sangre , Proteína ADAMTS13/metabolismo , Proteína C-Reactiva/análisis , Estudios de Casos y Controles , Tromboembolia Venosa/sangre , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/epidemiología , Tromboembolia Venosa/metabolismo , Factor de von Willebrand/análisis , Factor de von Willebrand/metabolismo
8.
Comput Math Methods Med ; 2022: 9610071, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36245842

RESUMEN

Objective: To investigate the diagnostic value of circulating integrins ß1, 2, and 3 in venous thrombosis (VTE). Materials and Methods: A total of 474 VTE patients and 306 patients with nonhigh risk for VTE as the control group were studied. Levels of adhering integrins ß1, 2, and 3 were detected by flow cytometry. Levels of circulating integrins ß1, 2, and 3 in serum were measured by enzyme-linked immunosorbent assay. Results: We found that integrins ß1, 2, and 3 were expressed highly both in serum and on the surface of leukocytes and platelets in venous thromboembolism. The levels of circulating integrins ß1, 2, and 3 are positively correlated with adhering integrins. It showed excellent clinical diagnostic performance of circulating integrins ß1, 2, and 3 in venous thromboembolism. Conclusions: Integrin subunit ß can be used as a diagnostic marker with high sensitivity and specificity for venous thromboembolism.


Asunto(s)
Integrinas , Tromboembolia Venosa , Citometría de Flujo , Humanos , Integrina beta1/metabolismo , Integrinas/metabolismo , Leucocitos , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/metabolismo
9.
World J Gastroenterol ; 28(26): 3132-3149, 2022 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-36051331

RESUMEN

BACKGROUND: The development of venous thromboembolism (VTE) is associated with high mortality among gastric cancer (GC) patients. Neutrophil extracellular traps (NETs) have been reported to correlate with the prothrombotic state in some diseases, but are rarely reported in GC patients. AIM: To investigate the effect of NETs on the development of cancer-associated thrombosis in GC patients. METHODS: The levels of NETs in blood and tissue samples of patients were analyzed by ELISA, flow cytometry, and immunofluorescence staining. NET generation and hypercoagulation of platelets and endothelial cells (ECs) in vitro were observed by immunofluorescence staining. NET procoagulant activity (PCA) was determined by fibrin formation and thrombin-antithrombin complex (TAT) assays. Thrombosis in vivo was measured in a murine model induced by flow stenosis in the inferior vena cava (IVC). RESULTS: NETs were likely to form in blood and tissue samples of GC patients compared with healthy individuals. In vitro studies showed that GC cells and their conditioned medium, but not gastric mucosal epithelial cells, stimulated NET release from neutrophils. In addition, NETs induced a hypercoagulable state of platelets by upregulating the expression of phosphatidylserine and P-selectin on the cells. Furthermore, NETs stimulated the adhesion of normal platelets on glass surfaces. Similarly, NETs triggered the conversion of ECs to hypercoagulable phenotypes by downregulating the expression of their intercellular tight junctions but upregulating that of tissue factor. Treatment of normal platelets or ECs with NETs augmented the level of plasma fibrin formation and the TAT complex. In the models of IVC stenosis, tumor-bearing mice showed a stronger ability to form thrombi, and NETs abundantly accumulated in the thrombi of tumor-bearing mice compared with control mice. Notably, the combination of deoxyribonuclease I, activated protein C, and sivelestat markedly abolished the PCA of NETs. CONCLUSION: GC-induced NETs strongly increased the risk of VTE development both in vitro and in vivo. NETs are potential therapeutic targets in the prevention and treatment of VTE in GC patients.


Asunto(s)
Trampas Extracelulares , Neoplasias Gástricas , Trombofilia , Trombosis , Tromboembolia Venosa , Animales , Constricción Patológica , Células Endoteliales/metabolismo , Trampas Extracelulares/metabolismo , Fibrina , Ratones , Neutrófilos/metabolismo , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/metabolismo , Trombosis/etiología , Tromboembolia Venosa/etiología , Tromboembolia Venosa/metabolismo
10.
J Thromb Haemost ; 20(10): 2350-2365, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35722954

RESUMEN

BACKGROUND: Breast cancer results in a three- to four-fold increased risk of venous thromboembolism (VTE), which is associated with reduced patient survival. Despite this, the mechanisms underpinning breast cancer-associated thrombosis remain poorly defined. Tumor cells can trigger endothelial cell (EC) activation resulting in increased von Willebrand factor (VWF) secretion. Importantly, elevated plasma VWF levels constitute an independent biomarker for VTE risk. Moreover, in a model of melanoma, treatment with low molecular weight heparin (LMWH) negatively regulated VWF secretion and attenuated tumor metastasis. OBJECTIVE: To investigate the role of VWF in breast cancer metastasis and examine the effect of LMWH in modulating EC activation and breast tumor transmigration. METHODS: von Willebrand factor levels were measured by ELISA. Primary ECs were used to assess tumor-induced activation, angiogenesis, tumor adhesion, and transendothelial migration. RESULTS AND CONCLUSION: Patients with metastatic breast cancer have markedly elevated plasma VWF:Ag levels that also correlate with poorer survival. MDA-MB-231 and MCF-7 breast cancer cells induce secretion of VWF, angiopoietin-2, and osteoprotegerin from ECs, which is further enhanced by the presence of platelets. Vascular endothelial growth factor-A (VEGF-A) plays an important role in modulating breast cancer-induced VWF release. Moreover, VEGF-A from breast tumor cells also contributes to a pro-angiogenic effect on ECs. VWF multimers secreted from ECs, in response to tumor-VEGF-A, mediate adhesion of breast tumor cells along the endothelium. LMWH inhibits VWF-breast tumor adhesion and transendothelial migration. Our findings highlight the significant crosstalk between tumor cells and the endothelium including increased VWF secretion which may contribute to tumor metastasis.


Asunto(s)
Neoplasias de la Mama , Tromboembolia Venosa , Angiopoyetina 2/metabolismo , Neoplasias de la Mama/metabolismo , Células Endoteliales/metabolismo , Femenino , Heparina de Bajo-Peso-Molecular/farmacología , Heparina de Bajo-Peso-Molecular/uso terapéutico , Humanos , Osteoprotegerina/metabolismo , Migración Transendotelial y Transepitelial , Factor A de Crecimiento Endotelial Vascular/metabolismo , Tromboembolia Venosa/metabolismo , Factor de von Willebrand/metabolismo
11.
Dis Markers ; 2021: 1776567, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925639

RESUMEN

BACKGROUND: This current study applied bioinformatics analysis to reveal the crosstalk between venous thromboembolism (VTE) and periodontitis, as well as the potential role of immune-related genes in this context. METHODS: Expression data were downloaded from the GEO database. Blood samples from venous thromboembolism (VTE) were used (GSE19151), while for periodontal disease, we used gingival tissue samples (GSE10334, GSE16134, and GSE23586). After batch correction, we used "limma" packages of R language for differential expression analysis (p value < 0.05, ∣logFC | ≥0.5). We used Venn diagrams to extract the differentially expressed genes common to VTE and periodontitis as potential crosstalk genes and applied functional enrichment analysis (GO biological process and KEGG pathway). The protein-protein interaction (PPI) network of crosstalk genes was constructed by Cytoscape software. The immune-related genes were downloaded from the literature. The Wilcoxon test was used to test the scores of immune infiltrating cells. The crosstalk genes were further screened by LASSO Logistic Regression. RESULTS: For periodontitis, 427 case and 136 control samples, and for VTE, 70 case and 63 control samples were included. The obtained PPI network had 1879 nodes and 2257 edges. Moreover, 782 immune genes and 28 cell types were included in the analysis. Over 90% of immune cells had different expressions in VTE and periodontitis. We obtained 12 significant pathways corresponding to crosstalk genes. CD3D, CSF3R, and CXCR4 acted as an immune gene and a crosstalk gene. We obtained a total of 12 shared biomarker crosstalk genes. Among those 12 biomarker crosstalk genes, 4 were immune genes (LGALS1, LSP1, SAMSN1, and WIPF1). CONCLUSION: Four biomarker crosstalk genes between periodontitis and VTE were also immune genes, i.e., LGALS1, LSP1, SAMSN1, and WIPF1. The findings of the current study need further validation and are a basis for development of biomarkers.


Asunto(s)
Biología Computacional , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Enfermedades Periodontales/genética , Mapas de Interacción de Proteínas , Tromboembolia Venosa/genética , Biomarcadores/metabolismo , Estudios de Casos y Controles , Bases de Datos Genéticas , Marcadores Genéticos , Humanos , Enfermedades Periodontales/complicaciones , Enfermedades Periodontales/inmunología , Enfermedades Periodontales/metabolismo , Tromboembolia Venosa/complicaciones , Tromboembolia Venosa/inmunología , Tromboembolia Venosa/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 41(11): 2681-2692, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34496636

RESUMEN

The immunoglobulin receptor GPVI (glycoprotein VI) is selectively expressed on megakaryocytes and platelets and is currently recognized as a receptor for not only collagen but also a variety of plasma and vascular proteins, including fibrin, fibrinogen, laminin, fibronectin, and galectin-3. Deficiency of GPVI is protective in mouse models of experimental thrombosis, pulmonary thromboembolism as well as in thromboinflammation, suggesting a role of GPVI in arterial and venous thrombus formation. In humans, platelet GPVI deficiency is associated with a mild bleeding phenotype, whereas a common variant rs1613662 in the GP6 gene is considered a risk factor for venous thromboembolism. However, preclinical studies on the inhibition of GPVI-ligand interactions are focused on arterial thrombotic complications. In this review we discuss the emerging evidence for GPVI in venous thrombus formation and leukocyte-dependent thromboinflammation, extending to venous thromboembolism, pulmonary thromboembolism, and cancer metastasis. We also recapitulate indications for circulating soluble GPVI as a biomarker of thrombosis-related complications. Collectively, we conclude that the current evidence suggests that platelet GPVI is also a suitable cotarget in the prevention of venous thrombosis due to its role in thrombus consolidation and platelet-leukocyte complex formation.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Inflamación/metabolismo , Activación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Tromboembolia Venosa/metabolismo , Trombosis de la Vena/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/patología , Fibrinolíticos/uso terapéutico , Humanos , Inflamación/sangre , Inflamación/tratamiento farmacológico , Inflamación/patología , Mediadores de Inflamación/sangre , Ligandos , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Transducción de Señal , Tromboembolia Venosa/sangre , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/patología , Trombosis de la Vena/sangre , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/patología
13.
Cells ; 10(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34200229

RESUMEN

Glioblastoma is among the tumor entities with an extreme thrombogenic potential and patients are at very high risk of developing a venous thromboembolism (VTE) over the course of the disease, with an incidence of up to 30% per year. Major efforts are currently being made to understand and gain novel insights into the underlying pathomechanisms of the development of VTE in patients with glioblastoma and to find appropriate biomarkers. Yet, patients with glioblastoma not only face a high thromboembolic risk but are also at risk of bleeding events. In the case of VTE, a therapeutic anticoagulation with low molecular weight heparin or, in the case of low bleeding risk, treatment with a direct oral anticoagulant, is recommended, according to recently published guidelines. With respect to an elevated bleeding risk in glioblastoma patients, therapeutic anticoagulation remains challenging in this patient group and prospective data for this vulnerable patient group are scarce, particularly with regard to direct oral anticoagulants.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Glioblastoma , Hemorragia , Heparina de Bajo-Peso-Molecular/uso terapéutico , Tromboembolia Venosa , Glioblastoma/complicaciones , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Hemorragia/metabolismo , Hemorragia/patología , Humanos , Guías de Práctica Clínica como Asunto , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/etiología , Tromboembolia Venosa/metabolismo , Tromboembolia Venosa/patología
14.
J Trauma Acute Care Surg ; 90(5): 845-852, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33797501

RESUMEN

BACKGROUND: The Pragmatic Randomized Optimal Platelet and Plasma Ratios (PROPPR) trial showed that 15% of patients developed venous thromboembolism (VTE) following hemorrhage, but the mechanisms are unknown. Since inflammation is associated with hypercoagulability and thrombosis, our goal was to compare the temporal inflammatory profile following hemorrhagic shock in patients with and without VTE. STUDY DESIGN: Secondary analysis was performed on data collected from PROPPR. Blood samples collected at 0 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, and 72 hours following admission were assayed on a 27-target cytokine panel, and compared between VTE (n = 83) and non-VTE (n = 475) patients. p < 0.05 indicated significance. RESULTS: Over time, both groups exhibited elevations in proinflammatory mediators interleukin (IL)-6, IL-8, IL-10, granulocyte colony-stimulating factor 57, monocyte chemoattractant protein 1 and macrophage inflammatory protein 1ß, and anti-inflammatory mediators IL-1ra and IL-10 (p < 0.05 vs. admission). Venous thromboembolism patients showed amplified responses for IL-6 (6-72 hours) and IL-8 (6-24 hours), which peaked at later time points, and granulocyte colony-stimulating factor 57 (12-24 hours), monocyte chemoattractant protein 1 (6-72 hours), and macrophage inflammatory protein-1 ß (2-12 hours) (p < 0.05 vs. non-VTE per time point) that peaked at similar time points to non-VTE patients. The anti-inflammatory responses were similar between groups, but the interleukin-mediated proinflammatory responses continued to rise after the peak anti-inflammatory response in the VTE group. The occurrence rate of adverse events was higher in VTE (97%) versus non-VTE (87%, p = 0.009) and was associated with higher inflammation. CONCLUSION: Patients with VTE following hemorrhagic shock exhibited a prolonged and amplified proinflammatory responses mediated by select interleukin, chemotactic, and glycoprotein cytokines that are not antagonized by anti-inflammatory mediators. This response is not related to randomization group, injury severity or degree of shock, but may be linked to adverse events. LEVEL OF EVIDENCE: Prognostic, level III.


Asunto(s)
Antiinflamatorios/uso terapéutico , Inflamación/complicaciones , Choque Hemorrágico/complicaciones , Tromboembolia Venosa/etiología , Adulto , Anticoagulantes/uso terapéutico , Citocinas/metabolismo , Femenino , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Método Simple Ciego , Estados Unidos , Tromboembolia Venosa/metabolismo , Adulto Joven
15.
Ann Vasc Surg ; 74: 389-399, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33819580

RESUMEN

OBJECTIVES: To explore the key genes, and correlated pathways in venous thromboembolism (VTE) via bioinformatic analysis, and expected our findings could contribute to the development of new biomarkers and therapeutic target for VTE. METHODS: Two VTE-related microarray expression profiles (GSE48000 and GSE19151) were downloaded from the Gene Expression Ominibus (GEO) database. Differentially expressed genes (DEGs) were analyzed using limma package, and overlapping DEGs were identified form the above two expression profiles. Then, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEEG) pathway enrichment analyses were performed by DAVID. Protein-protein interaction (PPI) network was constructed by using STRING and visualized with Cytoscape. Furthermore, module analysis plus centrality analysis of the PPI network were executed to identify the potential key genes. Finally, the pathway analysis was performed using GenCLiP 3.0. RESULTS: A total of 173 DEGs (125 upregulated and 48 downregulated) were identified. GO analysis demonstrated that DEGs were mainly enriched in viral life cycle, ribosome and structural constituent of ribosome. Meanwhile, KEGG pathway analysis showed that these genes were enriched in ribosome, Parkinson's disease and cell cycle. Additionally, one most significant module and 12 hub genes were found. Finally, 6 key genes, namely ISG15, RPS15A, MRPL13, ICT1, MRPL15 and RPLP0, with high centrality features were identified. These key genes were mainly involved in translation, metabolism of proteins and ribosome pathway. CONCLUSIONS: In summary, these 6 identified genes and correlated pathways should play an important role in VTE, which can provide new insight into the molecular mechanism, potential biomarkers and therapeutic targets associated with VTE.


Asunto(s)
Expresión Génica , Redes y Vías Metabólicas , Mapas de Interacción de Proteínas , Tromboembolia Venosa/genética , Tromboembolia Venosa/metabolismo , Biomarcadores , Biología Computacional , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos
16.
Blood ; 137(19): 2681-2693, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33529319

RESUMEN

Patients with isolated pulmonary embolism (PE) have a distinct clinical profile from those with deep vein thrombosis (DVT)-associated PE, with more pulmonary conditions and atherosclerosis. These findings suggest a distinct molecular pathophysiology and the potential involvement of alternative pathways in isolated PE. To test this hypothesis, data from 532 individuals from the Genotyping and Molecular Phenotyping of Venous ThromboEmbolism Project, a multicenter prospective cohort study with extensive biobanking, were analyzed. Targeted, high-throughput proteomics, machine learning, and bioinformatic methods were applied to contrast the acute-phase plasma proteomes of isolated PE patients (n = 96) against those of patients with DVT-associated PE (n = 276) or isolated DVT (n = 160). This resulted in the identification of shared molecular processes between PE phenotypes, as well as an isolated PE-specific protein signature. Shared processes included upregulation of inflammation, response to oxidative stress, and the loss of pulmonary surfactant. The isolated PE-specific signature consisted of 5 proteins: interferon-γ, glial cell line-derived neurotrophic growth factor, polypeptide N-acetylgalactosaminyltransferase 3, peptidyl arginine deiminase type-2, and interleukin-15 receptor subunit α. These proteins were orthogonally validated using cis protein quantitative trait loci. External replication in an independent population-based cohort (n = 5778) further validated the proteomic results and showed that they were prognostic for incident primary isolated PE in individuals without history of VTE (median time to event: 2.9 years; interquartile range: 1.6-4.2 years), supporting their possible involvement in the early pathogenesis. This study has identified molecular overlaps and differences between VTE phenotypes. In particular, the results implicate noncanonical pathways more commonly associated with respiratory and atherosclerotic disease in the acute pathophysiology of isolated PE.


Asunto(s)
Proteoma , Embolia Pulmonar/metabolismo , Transcriptoma , Proteínas de Fase Aguda/biosíntesis , Adulto , Anciano , Aterosclerosis/complicaciones , Comorbilidad , Conjuntos de Datos como Asunto , Femenino , Estudios de Seguimiento , Regulación de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Interferón gamma/biosíntesis , Interferón gamma/genética , Subunidad alfa del Receptor de Interleucina-15/biosíntesis , Subunidad alfa del Receptor de Interleucina-15/genética , Aprendizaje Automático , Masculino , Persona de Mediana Edad , N-Acetilgalactosaminiltransferasas/biosíntesis , N-Acetilgalactosaminiltransferasas/genética , Estrés Oxidativo , Estudios Prospectivos , Mapas de Interacción de Proteínas , Arginina Deiminasa Proteína-Tipo 2/biosíntesis , Arginina Deiminasa Proteína-Tipo 2/genética , Embolia Pulmonar/genética , Embolia Pulmonar/fisiopatología , Surfactantes Pulmonares , Sitios de Carácter Cuantitativo , Tromboembolia Venosa/metabolismo , Polipéptido N-Acetilgalactosaminiltransferasa
17.
Postgrad Med ; 133(5): 508-516, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33480813

RESUMEN

Myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (MF) are stem cell clonal neoplasms characterized by expansion of late myeloid cells. Thrombosis risk is elevated in MPNs and contributes significantly to morbidity and mortality. Current consensus guidelines make no specific recommendations regarding anticoagulant choice for the treatment of venous thromboembolism (VTE) in MPNs, with most evidence supporting the use of vitamin K antagonists (VKAs) for secondary prophylaxis. However, direct oral anticoagulants (DOACs) are now increasingly being used, although with limited data on safety and efficacy in MPNs specifically. The widespread adoption of DOACs is based on new, high-quality evidence demonstrating safety and efficacy of DOAC treatment for cancer-associated VTE. However, these studies include few if any MPN patients, and MPNs have disease-specific considerations that may elevate thrombosis and bleeding risk. The purpose of this review is to discuss evidence behind current treatment recommendations for thrombosis in MPNs, with special attention to the use of DOACs.


Asunto(s)
Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/metabolismo , Neoplasias/tratamiento farmacológico , Tromboembolia Venosa/prevención & control , Humanos , Mutación , Trastornos Mieloproliferativos/complicaciones , Neoplasias/metabolismo , Prevención Secundaria , Tromboembolia Venosa/metabolismo
18.
Respir Investig ; 59(3): 327-334, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33386292

RESUMEN

BACKGROUND: The risk of venous thromboembolism (VTE) is increased 7-fold in patients with cancer than in those without. Low-molecular-weight heparin is the standard treatment for cancer-associated VTE. Direct oral anticoagulants (DOACs) are not inferior to low-molecular-weight heparin with respect to the general outcome of recurrent VTE. Warfarin is associated with a risk of bleeding when used in combination with gefitinib or erlotinib which are epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs). It is unclear, however, whether combination treatments with EGFR-TKIs and DOACs pose the same risk. We aimed to identify anticancer drugs and anticoagulants that can be used safely in combination, as accompanying research to an observational study on VTE incidence rates in lung cancer patients (Rising-VTE/NEJ037 study). METHODS: Twelve patients receiving EFGR-TKI monotherapy and VTE treatment were enrolled. Blood samples were collected in time series after the first dose of edoxaban, and further samples were collected within 8-15 days after administering EGFR-TKIs. The pharmacokinetics (PK) of edoxaban were analyzed using a non-compartmental model. RESULTS: Edoxaban concentrations (30 mg once daily) were measured in eight patients. PK analyses showed no significant differences before and after co-administration of EGFR-TKIs (gefitinib, erlotinib, and afatinib). CONCLUSIONS: Our findings indicate that the PK of edoxaban was not considerably affected by co-administration of EGFR-TKIs (gefitinib, erlotinib, and afatinib).


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Inhibidores del Factor Xa/farmacocinética , Neoplasias Pulmonares/metabolismo , Mutación , Piridinas/farmacocinética , Tiazoles/farmacocinética , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/metabolismo , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/complicaciones , Carcinoma de Pulmón de Células no Pequeñas/genética , Interacciones Farmacológicas , Quimioterapia Combinada , Receptores ErbB/genética , Clorhidrato de Erlotinib/administración & dosificación , Inhibidores del Factor Xa/administración & dosificación , Femenino , Gefitinib/administración & dosificación , Humanos , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/genética , Masculino , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridinas/administración & dosificación , Tiazoles/administración & dosificación , Tromboembolia Venosa/etiología
20.
Front Immunol ; 11: 574997, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324398

RESUMEN

Membranous nephropathy (MN) is a rare autoimmune kidney disease. Most autoimmune diseases are associated with a pro-inflammatory Th17-immune response, but little is known about immune dysregulation in MN. In China, MN was associated with exposure to fine air particulate matter (PM2.5) that could act as a danger signal and redirect immune response toward the Th2 or Th17 pathway. We aimed to analyze the cytokine profile of MN patients and to study the possible environmental factors involved in this immune reorientation, as well as the consequences on the prognosis of the disease. In this prospective study, 59 MN patients filled a comprehensive lifestyle questionnaire. Peripheral blood cells from MN patients were stimulated in vitro to measure the cytokines produced in supernatant. Cytokine profiles of MN patients were compared to 28 healthy donors and analyzed regarding individual PM2.5 exposure. Compared to healthy donors, MN patients had higher serum levels of Th17 and Th2 cytokines IL-17A (62 pg/ml [IQR, 16-160] versus 31 [IQR, 13-51], P=0.035), IL-6 (66767 pg/ml [IQR, 36860-120978] versus 27979 [IQR, 18672-51499], P=0.001), and IL-4 (12 pg/ml [IQR, 0-33] versus 0 pg/ml [IQR, 0-0], P=0.0003), respectively, as well as a deficiency of Th1 and regulatory T cell cytokines IFN-γ (5320 pg/ml [IQR, 501-14325] versus 18037 [IQR, 4889-31329], P=0.0005) and IL-10 (778 pg/ml [IQR, 340-1247] versus 1102 [IQR, 737-1652], P=0.04), respectively. MN patients with high IL-17A levels lived in areas highly exposed to PM2.5: 51 µg/m3 versus 31 µg/m3 for patients with low IL-17A levels (P=0.002) while the World Health Organization recommends an exposition below 10 µg/m3. MN patients with Th17-mediated inflammation had more venous thromboembolic events (P=0.03) and relapsed more often (P=0.0006). Rituximab treatment induced Th1 and regulatory T cell cytokines but did not impact Th17 cytokines. MN patients with Th17-mediated inflammation which appears to be related to an urban environment have worse prognosis. Alternative strategies targeting dysregulated cytokine balance could be considered for these patients at high risk of relapse.


Asunto(s)
Citocinas/sangre , Contaminantes Ambientales/efectos adversos , Glomerulonefritis Membranosa/inmunología , Material Particulado/efectos adversos , Células Th17/inmunología , Tromboembolia Venosa/inmunología , Adulto , Anciano , Antiinflamatorios/uso terapéutico , Biomarcadores/sangre , Estudios de Casos y Controles , Células Cultivadas , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Glomerulonefritis Membranosa/sangre , Glomerulonefritis Membranosa/tratamiento farmacológico , Glomerulonefritis Membranosa/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Recurrencia , Medición de Riesgo , Factores de Riesgo , Rituximab/uso terapéutico , Células Th17/efectos de los fármacos , Células Th17/metabolismo , Salud Urbana , Tromboembolia Venosa/sangre , Tromboembolia Venosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...