Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.872
Filtrer
2.
J Orthop Surg Res ; 19(1): 505, 2024 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-39182115

RÉSUMÉ

BACKGROUND: Deep vein thrombosis (DVT) of lower extremity is a common complications after total knee arthroplasty (TKA). The purpose of this study was to evaluate the risk factors for DVT after TKA and analyze the expression of miR-199b-5p and nitric oxide (NO) before and after TKA, as well as their predictive value for DVT. METHODS: Basic clinical information of 121 patients with TKA was analyzed retrospectively. RT-qPCR was used to detect the relative expression level of miR-199b-5p in patients before and after TKA treatment. Based on the occurrence of DVT, patients were divided into DVT and non-DVT groups. Logistic regression analysis evaluated the risk factors of DVT. The receiver operating characteristic (ROC) curve assessed the predictive value of postoperative miR-199b-5p level, preoperative NO level, and their combination in DVT. The target genes of miR-199b-5p and their functions were predicted and annotated using bioinformatics analysis. RESULTS: The level of miR-199b-5p after TKA was upregulated compared with that before TKA (P < 0.001). DVT occurred in 20 of 121 patients after TKA, with an incidence of 16.53%. Multivariate analysis showed that age, family history of DVT, decrease of NO and increase of miR-199b-5p were risk factors for DVT after TKA (P < 0.05). The ROC curve showed that both miR-199b-5p and NO had certain diagnostic value for DVT, but the combination of miR-199b-5p and NO had the highest diagnostic accuracy (P < 0.001). CONCLUSION: This study showed that the expression of miR-199b-5p was up-regulated after TKA, and miR-199b-5p levels were higher in DVT patients than in non-DVT patients. miR-199b-5p combined with NO is of great value in the diagnosis of DVT after TKA.


Sujet(s)
Arthroplastie prothétique de genou , microARN , Monoxyde d'azote , Valeur prédictive des tests , Thrombose veineuse , Humains , Arthroplastie prothétique de genou/effets indésirables , Mâle , Femelle , microARN/génétique , Monoxyde d'azote/métabolisme , Sujet âgé , Adulte d'âge moyen , Thrombose veineuse/étiologie , Thrombose veineuse/génétique , Études rétrospectives , Complications postopératoires/étiologie , Facteurs de risque
3.
BMC Cardiovasc Disord ; 24(1): 383, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054435

RÉSUMÉ

BACKGROUND: The aim of this study was to explore the genetic effects of hormones modulated through the pituitary-thyroid/adrenal/gonadal axis on the risk of developing venous thromboembolism (VTE) and to investigate the potentially causal relationships between them. METHODS: A two-sample Mendelian randomization (MR) design was used. The single-nucleotide polymorphisms (SNPs) used as instrumental variables for various hormones and hormone-mediated diseases were derived from published genome-wide association studies (GWASs). Summary statistics for the risk of developing VTE (including deep venous thrombosis [DVT] and pulmonary embolism [PE]) were obtained from the UK Biobank and the FinnGen consortium. Inverse-variance weighting (IVW) was applied as the primary method to analyse causal associations. Other MR methods were used for supplementary estimates and sensitivity analysis. RESULTS: A genetic predisposition to greater free thyroxine (FT4) concentrations was associated with a greater risk of developing DVT (OR = 1.0007, 95%CI [1.0001-1.0013], p = 0.0174) and VTE (OR = 1.0008, 95%CI [1.0002-1.0013], p = 0.0123). Genetically predicted hyperthyroidism was significantly associated with an increased risk of developing DVT (OR = 1.0685, 95%CI [1.0139-1.1261], p = 0.0134) and VTE (OR = 1.0740, 95%CI [1.0165-1.1348], p = 0.0110). According to the initial MR analysis, testosterone concentrations were positively associated with the risk of developing VTE (OR = 1.0038, 95%CI [1.004-1.0072], p = 0.0285). After sex stratification, estradiol concentrations were positively associated with the risk of developing DVT (OR = 1.0143, 95%CI [1.0020-1.0267], p = 0.0226) and VTE (OR = 1.0156, 95%CI [1.0029-1.0285], p = 0.0158) in females, while the significant relationship between testosterone and VTE did not persist. SHBG rs858518 was identified as the only SNP that was associated with an increased risk of developing VTE, mediated by estradiol, in females. CONCLUSIONS: Genetically predicted hyperthyroidism and increased FT4 concentrations were positively associated with the risk of developing VTE. The effects of genetically predicted sex hormones on the risk of developing VTE differed between males and females. Greater genetically predicted estradiol concentrations were associated with an increased risk of developing VTE in females, while the SHBG rs858518 variant may become a potential prevention and treatment target for female VTE.


Sujet(s)
Prédisposition génétique à une maladie , Étude d'association pangénomique , Analyse de randomisation mendélienne , Polymorphisme de nucléotide simple , Thromboembolisme veineux , Humains , Thromboembolisme veineux/génétique , Thromboembolisme veineux/épidémiologie , Thromboembolisme veineux/diagnostic , Thromboembolisme veineux/sang , Facteurs de risque , Appréciation des risques , Femelle , Mâle , Thyroxine/sang , Phénotype , Marqueurs biologiques/sang , Thrombose veineuse/génétique , Thrombose veineuse/épidémiologie , Thrombose veineuse/sang , Thrombose veineuse/diagnostic , Facteurs sexuels , Testostérone/sang , Embolie pulmonaire/génétique , Embolie pulmonaire/épidémiologie , Embolie pulmonaire/sang , Embolie pulmonaire/diagnostic
4.
Sci Rep ; 14(1): 15884, 2024 07 10.
Article de Anglais | MEDLINE | ID: mdl-38987624

RÉSUMÉ

Behçet's disease (BD) is a multifaceted autoimmune disorder affecting multiple organ systems. Vascular complications, such as venous thromboembolism (VTE), are highly prevalent, affecting around 50% of individuals diagnosed with BD. This study aimed to identify potential biomarkers for VTE in BD patients. Three microarray datasets (GSE209567, GSE48000, GSE19151) were retrieved for analysis. Differentially expressed genes (DEGs) associated with VTE in BD were identified using the Limma package and weighted gene co-expression network analysis (WGCNA). Subsequently, potential diagnostic genes were explored through protein-protein interaction (PPI) network analysis and machine learning algorithms. A receiver operating characteristic (ROC) curve and a nomogram were constructed to evaluate the diagnostic performance for VTE in BD patients. Furthermore, immune cell infiltration analyses and single-sample gene set enrichment analysis (ssGSEA) were performed to investigate potential underlying mechanisms. Finally, the efficacy of listed drugs was assessed based on the identified signature genes. The limma package and WGCNA identified 117 DEGs related to VTE in BD. A PPI network analysis then selected 23 candidate hub genes. Four DEGs (E2F1, GATA3, HDAC5, and MSH2) were identified by intersecting gene sets from three machine learning algorithms. ROC analysis and nomogram construction demonstrated high diagnostic accuracy for these four genes (AUC: 0.816, 95% CI: 0.723-0.909). Immune cell infiltration analysis revealed a positive correlation between dysregulated immune cells and the four hub genes. ssGSEA provided insights into potential mechanisms underlying VTE development and progression in BD patients. Additionally, therapeutic agent screening identified potential drugs targeting the four hub genes. This study employed a systematic approach to identify four potential hub genes (E2F1, GATA3, HDAC5, and MSH2) and construct a nomogram for VTE diagnosis in BD. Immune cell infiltration analysis revealed dysregulation, suggesting potential macrophage involvement in VTE development. ssGSEA provided insights into potential mechanisms underlying BD-induced VTE, and potential therapeutic agents were identified.


Sujet(s)
Maladie de Behçet , Marqueurs biologiques , Biologie informatique , Analyse de profil d'expression de gènes , Cartes d'interactions protéiques , Humains , Maladie de Behçet/génétique , Maladie de Behçet/complications , Maladie de Behçet/diagnostic , Biologie informatique/méthodes , Cartes d'interactions protéiques/génétique , Marqueurs biologiques/sang , Réseaux de régulation génique , Thrombose veineuse/génétique , Thrombose veineuse/étiologie , Thrombose veineuse/diagnostic , Thromboembolisme veineux/génétique , Thromboembolisme veineux/étiologie , Thromboembolisme veineux/diagnostic , Thromboembolisme veineux/sang , Facteur de transcription GATA-3/génétique , Courbe ROC , Histone deacetylases/génétique , Apprentissage machine
5.
Sci Rep ; 14(1): 15113, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956421

RÉSUMÉ

The aims of this study were to determine whether human umbilical cord mesenchymal stem cells (hucMSCs) modified by miRNA-25-3p (miR-25-3p) overexpression could promote venous endothelial cell proliferation and attenuate portal endothelial cell injury. HucMSCs and human umbilical vein endothelial cells (HUVEC) were isolated and cultured from human umbilical cord and characterized. Lentiviral vectors expressing miRNA-25-3p were transfected into hucMSCs and confirmed by PCR. We verified the effect of miR-25-3p-modified hucMSCs on HUVEC by cell co-culture and cell supernatant experiments. Subsequently, exosomes of miR-25-3p-modified hucMSCs were isolated from cell culture supernatants and characterized by WB, NTA and TEM. We verified the effects of miR-25-3p-modified exosomes derived from hucMSCs on HUVEC proliferation, migration, and angiogenesis by in vitro cellular function experiments. Meanwhile, we further examined the downstream target genes and signaling pathways potentially affected by miR-25-3p-modified hucMSC-derived exosomes in HUVEC. Finally, we established a rat portal vein venous thrombosis model by injecting CM-DiR-labeled hucMSCs intravenously into rats and examining the homing of cells in the portal vein by fluorescence microscopy. Histological and immunohistochemical experiments were used to examine the effects of miRNA-25-3p-modified hucMSCs on the proliferation and damage of portal vein endothelial cells. Primary hucMSCs and HUVECs were successfully isolated, cultured and characterized. Primary hucMSCs were modified with a lentiviral vector carrying miR-25-3p at MOI 80. Co-culture and cell supernatant intervention experiments showed that overexpression of miRNA-25-3p in hucMSCs enhanced HUVEC proliferation, migration and tube formation in vitro. We successfully isolated and characterized exosomes of miR-25-3p-modified hucMSCs, and exosome intervention experiments demonstrated that miR-25-3p-modified exosomes derived from hucMSCs similarly enhanced the proliferation, migration, and angiogenesis of HUVECs. Subsequent PCR and WB analyses indicated PTEN/KLF4/AKT/ERK1/2 as potential pathways of action. Analysis in a rat portal vein thrombosis model showed that miR-25-3p-modified hucMSCs could homing to damaged portal veins. Subsequent histological and immunohistochemical examinations demonstrated that intervention with miR-25-3p overexpression-modified hucMSCs significantly reduced damage and attenuated thrombosis in rat portal veins. The above findings indicate suggest that hucMSCs based on miR-25-3p modification may be a promising therapeutic approach for use in venous thrombotic diseases.


Sujet(s)
Prolifération cellulaire , Exosomes , Cellules endothéliales de la veine ombilicale humaine , Cellules souches mésenchymateuses , microARN , Veine porte , microARN/génétique , microARN/métabolisme , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Rats , Exosomes/métabolisme , Exosomes/génétique , Veine porte/métabolisme , Mouvement cellulaire/génétique , Rat Sprague-Dawley , Mâle , Thrombose veineuse/génétique , Thrombose veineuse/métabolisme , Thrombose veineuse/anatomopathologie , Thrombose veineuse/thérapie , Cellules cultivées , Techniques de coculture , Transduction du signal , Cordon ombilical/cytologie
6.
J Am Heart Assoc ; 13(15): e034492, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39028040

RÉSUMÉ

BACKGROUND: Venous thromboembolism is associated with endothelial cell activation that contributes to the inflammation-dependent activation of the coagulation system. Cellular damage is associated with the release of different species of extracellular RNA (eRNA) involved in inflammation and coagulation. TLR3 (toll-like receptor 3), which recognizes (viral) single-stranded or double-stranded RNAs and self-RNA fragments, might be the receptor of these species of eRNA during venous thromboembolism. Here, we investigate how the TLR3/eRNA axis contributes to venous thromboembolism. METHODS AND RESULTS: Thrombus formation and size in wild-type and TLR3 deficient (-/-) mice were monitored by ultrasonography after venous thrombosis induction using the ferric chloride and stasis models. Mice were treated with RNase I, with polyinosinic-polycytidylic acid, a TLR3 agonist, or with RNA extracted from murine endothelial cells. Gene expression and signaling pathway activation were analyzed in HEK293T cells overexpressing TLR3 in response to eRNA or in human umbilical vein endothelial cells transfected with a small interference RNA against TLR3. Plasma clot formation on treated human umbilical vein endothelial cells was analyzed. Thrombosis exacerbated eRNA release in vivo and increased eRNA content within the thrombus. RNase I treatment reduced thrombus size compared with vehicle-treated mice (P<0.05). Polyinosinic-polycytidylic acid and eRNA treatments increased thrombus size in wild-type mice (P<0.01 and P<0.05), but not in TLR3-/- mice, by reinforcing neutrophil recruitment (P<0.05). Mechanistically, TLR3 activation in endothelial cells promotes CXCL5 (C-X-C motif chemokine 5) secretion (P<0.001) and NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation (P<0.05). Finally, eRNA triggered plasma clot formation in vitro (P<0.01). CONCLUSIONS: We show that eRNA and TLR3 activation enhance venous thromboembolism through neutrophil recruitment possibly through secretion of CXCL5, a potent neutrophil chemoattractant.


Sujet(s)
Modèles animaux de maladie humaine , Cellules endothéliales de la veine ombilicale humaine , Souris de lignée C57BL , Souris knockout , Infiltration par les neutrophiles , Récepteur de type Toll-3 , Thrombose veineuse , Animaux , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Thrombose veineuse/métabolisme , Thrombose veineuse/génétique , Thrombose veineuse/anatomopathologie , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Transduction du signal , Cellules HEK293 , Lésions du système vasculaire/métabolisme , Lésions du système vasculaire/génétique , Lésions du système vasculaire/anatomopathologie , Granulocytes neutrophiles/métabolisme , ARN/génétique , Mâle , Souris , Poly I-C/pharmacologie , Coagulation sanguine
7.
J Thromb Haemost ; 22(9): 2393-2403, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38908832

RÉSUMÉ

Venous thromboembolic disease (VTE) is a prevalent and potentially life-threatening vascular disease, including both deep vein thrombosis and pulmonary embolism. This review will focus on recent insights into the heritable factors that influence an individual's risk for VTE. Here, we will explore not only the discovery of new genetic risk variants but also the importance of functional characterization of these variants. These genome-wide studies should lead to a better understanding of the biological role of genes inside and outside of the canonical coagulation system in thrombus formation and lead to an improved ability to predict an individual's risk of VTE. Further understanding of the molecular mechanisms altered by genetic variation in VTE risk will be accelerated by further human genome sequencing efforts and the use of functional genetic screens.


Sujet(s)
Prédisposition génétique à une maladie , Variation génétique , Thromboembolisme veineux , Humains , Thromboembolisme veineux/génétique , Facteurs de risque , Étude d'association pangénomique , Coagulation sanguine/génétique , Embolie pulmonaire/génétique , Phénotype , Thrombose veineuse/génétique
8.
J Pharm Biomed Anal ; 248: 116290, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38878456

RÉSUMÉ

TIMP metallopeptidase inhibitor 3 (TIMP-3) may contribute to the pathogenesis of venous thromboembolism (VTE). However, few studies have investigated the effect of TIMP-3 on VTE. Therefore, a two-sample Mendelian randomization (MR) analysis was conducted to investigate the association between TIMP-3 levels and VTE. Seven independent single-nucleotide polymorphisms (SNPs) for TIMP-3 levels were obtained from a published genome-wide association study (the KORA Consortium, including 997 Europeans). We obtained outcome datasets for VTE, pulmonary embolism (PE), and deep vein thrombosis (DVT) from the FinnGen Consortium. The primary analytical method used in the MR analysis was the inverse variance weighted (IVW) method. To enhance the robustness of the MR results, some other MR methods including weighted median, MR-Egger, and MR-PRESSO were conducted. Moreover, several sensitivity analyses were performed to identify potential horizontal pleiotropy and heterogeneity. In primary IVW MR analyses, per log increase in genetically predicted TIMP-3 levels were positively associated with the incidence of VTE (odds ratio [OR], 1.03; 95 % confidence interval (CI), 1.01, 1.06; P = 0.010), PE (OR, 1.04; 95 % CI, 1.01, 1.08; P = 0.009), and DVT (OR, 1.06; 95 % CI, 1.02, 1.10; P= 0.003). The results of the weighted median, MR-Egger, and MR-PRESSO were similar to the main findings. No unbalanced pleiotropy or heterogeneity was observed. The study suggests that genetically predicted high levels of TIMP-3 may be associated with an increased risk of VTE. These findings indicate that strategies targeting TIMP-3 may provide a basis for the prevention and treatment of VTE. Further investigation is required to clarify this potential mechanism.


Sujet(s)
Étude d'association pangénomique , Analyse de randomisation mendélienne , Polymorphisme de nucléotide simple , Inhibiteur tissulaire de métalloprotéinase-3 , Thromboembolisme veineux , Humains , Analyse de randomisation mendélienne/méthodes , Thromboembolisme veineux/génétique , Thromboembolisme veineux/épidémiologie , Inhibiteur tissulaire de métalloprotéinase-3/génétique , Étude d'association pangénomique/méthodes , Prédisposition génétique à une maladie , Embolie pulmonaire/génétique , Embolie pulmonaire/épidémiologie , Embolie pulmonaire/sang , Facteurs de risque , Thrombose veineuse/génétique , Thrombose veineuse/épidémiologie
9.
J Thromb Haemost ; 22(9): 2531-2542, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38838952

RÉSUMÉ

BACKGROUND: The thrombin generation assay (TGA) evaluates the potential of plasma to generate thrombin over time, providing a global picture of an individual's hemostatic balance. OBJECTIVES: This study aimed to identify novel biological determinants of thrombin generation using a multiomics approach. METHODS: Associations between TGA parameters and plasma levels of 377 antibodies targeting 236 candidate proteins for cardiovascular risk were tested using multiple linear regression analysis in 770 individuals with venous thrombosis from the Marseille Thrombosis Association (MARTHA) study. Proteins associated with at least 3 TGA parameters were selected for validation in an independent population of 536 healthy individuals (Etablissement Français du Sang Alpes-Méditerranée [EFS-AM]). Proteins with strongest associations in both groups underwent additional genetic analyses and in vitro experiments. RESULTS: Eighteen proteins were associated (P < 1.33 × 10⁻4) with at least 3 TGA parameters in MARTHA, among which 13 demonstrated a similar pattern of associations in EFS-AM. Complement proteins C5 and C9 had the strongest associations in both groups. Ex vivo supplementation of platelet-poor plasma with purified C9 protein had a significant dose-dependent effect on TGA parameters. No effect was observed with purified C5. Several single nucleotide polymorphisms associated with C5 and C9 plasma levels were identified, with the strongest association for the C5 missense variant rs17611, which was associated with a decrease in C5 levels, endogenous thrombin potential, and peak in MARTHA. No association of this variant with TGA parameters was observed in EFS-AM. CONCLUSION: This study identified complement proteins C5 and C9 as potential determinants of thrombin generation. Further studies are warranted to establish causality and elucidate the underlying mechanisms.


Sujet(s)
Complément C5 , Complément C9 , Polymorphisme de nucléotide simple , Thrombine , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Marqueurs biologiques/sang , Coagulation sanguine , Tests de coagulation sanguine , Études cas-témoins , Complément C5/analyse , Modèles linéaires , Phénotype , Facteurs de risque , Thrombine/métabolisme , Thrombose veineuse/sang , Thrombose veineuse/génétique , Thrombose veineuse/immunologie , Complément C9/analyse
10.
Mol Med ; 30(1): 84, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867168

RÉSUMÉ

BACKGROUND: Deep vein thrombosis (DVT) is a common vascular surgical disease caused by the coagulation of blood in the deep veins, and predominantly occur in the lower limbs. Endothelial progenitor cells (EPCs) are multi-functional stem cells, which are precursors of vascular endothelial cells. EPCs have gradually evolved into a promising treatment strategy for promoting deep vein thrombus dissolution and recanalization through the stimulation of various physical and chemical factors. METHODS: In this study, we utilized a mouse DVT model and performed several experiments including qRT-PCR, Western blot, tube formation, wound healing, Transwell assay, immunofluorescence, flow cytometry analysis, and immunoprecipitation to investigate the role of HOXD9 in the function of EPCs cells. The therapeutic effect of EPCs overexpressing HOXD9 on the DVT model and its mechanism were also explored. RESULTS: Overexpression of HOXD9 significantly enhanced the angiogenesis and migration abilities of EPCs, while inhibiting cell apoptosis. Additionally, results indicated that HOXD9 specifically targeted the HRD1 promoter region and regulated the downstream PINK1-mediated mitophagy. Interestingly, intravenous injection of EPCs overexpressing HOXD9 into mice promoted thrombus dissolution and recanalization, significantly decreasing venous thrombosis. CONCLUSIONS: The findings of this study reveal that HOXD9 plays a pivotal role in stimulating vascular formation in endothelial progenitor cells, indicating its potential as a therapeutic target for DVT management.


Sujet(s)
Modèles animaux de maladie humaine , Progéniteurs endothéliaux , Protéines à homéodomaine , Mitophagie , Néovascularisation physiologique , Thrombose veineuse , Animaux , Progéniteurs endothéliaux/métabolisme , Souris , Thrombose veineuse/métabolisme , Thrombose veineuse/génétique , Thrombose veineuse/thérapie , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Mitophagie/génétique , Néovascularisation physiologique/génétique , Mouvement cellulaire , Mâle , Apoptose , Humains ,
11.
Thromb Res ; 240: 109044, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38824799

RÉSUMÉ

Protein C (PC), a vitamin K-dependent serine protease zymogen in plasma, can be activated by thrombin-thrombomodulin(TM) complex, resulting in the formation of activated protein C (APC). APC functions to downregulate thrombin generation by inactivating active coagulation factors V(FVa) and VIII(FVIIIa). Deficiency in PC increases the risk of venous thromboembolism (VTE). We have identified two unrelated VTE patients with the same heterozygous mutation (c.1384 T > C, p.Ter462GlnextTer17) in PROC. To comprehend the role of this mutation in VTE development, we expressed recombinant PC-Ter462GlnextTer17 in mammalian cells and evaluated its characteristics using established coagulation assay systems. Functional studies revealed a significant impairment in the activation of the mutant by thrombin or thrombin-TM complex. Furthermore, APC-Ter462GlnextTer17 demonstrated diminished hydrolytic activity towards the chromogenic substrate S2366. APTT and FVa degradation assays showed that both the anticoagulant activity of the mutant protein was markedly impaired, regardless of whether protein S was present or absent. These results were further supported by a thrombin generation assay conducted using purified and plasma-based systems. In conclusion, the Ter462GlnextTer17 mutation introduces a novel tail at the C-terminus of PC, leading to impaired activity in both PC zymogen activation and APC's anticoagulant function. This impairment contributes to thrombosis in individuals carrying this heterozygous mutation and represents a genetic risk factor for VTE.


Sujet(s)
Mutation , Protéine C , Thrombose veineuse , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Protéine C/métabolisme , Protéine C/génétique , Thrombose veineuse/génétique
12.
Eur J Med Res ; 29(1): 327, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38877527

RÉSUMÉ

BACKGROUND: Some previous observational studies have linked deep venous thrombosis (DVT) to thyroid diseases; however, the findings were contradictory. This study aimed to investigate whether some common thyroid diseases can cause DVT using a two-sample Mendelian randomization (MR) approach. METHODS: This two-sample MR study used single nucleotide polymorphisms (SNPs) identified by the FinnGen genome-wide association studies (GWAS) to be highly associated with some common thyroid diseases, including autoimmune hyperthyroidism (962 cases and 172,976 controls), subacute thyroiditis (418 cases and 187,684 controls), hypothyroidism (26,342 cases and 59,827 controls), and malignant neoplasm of the thyroid gland (989 cases and 217,803 controls. These SNPs were used as instruments. Outcome datasets for the GWAS on DVT (6,767 cases and 330,392 controls) were selected from the UK Biobank data, which was obtained from the Integrative Epidemiology Unit (IEU) open GWAS project. The inverse variance weighted (IVW), MR-Egger and weighted median methods were used to estimate the causal association between DVT and thyroid diseases. The Cochran's Q test was used to quantify the heterogeneity of the instrumental variables (IVs). MR Pleiotropy RESidual Sum and Outlier test (MR-PRESSO) was used to detect horizontal pleiotropy. When the causal relationship was significant, bidirectional MR analysis was performed to determine any reverse causal relationships between exposures and outcomes. RESULTS: This MR study illustrated that autoimmune hyperthyroidism slightly increased the risk of DVT according to the IVW [odds ratio (OR) = 1.0009; p = 0.024] and weighted median methods [OR = 1.001; p = 0.028]. According to Cochran's Q test, there was no evidence of heterogeneity in IVs. Additionally, MR-PRESSO did not detect horizontal pleiotropy (p = 0.972). However, no association was observed between other thyroid diseases and DVT using the IVW, weighted median, and MR-Egger regression methods. CONCLUSIONS: This study revealed that autoimmune hyperthyroidism may cause DVT; however, more evidence and larger sample sizes are required to draw more precise conclusions.


Sujet(s)
Étude d'association pangénomique , Analyse de randomisation mendélienne , Polymorphisme de nucléotide simple , Maladies de la thyroïde , Thrombose veineuse , Humains , Thrombose veineuse/génétique , Thrombose veineuse/épidémiologie , Analyse de randomisation mendélienne/méthodes , Maladies de la thyroïde/génétique , Maladies de la thyroïde/épidémiologie , Maladies de la thyroïde/complications , Prédisposition génétique à une maladie , Hyperthyroïdie/génétique , Hyperthyroïdie/complications
13.
Int J Hematol ; 120(2): 179-185, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38801563

RÉSUMÉ

Congenital antithrombin (AT) or serpin C1 deficiency, caused by a SERPINC1 abnormality, is a high-risk factor for venous thrombosis. SERPINC1 is prone to genetic rearrangement, because it contains numerous Alu elements. In this study, a Japanese patient who developed deep vein thrombosis during pregnancy and exhibited low AT activity underwent SERPINC1 gene analysis using routine methods: long-range polymerase chain reaction (PCR) and real-time PCR. Sequencing using long-range PCR products revealed no pathological variants in SERPINC1 exons or exon-intron junctions, and all the identified variants were homozygous, suggesting a deletion in one SERPINC1 allele. Copy number quantification for each SERPINC1 exon using real-time PCR revealed half the number of exon 1 and 2 copies compared with controls. Moreover, a deletion region was deduced by quantifying the 5'-upstream region copy number of SERPINC1 for each constant region. Direct long-range PCR sequencing with primers for the 5'-end of each presumed deletion region revealed a large Alu-mediated deletion (∼13 kb) involving SERPINC1 exons 1 and 2. Thus, a large deletion was identified in SERPINC1 using conventional PCR methods.


Sujet(s)
Déficit en antithrombine III , Antithrombine-III , Réaction de polymérisation en chaine en temps réel , Délétion de séquence , Humains , Femelle , Antithrombine-III/génétique , Déficit en antithrombine III/génétique , Adulte , Grossesse , Exons/génétique , Thrombose veineuse/génétique , Séquences Alu/génétique , Délétion de gène
14.
Neurology ; 102(11): e209445, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38759137

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Gene-gene interactions likely contribute to the etiology of multifactorial diseases such as cerebral venous thrombosis (CVT) and could be one of the main sources of known missing heritability. We explored Factor XI (F11) and ABO gene interactions among patients with CVT. METHODS: Patients with CVT of European ancestry from the large Bio-Repository to Establish the Aetiology of Sinovenous Thrombosis (BEAST) international collaboration were recruited. Codominant modelling was used to determine interactions between genome-wide identified F11 and ABO genes with CVT status. RESULTS: We studied 882 patients with CVT and 1,205 ethnically matched control participants (age: 42 ± 15 vs 43 ± 12 years, p = 0.08: sex: 71% male vs 68% female, p = 0.09, respectively). Individuals heterozygous (AT) for the risk allele (T) at both loci (rs56810541/F11 and rs8176645/ABO) had a 3.9 (95% CI 2.74-5.71, p = 2.75e-13) increase in risk of CVT. Individuals homozygous (TT) for the risk allele at both loci had a 13.9 (95% CI 7.64-26.17, p = 2.0e-15) increase in risk of CVT. The presence of a non-O blood group (A, B, AB) combined with TT/rs56810541/F11 increased CVT risk by OR = 6.8 (95% CI 4.54-10.33, p = 2.00e15), compared with blood group-O combined with AA. DISCUSSION: Interactions between factor XI and ABO genes increase risk of CVT by 4- to 14-fold.


Sujet(s)
Système ABO de groupes sanguins , Facteur XI , Thrombose veineuse , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Système ABO de groupes sanguins/génétique , Épistasie/génétique , Facteur XI/génétique , Galactosyltransferases , Prédisposition génétique à une maladie/génétique , Thrombose intracrânienne/génétique , Polymorphisme de nucléotide simple , Thrombose veineuse/génétique
15.
J Matern Fetal Neonatal Med ; 37(1): 2352089, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38812363

RÉSUMÉ

OBJECTIVE: Deep vein thrombosis (DVT) is a common complication in obstetrics that needs early interaction. The study examined the expression change and clinical value of long non-coding RNA (lncRNA) colorectal neoplasia differentially expressed (CRNDE) in DVT early diagnosis. METHODS: One hundred patients with DVT after delivery and 100 healthy parturients without DVT were enrolled. Serum samples were collected one day before delivery and received qRT-PCR for mRNA detection. Prenatal coagulation markers including prothrombin time (PT), activated partial prothrombin time (APTT), fibrinogen (FIB) and thrombin time (TT), D-dimer (D-D), thrombomodulin (TM), and peroxidase anti-peroxidase soluble complex (PAP) were tested. The receiver operating characteristic (ROC) curve was drawn for the diagnostic value assessment. RESULTS: LncRNA CRNDE levels increased remarkably in the serum of DVT patients compared with the healthy controls, which were negatively correlated with serum concentration of PT, APTT, and TT while positively correlated with FIB, D-D, TM, and PAP. Serum CRNDE (HR = 5.973, 95% CI = 2.990-11.933, p < .001) was independently related to the occurrence of DVT after delivery. Then, ROC curve using serum CRNDE showed a good diagnostic value for DVT with the AUC of 0.899. ROC curve of ultrasonography combined with CRNDE produced an AUC of 0.968, and both sensitivity and specificity were enhanced compared to a single indicator. CONCLUSIONS: The increase of CRNDE level was an independent risk factor for postpartum DVT. Prenatal ultrasonography combined with CRNDE can improve the predictive efficacy for DVT.


Sujet(s)
Valeur prédictive des tests , ARN long non codant , Échographie prénatale , Thrombose veineuse , Humains , Femelle , ARN long non codant/sang , Grossesse , Adulte , Thrombose veineuse/génétique , Thrombose veineuse/diagnostic , Thrombose veineuse/sang , Études cas-témoins , Période du postpartum/sang , Membre inférieur/vascularisation , Membre inférieur/imagerie diagnostique , Marqueurs biologiques/sang , Courbe ROC
16.
Ann Rheum Dis ; 83(9): 1132-1143, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-38609158

RÉSUMÉ

INTRODUCTION: Systemic lupus erythematosus with antiphospholipid syndrome (SLE-APS) represents a challenging SLE endotype whose molecular basis remains unknown. METHODS: We analysed whole-blood RNA-sequencing data from 299 patients with SLE (108 SLE-antiphospholipid antibodies (aPL)-positive, including 67 SLE-APS; 191 SLE-aPL-negative) and 72 matched healthy controls (HC). Pathway enrichment analysis, unsupervised weighted gene coexpression network analysis and machine learning were applied to distinguish disease endotypes. RESULTS: Patients with SLE-APS demonstrated upregulated type I and II interferon (IFN) pathways compared with HC. Using a 100-gene random forests model, we achieved a cross-validated accuracy of 75.6% in distinguishing these two states. Additionally, the comparison between SLE-APS and SLE-aPL-negative revealed 227 differentially expressed genes, indicating downregulation of IFN-α and IFN-γ signatures, coupled with dysregulation of the complement cascade, B-cell activation and neutrophil degranulation. Unsupervised analysis of SLE transcriptome identified 21 gene modules, with SLE-APS strongly linked to upregulation of the 'neutrophilic/myeloid' module. Within SLE-APS, venous thromboses positively correlated with 'neutrophilic/myeloid' and 'B cell' modules, while arterial thromboses were associated with dysregulation of 'DNA damage response (DDR)' and 'metabolism' modules. Anticardiolipin and anti-ß2GPI positivity-irrespective of APS status-were associated with the 'neutrophilic/myeloid' and 'protein-binding' module, respectively. CONCLUSIONS: There is a hierarchical upregulation and-likely-dependence on IFN in SLE with the highest IFN signature observed in SLE-aPL-negative patients. Venous thrombotic events are associated with neutrophils and B cells while arterial events with DDR and impaired metabolism. This may account for their differential requirements for anticoagulation and provide rationale for the potential use of mTOR inhibitors such as sirolimus and the direct fIIa inhibitor dabigatran in SLE-APS.


Sujet(s)
Syndrome des anticorps antiphospholipides , Analyse de profil d'expression de gènes , Lupus érythémateux disséminé , Transcriptome , Humains , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/sang , Syndrome des anticorps antiphospholipides/génétique , Syndrome des anticorps antiphospholipides/sang , Syndrome des anticorps antiphospholipides/complications , Femelle , Mâle , Analyse de profil d'expression de gènes/méthodes , Adulte , Adulte d'âge moyen , Études cas-témoins , Interférons , Thrombose veineuse/génétique , Thrombose veineuse/sang , Anticorps antiphospholipides/sang
17.
Curr Med Sci ; 44(2): 369-379, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38619683

RÉSUMÉ

OBJECTIVE: Matrix metalloproteinase 13 (MMP13) is an extracellular matrix protease that affects the progression of atherosclerotic plaques and arterial thrombi by degrading collagens, modifying protein structures and regulating inflammatory responses, but its role in deep vein thrombosis (DVT) has not been determined. The purpose of this study was to investigate the potential effects of MMP13 and MMP13-related genes on the formation of DVT. METHODS: We altered the expression level of MMP13 in vivo and conducted a transcriptome study to examine the expression and relationship between MMP13 and MMP13-related genes in a mouse model of DVT. After screening genes possibly related to MMP13 in DVT mice, the expression levels of candidate genes in human umbilical vein endothelial cells (HUVECs) and the venous wall were evaluated. The effect of MMP13 on platelet aggregation in HUVECs was investigated in vitro. RESULTS: Among the differentially expressed genes, interleukin 1 beta, podoplanin (Pdpn), and factor VIII von Willebrand factor (F8VWF) were selected for analysis in mice. When MMP13 was inhibited, the expression level of PDPN decreased significantly in vitro. In HUVECs, overexpression of MMP13 led to an increase in the expression level of PDPN and induced platelet aggregation, while transfection of PDPN-siRNA weakened the ability of MMP13 to increase platelet aggregation. CONCLUSIONS: Inhibiting the expression of MMP13 could reduce the burden of DVT in mice. The mechanism involves downregulating the expression of Pdpn through MMP13, which could provide a novel gene target for DVT diagnosis and treatment.


Sujet(s)
Thrombose veineuse , Animaux , Humains , Souris , Modèles animaux de maladie humaine , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Matrix Metalloproteinase 13/génétique , Agrégation plaquettaire , Thrombose veineuse/génétique
18.
J Thromb Thrombolysis ; 57(5): 815-827, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38643313

RÉSUMÉ

Venous thromboembolism (VTE) is a life-threatening haemostatic disease frequently diagnosed among the cancer population. The Khorana Score is currently the primal risk assessment model to stratify oncological patients according to their susceptibility to VTE, however, it displays a limited performance. Meanwhile, intensive research on VTE pathophysiology in the general population has uncovered a range of single-nucleotide polymorphisms (SNPs) associated with the condition. Nonetheless, their predictive ability concerning cancer-associated thrombosis (CAT) is controversial. Cervical cancer (CC) patients undergoing chemoradiotherapy often experience VTE, which negatively affects their survival. Thus, aiming for an improvement in thromboprophylaxis, new thrombotic biomarkers, including SNPs, are currently under investigation. In this study, the predictive capability of haemostatic gene SNPs on CC-related VTE and their prognostic value regardless of VTE were explored. Six SNPs in haemostatic genes were evaluated. A total of 401 CC patients undergoing chemoradiotherapy were enrolled in a retrospective cohort study. The implications for the time to VTE occurrence and overall survival (OS) were assessed. CAT considerably impacted the CC patients' OS (log-rank test, P < 0.001). SERPINE1 rs2070682 (T > C) showed a significant association with the risk of CC-related VTE (CC/CT vs. TT, log-rank test, P = 0.002; C allele, Cox model, hazard ratio (HR) = 6.99 and P = 0.009), while F2 rs1799963 (G > A) demonstrated an important prognostic value regardless of VTE (AA/AG vs. GG, log-rank test, P = 0.020; A allele, Cox model, HR = 2.76 and P = 0.026). For the remaining SNPs, no significant associations were detected. The polymorphisms SERPINE1 rs2070682 and F2 rs1799963 could be valuable tools in clinical decision-making, aiding in thromboprophylaxis and CC management, respectively.


Sujet(s)
Inhibiteur-1 d'activateur du plasminogène , Polymorphisme de nucléotide simple , Tumeurs du col de l'utérus , Thrombose veineuse , Humains , Femelle , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/complications , Adulte d'âge moyen , Inhibiteur-1 d'activateur du plasminogène/génétique , Thrombose veineuse/génétique , Thrombose veineuse/diagnostic , Thrombose veineuse/étiologie , Études rétrospectives , Thromboembolisme veineux/génétique , Thromboembolisme veineux/étiologie , Thromboembolisme veineux/diagnostic , Sujet âgé , Adulte , Chimioradiothérapie/effets indésirables , Pronostic , Appréciation des risques/méthodes , Hémostase/génétique
19.
Circ Res ; 134(10): e93-e111, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38563147

RÉSUMÉ

BACKGROUND: Endothelial activation promotes the release of procoagulant extracellular vesicles and inflammatory mediators from specialized storage granules. Endothelial membrane exocytosis is controlled by phosphorylation. We hypothesized that the absence of PTP1B (protein tyrosine phosphatase 1B) in endothelial cells promotes venous thromboinflammation by triggering endothelial membrane fusion and exocytosis. METHODS: Mice with inducible endothelial deletion of PTP1B (End.PTP1B-KO) underwent inferior vena cava ligation to induce stenosis and venous thrombosis. Primary endothelial cells from transgenic mice and human umbilical vein endothelial cells were used for mechanistic studies. RESULTS: Vascular ultrasound and histology showed significantly larger venous thrombi containing higher numbers of Ly6G (lymphocyte antigen 6 family member G)-positive neutrophils in mice with endothelial PTP1B deletion, and intravital microscopy confirmed the more pronounced neutrophil recruitment following inferior vena cava ligation. RT2 PCR profiler array and immunocytochemistry analysis revealed increased endothelial activation and adhesion molecule expression in primary End.PTP1B-KO endothelial cells, including CD62P (P-selectin) and VWF (von Willebrand factor). Pretreatment with the NF-κB (nuclear factor kappa B) kinase inhibitor BAY11-7082, antibodies neutralizing CD162 (P-selectin glycoprotein ligand-1) or VWF, or arginylglycylaspartic acid integrin-blocking peptides abolished the neutrophil adhesion to End.PTP1B-KO endothelial cells in vitro. Circulating levels of annexin V+ procoagulant endothelial CD62E+ (E-selectin) and neutrophil (Ly6G+) extracellular vesicles were also elevated in End.PTP1B-KO mice after inferior vena cava ligation. Higher plasma MPO (myeloperoxidase) and Cit-H3 (citrullinated histone-3) levels and neutrophil elastase activity indicated neutrophil activation and extracellular trap formation. Infusion of End.PTP1B-KO extracellular vesicles into C57BL/6J wild-type mice most prominently enhanced the recruitment of endogenous neutrophils, and this response was blunted in VWF-deficient mice or by VWF-blocking antibodies. Reduced PTP1B binding and tyrosine dephosphorylation of SNAP23 (synaptosome-associated protein 23) resulting in increased VWF exocytosis and neutrophil adhesion were identified as mechanisms, all of which could be restored by NF-κB kinase inhibition using BAY11-7082. CONCLUSIONS: Our findings show that endothelial PTP1B deletion promotes venous thromboinflammation by enhancing SNAP23 phosphorylation, endothelial VWF exocytosis, and neutrophil recruitment.


Sujet(s)
Exocytose , Souris knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Thrombose veineuse , Facteur de von Willebrand , Animaux , Protein Tyrosine Phosphatase, Non-Receptor Type 1/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 1/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 1/déficit , Humains , Souris , Facteur de von Willebrand/métabolisme , Facteur de von Willebrand/génétique , Thrombose veineuse/métabolisme , Thrombose veineuse/génétique , Thrombose veineuse/anatomopathologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Inflammation/métabolisme , Inflammation/génétique , Souris de lignée C57BL , Granulocytes neutrophiles/métabolisme , Cellules endothéliales/métabolisme , Cellules cultivées , Veine cave inférieure/métabolisme , Veine cave inférieure/anatomopathologie , Mâle , Infiltration par les neutrophiles , Facteur de transcription NF-kappa B/métabolisme
20.
Oncogene ; 43(21): 1631-1643, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38589675

RÉSUMÉ

Androgen deprivation therapy (ADT) is the first line of treatment for metastatic prostate cancer (PCa) that effectively delays the tumor progression. However, it also increases the risk of venous thrombosis event (VTE) in patients, a leading cause of mortality. How a pro-thrombotic cascade is induced by ADT remains poorly understood. Here, we report that protein disulfide isomerase A2 (PDIA2) is upregulated in PCa cells to promote VTE formation and enhance PCa cells resistant to ADT. Using various in vitro and in vivo models, we demonstrated a dual function of PDIA2 that enhances tumor-mediated pro-coagulation activity via tumor-derived extracellular vehicles (EVs). It also stimulates PCa cell proliferation, colony formation, and xenograft growth androgen-independently. Mechanistically, PDIA2 activates the tissue factor (TF) on EVs through its isomerase activity, which subsequently triggers a pro-thrombotic cascade in the blood. Additionally, TF-containing EVs can activate the Src kinase inside PCa cells to enhance the AR signaling ligand independently. Androgen deprivation does not alter PDIA2 expression in PCa cells but enhances PDIA2 translocation to the cell membrane and EVs via suppressing the clathrin-dependent endocytic process. Co-recruitment of AR and FOXA1 to the PDIA2 promoter is required for PDIA2 transcription under androgen-deprived conditions. Importantly, blocking PDIA2 isomerase activity suppresses the pro-coagulation activity of patient plasma, PCa cell, and xenograft samples as well as castrate-resistant PCa xenograft growth. These results demonstrate that PDIA2 promotes VTE and tumor progression via activating TF from tumor-derived EVs. They rationalize pharmacological inhibition of PDIA2 to suppress ADT-induced VTE and castrate-resistant tumor progression.


Sujet(s)
Évolution de la maladie , Tumeurs prostatiques résistantes à la castration , Protein Disulfide-Isomerases , Thrombose veineuse , Animaux , Humains , Mâle , Souris , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/effets indésirables , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Protein Disulfide-Isomerases/métabolisme , Protein Disulfide-Isomerases/génétique , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Thromboplastine/métabolisme , Thromboplastine/génétique , Thrombose veineuse/métabolisme , Thrombose veineuse/induit chimiquement , Thrombose veineuse/anatomopathologie , Thrombose veineuse/génétique , Thrombose veineuse/étiologie , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE