Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 217
Filtrar
1.
Int Angiol ; 41(6): 457-463, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36326144

RESUMEN

BACKGROUND: Vein-specific inflammation leads to vascular smooth muscle cells proliferation and extracellular matrix degradation of vein wall. This process is known as remodeling and is promoted by "trapped" leukocytes. Monocyte chemoattractant protein 1 (MCP-1) is a chemokine responsible for trafficking of leukocytes from blood to vein wall. The aim of this study was to measure the MCP-1 concentration in varicose veins blood before and after venoactive drug therapy and to compare it with a concentration of blood from varicose veins of subjects who did not receive drug treatment. METHODS: Non-randomized comparative study was conducted on 30 patients with primary varicose veins. 20 patients of the study group received diosmin 900 mg/hesperidin 100 mg once daily. 10 controls received no treatment. MCP-1 level was measured (pg/mL) in the blood from varicose veins twice, at the day of inclusion and after 60 days. Legs discomfort related to chronic venous disease (CVD) symptoms was measured with 10-cm Visual Analogue Scale (VAS) at inclusion and at completion of the study. RESULTS: Median (interquartile range, IQR) MCP-1 concentrations in treatment and control groups at inclusion were 171.9 (124.4-216.0) and 157.0 (120.1-163.1), resp., P=0.285. After 60 days of treatment MCP-1 level decreased, but non-significantly to 152.3 (124.1-178.3). In patients who did not receive treatment chemokine level slightly increased to 163.0 (134.0-172.9). Median changes over time were -6.6 (-30.9-7.4) and 10.6 (-3.7-19.2) in the study and control groups, resp. (P=0.048). After 60 days in 12 of 19 and 2 of 9 patients of treatments and control groups MCP-1 decreased (P=0.103). Odds ratio for MCP-1 decreasing was 9.5 (95% CI 1.1-81.5, P=0.043) for those who received venoactive drug. Mean (± standard deviation [SD]) legs discomfort significantly dropped in the study group from 5.7 (±2.5) to 1.9 (±2.2) (P=0.0003), while in controls no changes were registered: 3.4 (±1.3) and 3.5 (± 1.4), resp., P=0.28). Mean difference of VAS at baseline and at follow-up was -3.5 (±2.6) and 0.9 (±2.1), resp. (P<0.0001). CONCLUSIONS: Plasma concentration of MCP-1 in varicose veins blood demonstrates a tendency to decrease under two months treatment with a venoactive drug. Future studies are needed to reveal a possible role of MCP-1 as a target considering its role in varicose veins pathogenesis.


Asunto(s)
Quimiocina CCL2 , Várices , Humanos , Quimiocina CCL2/uso terapéutico , Várices/tratamiento farmacológico , Várices/metabolismo , Venas , Quimiocinas/uso terapéutico , Enfermedad Crónica
2.
Vascul Pharmacol ; 146: 107097, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35963524

RESUMEN

BACKGROUND: A chronic change in hemodynamic forces might activate the pathophysiological process of maladaptive venous remodeling. Biomechanical stretching stimulates venous smooth muscle cells (SMCs) in the media, and biomechanical loads exceeding physiological levels affect the intrinsic circadian rhythm and cellular phenotype. This study aimed to investigate the changes in the expression patterns of circadian clock genes under biomechanical stretching and their role in the regulation of the SMC phenotype. METHODS: Circadian genes were detected in venous specimens and venous SMCs from patients with varicose veins (VVs) and patients with autologous vein grafts (normal veins). Molecular mechanism studies of SMC phenotypic switching under biomechanical stretching were performed in human umbilical venous SMCs (HUVSMCs). RESULTS: CLOCK upregulation was observed in VVs. The circadian rhythm was disrupted in venous SMCs derived from VVs. In addition, CLOCK expression and cell proliferation and migration were increased in HUVSMCs exposed to biomechanical stretch. CLOCK overexpression activated NF-κB signaling and phenotypic transformation in HUVSMCs, whereas CLOCK depletion had inhibitory effects on these pathways. Further experiments revealed that the CLOCK protein regulates phenotypic and functional transformation via the RHOA/ROCK1 pathway. CONCLUSIONS: Our results demonstrate that CLOCK is a crucial regulator of the SMC phenotype under mechanical stretch. The CLOCK/RHOA/ROCK1 pathway is important in phenotypic adaptation, and targeting RHOA/ROCK1 could potentially reverse stretch-induced phenotypic switching.


Asunto(s)
Músculo Liso Vascular , Várices , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Proliferación Celular , Células Cultivadas , Humanos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Fenotipo , Regulación hacia Arriba , Várices/metabolismo , Quinasas Asociadas a rho/metabolismo
3.
Nat Commun ; 13(1): 3065, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35654884

RESUMEN

Varicose veins affect one-third of Western society, with a significant subset of patients developing venous ulceration, costing $14.9 billion annually in the USA. Current management consists of either compression stockings, or surgical ablation for more advanced disease. Most varicose veins patients report a positive family history, and heritability is ~17%. We describe the largest two-stage genome-wide association study of varicose veins in 401,656 individuals from UK Biobank, and replication in 408,969 individuals from 23andMe (total 135,514 cases and 675,111 controls). Forty-nine signals at 46 susceptibility loci were discovered. We map 237 genes to these loci, several of which are biologically plausible and tractable to therapeutic targeting. Pathway analysis identified enrichment in extracellular matrix biology, inflammation, (lymph)angiogenesis, vascular smooth muscle cell migration, and apoptosis. Using a polygenic risk score (PRS) derived in an independent cohort, we demonstrate its predictive utility and correlation with varicose veins surgery.


Asunto(s)
Estudio de Asociación del Genoma Completo , Várices , Movimiento Celular , Estudios de Cohortes , Matriz Extracelular/metabolismo , Humanos , Várices/genética , Várices/metabolismo , Várices/terapia
4.
Int J Med Sci ; 19(1): 186-194, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34975312

RESUMEN

Chronic Venous Disease (CVD) refers to a wide variety of venous disorders being the varicose veins its most common manifestation. It is well-established the link between pregnancy and the risk of suffering CVD, due to hormonal or haematological factors, especially during the third trimester. In the same manner, previous studies have demonstrated the detrimental effect of this condition in the placental tissue of pregnant women, including in the normal physiology and the metabolomic profile of this organ. In this context, the aim of this study was to evaluate the glucose homeostasis in the placental tissue of women presenting CVD. Through immunohistochemistry, we studied the protein expression of the glucose transporter 1 (GLUT-1), Phosphoglycerate kinase 1 (PGK1), aldolase (ALD), Glyceraldehyde-3-phosphate dehydrogenase (GA3PDH) and lactate dehydrogenase (LDH). Our results have reported a significative increase in the expression of GLUT-1, PGK1, ALD, GA3PDH and the isoenzyme LDHA in placentas of women with CVD. This work has proven for the first-time an altered glucose metabolism in the placental tissue of women affected by CVD, what may aid to understand the pathophysiological mechanisms of this condition in more distant organs such as placenta. Furthermore, our research also supports the basis for further studies in the metabolic phenotyping of the human placenta due to CVD, which may be considered during the late pregnancy in these women.


Asunto(s)
Glucólisis , Placenta/metabolismo , Complicaciones del Embarazo/metabolismo , Enfermedades Vasculares/metabolismo , Enfermedad Crónica , Femenino , Fructosa-Bifosfato Aldolasa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Humanos , Inmunohistoquímica , L-Lactato Deshidrogenasa/metabolismo , Fosfoglicerato Quinasa/metabolismo , Placenta/irrigación sanguínea , Embarazo , Várices/metabolismo
5.
Ann Vasc Surg ; 79: 335-347, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34648856

RESUMEN

BACKGROUND: Superficial thrombophlebitis (ST) is a frequent pathology, but its exact incidence remains to be determined. This study tested the hypothesis whether relationships exist among smooth muscle cells (SMCs) derived from ST, varicose great saphenous veins (VGSVs), and normal great saphenous veins (GSVs). METHODS: Forty-one samples of ST, VGSVs, and GSVs were collected. SMCs were isolated and cultured. Proliferation, migration, adhesion, and senescence in SMCs from the three vein walls were compared by various methods. Bax, Bcl-2, caspase-3, matrix metalloproteinase-2 (MMP-2), MMP-9, tissue inhibitor of metalloproteinase-1 (TIMP-1), and TIMP-2 messenger RNA (mRNA) and protein expressions were detected by fluorescence quantitative PCR and Western blot. RESULTS: An obvious decrease in cytoskeletal filaments was observed in thrombophlebitic vascular smooth muscle cells (TVSMCs). The quantity of proliferation, migration, adhesion, and senescence in TVSMCs was significantly higher than in varicose vascular smooth muscle cells and normal vascular smooth muscle cells (NVSMCs) (all P < 0.05). Bax and caspase-3 mRNA and protein expression were decreased, while Bcl-2 mRNA and protein expression were increased in the TVSMCs compared with the varicose vascular smooth muscle cells and the NVSMCs (all P < 0.05). MMP-2, MMP-9, TIMP-1, and TIMP-2 mRNA and protein expression were significantly increased in the TVSMCs compared with the VVGSVs and the NVSMCs (all P < 0.05). CONCLUSION: SMCs derived from ST are more dedifferentiated and demonstrate increased cell proliferation, migration, adhesion, and senescence, as well as obviously decreased cytoskeletal filaments. These results suggest that the phenotypic and functional differences could be related to the presence of atrophic and hypertrophic vein segments during the disease course among SMCs derived from ST, VGSVs, and GSVs.


Asunto(s)
Desdiferenciación Celular , Citoesqueleto/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Tromboflebitis/patología , Várices/patología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Estudios de Casos y Controles , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Senescencia Celular , Citoesqueleto/metabolismo , Femenino , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fenotipo , Vena Safena/metabolismo , Vena Safena/patología , Tromboflebitis/genética , Tromboflebitis/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Várices/genética , Várices/metabolismo
6.
Cells ; 10(12)2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34944071

RESUMEN

Chronic venous diseases, including varicose veins, are characterized by hemodynamic disturbances due to valve defects, venous insufficiency, and orthostatism. Veins are physiologically low shear stress systems, and how altered hemodynamics drives focal endothelial dysfunction and causes venous remodeling is unknown. Here we demonstrate the occurrence of endothelial to mesenchymal transition (EndMT) in human varicose veins. Moreover, the BMP4-pSMAD5 pathway was robustly upregulated in varicose veins. In vitro flow-based assays using human vein, endothelial cells cultured in microfluidic chambers show that even minimal disturbances in shear stress as may occur in early stages of venous insufficiency induce BMP4-pSMAD5-based phenotype switching. Furthermore, low shear stress at uniform laminar pattern does not induce EndMT in venous endothelial cells. Targeting the BMP4-pSMAD5 pathway with small molecule inhibitor LDN193189 reduced SNAI1/2 expression in venous endothelial cells exposed to disturbed flow. TGFß inhibitor SB505124 was less efficient in inhibiting EndMT in venous endothelial cells exposed to disturbed flow. We conclude that disturbed shear stress, even in the absence of any oscillatory flow, induces EndMT in varicose veins via activation of BMP4/pSMAD5-SNAI1/2 signaling. The present findings serve as a rationale for the possible use of small molecular mechanotherapeutics in the management of varicose veins.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Células Endoteliales/patología , Mesodermo/patología , Transducción de Señal , Proteína Smad5/metabolismo , Estrés Mecánico , Várices/metabolismo , Várices/patología , Adulto , Anciano , Biomarcadores/metabolismo , Células Endoteliales/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Neointima/patología , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Reología/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Adulto Joven
7.
J Vasc Surg Venous Lymphat Disord ; 9(1): 242-251, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32360331

RESUMEN

BACKGROUND: Lower limb venous varicosities (VVs) are clinically common; however, their molecular underpinnings are far from well elucidated. Previous studies have demonstrated that the phenotypic transition of vascular smooth muscle cells (VSMCs) plays a critical role in VV pathogenesis and that c-fos is upregulated in VSMCs from VVs. The present study investigated the histologic and cytologic changes in VVs and the correlation between c-fos upregulation and VSMC phenotypic switching. METHODS: Thirty-four patients with VVs (VV group) and 13 patients undergoing coronary artery bypass using autologous great saphenous vein segments (normal vein [NV] group) were enrolled in the present study. The great saphenous veins of both groups were harvested for subsequent experiments. Hematoxylin and eosin staining was performed for vein morphologic analysis. Real-time quantitative polymerase chain reaction, immunohistochemistry, and Western blot assays were used to assess mRNA and protein expression of c-fos, α-smooth muscle actin (α-SMA), and osteopontin (OPN). Simple linear regression was used to evaluate the correlation between c-fos and OPN/α-SMA. Primary VSMCs were isolated from both groups and cultured in vitro. A cell counting kit-8 assay and scratch-wound assay were used to analyze the proliferation and migration abilities of the cells, respectively. RESULTS: The mean age of the patients in the NV and VV groups was 61.4 ± 3.8 years and 59.5 ± 10.4 years, respectively. The vein cavities of the VV group were dilated, and the arrangement of the cells was disordered. The tunica media of the VV group was thicker than that of the NV group owing to the accumulation and proliferation of VSMCs. Significantly elevated mRNA levels of c-fos and OPN were observed in the VV group compared with the NV group, and a positive correlation was further demonstrated between the mRNA levels of c-fos and OPN/α-SMA (R2, 0.5524; P < .001). The VSMCs derived from the VV group were more numerous (as shown by the cell counting kit-8 assay) and had a significantly greater migration speed (as shown by the scratch-wound assay) than those derived from the NV group. Moreover, the protein expression of c-fos was significantly upregulated in VSMCs derived from the VV group, and this change was accompanied by a decrease in α-SMA and an increase in OPN expression. CONCLUSIONS: Both mRNA and protein expression of c-fos were upregulated in VV specimens, and the phenotypic biomarkers (OPN/α-SMA) were altered concurrently. VSMCs derived from VVs showed increased proliferation and migration abilities. Upregulation of c-fos might play a role in the phenotypic switching of VSMCs and subsequently participate in the pathogenesis of VVs. CLINICAL RELEVANCE: C-fos is an immediate early gene owing to the transient and rapid change in its expression in response to stimuli. It is involved in the regulation of cell proliferation, cell growth, and cell movement. In the present study, varicose vein specimens showed increased mRNA and protein expression of c-fos, accompanied by altered phenotypic biomarkers. The upregulation of the c-fos gene in smooth muscle cells cultured from varicose vein specimens might be associated with phenotypic switching and functional disturbance. These results could contribute to the exploration of the molecular mechanisms underlying the pathogenesis of varicose veins and the development of new therapeutic strategies.


Asunto(s)
Extremidad Inferior/irrigación sanguínea , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Várices/metabolismo , Actinas/genética , Actinas/metabolismo , Anciano , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/patología , Músculo Liso Vascular/cirugía , Miocitos del Músculo Liso/patología , Osteopontina/genética , Osteopontina/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-fos/genética , Transducción de Señal , Regulación hacia Arriba , Várices/genética , Várices/patología , Várices/cirugía , Venas/metabolismo , Venas/patología , Venas/cirugía
8.
J Vasc Surg Venous Lymphat Disord ; 9(2): 428-434, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32679199

RESUMEN

OBJECTIVE: The objective of this study was to analyze whether compression stocking therapy in the human varicose vein wall may change the levels of biomarkers associated with vein insufficiency. METHODS: Dilated collateral varicose vein samples were obtained from patients showing chronic venous disease (class 2 of the Clinical, Etiology, Anatomy, and Pathophysiology classification). Before elective surgery, 12 patients underwent compression stocking therapy (for 1 month) and 9 patients did not (control group). Expression levels of biomarkers associated with endothelial functionality (nitric oxide synthase 3), inflammation (interleukin-6, interleukin-10), oxidative stress (Gp91phox subunit of NADPH oxidase), and coagulation (factor Xa) were determined. P-selectin, an inflammatory and thrombosis-related biomarker, was also measured. RESULTS: Compression stockings increased the content of nitric oxide synthase 3 (control, 16.48 [16.04-17.40] AU; compression, 83.71 [67.70-91.85] AU; P < .001) in the varicose vein wall that was accompanied by reduction of both interleukin-6 levels (control, 38.72 [33.48-48.52] pg/µg protein; compression, 14.49 [11.05-17.41] pg/µg protein; P = .001) and the expression of Gp91phox subunit of NADPH oxidase (control, 63.24 [53.79-77.03] AU; compression, 36.85 [35.66-52.27] AU; P < .010). P-selectin (control, 77.37 [61.86-85.00] AU; compression, 54.31 [49.60-67.50] AU; P = .017) and factor Xa (control, 90.78 [75.02-100.00] AU; compression, 14.50 [13.77-36.20] AU; P < .001) were also reduced in the varicose vein wall of compression stocking-treated patients. However, P-selectin lost its statistical significance after adjustment by dyslipidemia. CONCLUSIONS: In the varicose vein wall, compression stocking therapy improved the content levels of biomarkers associated with endothelial functionality, inflammation, oxidative stress, and coagulation.


Asunto(s)
Coagulación Sanguínea , Mediadores de Inflamación/metabolismo , Estrés Oxidativo , Vena Safena/metabolismo , Medias de Compresión , Várices/terapia , Insuficiencia Venosa/terapia , Adulto , Biomarcadores/metabolismo , Factor Xa/metabolismo , Femenino , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , NADPH Oxidasa 2/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proyectos Piloto , Vena Safena/patología , Vena Safena/cirugía , Resultado del Tratamiento , Várices/metabolismo , Várices/patología , Procedimientos Quirúrgicos Vasculares , Insuficiencia Venosa/metabolismo , Insuficiencia Venosa/patología
9.
Vasc Med ; 25(6): 588-597, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32877311

RESUMEN

Fibronectin (FN) circulating in the blood and produced by cells provides the basis of the extracellular matrix (ECM) formed in healing acute wounds. The time-dependent deposition of FN by macrophages, its synthesis by fibroblasts and myofibroblasts, and later degradation in the remodeled granulation tissue are a prerequisite for successful healing of wounds. However, the pattern of FN expression and deposition in skin lesions is disturbed. The degradation of the ECM components including FN in varicose veins prevails over ECM synthesis and deposition. FN is inconspicuous in the fibrotic lesions in lipodermatosclerosis, while tenascin-C containing FN-like peptide sequences are prominent. FN is produced in large amounts by fibroblasts at the edge of venous ulcers but FN deposition at the wound bed is impaired. Both the proteolytic environment in the wounds and the changed function of the ulcer fibroblasts may be responsible for the poor healing of venous ulcers. The aim of this review is to describe the current knowledge of FN pathophysiology in chronic venous diseases. In view of the fact that FN plays a crucial role in organizing the ECM, further research focused on FN metabolism in venous diseases may bring results applicable to the treatment of the diseases.


Asunto(s)
Fibronectinas/metabolismo , Várices/metabolismo , Venas/metabolismo , Insuficiencia Venosa/metabolismo , Animales , Enfermedad Crónica , Dermatitis/metabolismo , Dermatitis/patología , Humanos , Esclerodermia Localizada/metabolismo , Esclerodermia Localizada/patología , Transducción de Señal , Várices/patología , Várices/fisiopatología , Venas/patología , Venas/fisiopatología , Insuficiencia Venosa/patología , Insuficiencia Venosa/fisiopatología
10.
J Cell Physiol ; 235(12): 9538-9556, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32529639

RESUMEN

Despite the high prevalence of varicose veins, the underlying pathogenesis of this disease remains unclear. The present study aims to explore the role of insulin-like growth factor binding protein 6 (IGFBP6) in vascular smooth muscle cells (VSMCs). Using a protein array approach, we identified several differentially expressed proteins between varicose great saphenous veins and normal great saphenous veins. Bioinformatic analysis showed that IGFBP6 was closely related to cell proliferation. Further validation confirmed that IGFBP6 was one of the most highly expressed proteins in varicose vein tissue. Knocking down IGFBP6 in VSMCs significantly attenuated cell proliferation and induced the S phase arrest during the cell cycle. Further experiments demonstrated that IGFBP6 knockdown increased cyclin E ubiquitination, which reduced expression of cyclin E and phosphorylation of CDK2. Furthermore, IGFBP6 knockdown arrested centrosome replication, which subsequently influenced VSMC morphology. Ultimately, IGFBP6 was validated to be involved in VSMC proliferation in varicose vein tissues. The present study reveals that IGFBP6 is closely correlated with VSMC biological function and provides unprecedented insights into the underlying pathogenesis of varicose veins.


Asunto(s)
Ciclina E/genética , Quinasa 2 Dependiente de la Ciclina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteoma/genética , Proliferación Celular/genética , Femenino , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Análisis por Matrices de Proteínas , Vena Safena/metabolismo , Várices/metabolismo , Várices/patología
11.
Int J Mol Sci ; 21(7)2020 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-32260158

RESUMEN

Pregnancy is a period in a woman's life associated with an increased risk of developing lower extremity chronic venous disease (CVD). Pregnancy-associated CVD is associated with changes in placental villi. We investigated angiogenesis and lymphangiogenesis in the placental villi of women with CVD during pregnancy compared with healthy controls with no history of CVD (HC). An observational, analytical, and prospective cohort study was conducted on 114 women in their third trimester of pregnancy (32 weeks). Sixty-two participants were clinically diagnosed with CVD. In parallel, 52 controls with no history of CVD (HC) were studied. Gene and protein expression of CD31, podoplanin (D2-40), Flt-1, and placental growth factor (PIGF) was analysed by real-time polymerase chain reaction (RT-qPCR) and immunohistochemistry. CD31 and D2-40 gene expression was significantly greater in the placental villi of women with CVD, as were the numbers of vessels positive for CD31 and D2-40. Significantly higher gene and protein expression of Flt-1 and PIGF was observed in the placental villi of women with CVD. Histological analysis showed more placental villi with periodic acid of Schiff (PAS)-positive material in women with CVD. Our results show a connection between pregnancy-associated CVD and leading to higher proangiogenic and lymphangiogenic activity in placental villi.


Asunto(s)
Vellosidades Coriónicas/metabolismo , Glicoproteínas de Membrana/genética , Factor de Crecimiento Placentario/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Regulación hacia Arriba , Várices/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Adulto , Biomarcadores/metabolismo , Estudios de Casos y Controles , Femenino , Edad Gestacional , Humanos , Edad Materna , Glicoproteínas de Membrana/metabolismo , Factor de Crecimiento Placentario/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Complicaciones Cardiovasculares del Embarazo , Estudios Prospectivos , Várices/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
13.
Heart Vessels ; 35(1): 1-13, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31227876

RESUMEN

Recent findings have suggested that the primary factors for development of chronic venous disease (CVD), which commonly manifests as varicose veins (VV), are due to structural and biochemical modifications of the vessel wall. The aim of this exploratory study was to characterize by flow cytometry the endothelial cells (EC) mechanically extracted from the varicose saphenous veins (VSV) segments of patients submitted to VV surgery, and to compare the expression of cell surface molecules in these EC with that observed in the EC from the graft SV (GSV) of patients undergoing bypass surgery. EC were isolated from distal- (varicose trunk) and from proximal- (nearly normal) VSV segments of 30 patients submitted to VV surgery, and from proximal GSV segments of 20 patients submitted to bypass surgery (control group), using a mechanical method, and their immunophenotype was characterized by flow cytometry. EC were identified as being CD45negCD146brightCD31bright, and analyzed for expression of activation-related (CD54, CD62E, CD106), procoagulant (CD142), and cell junction (CD31, CD146) molecules, and for the scavenger receptor, CD36. The EC harvested from the SV segments of CVD patients had lower expression of all the molecules evaluated, in comparison to controls; these differences were more evident for the EC isolated from the distal-VSV. The EC extracted from the proximal- and distal-VSV segments of the CVD patients also differ from each other, the first having lower levels of CD62E, CD106, CD142 and CD36. Groups did not match for gender and controls were heterogeneous concerning the underlying pathologies, which may have a confounding effect. Our study revealed that the EC isolated from varicose (distal) and nearly normal (proximal) VSV segments of the CVD patients differ phenotypically from each other, and from the EC of the control group. The VSV segments more affected by the CVD have the lowest expression of the studied markers. We hypothesize that CVD is associated with a decrease on the EC surface molecules, causing EC dysfunctionality. Further studies with a large number of gender-matched participants are needed, to confirm the results obtained in this exploratory study.


Asunto(s)
Antígenos CD/metabolismo , Células Endoteliales/metabolismo , Citometría de Flujo , Vena Safena/metabolismo , Várices/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Estudios de Casos y Controles , Enfermedad Crónica , Células Endoteliales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Vena Safena/patología , Vena Safena/cirugía , Várices/patología , Várices/cirugía
14.
Int Angiol ; 38(3): 185-193, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31058480

RESUMEN

BACKGROUND: Varicose great saphenous veins (VGSVs) are a common disorder with a high incidence, but the pathogenesis is unclear. This study was designed to measure the changes in matrix metalloproteinases (MMPs) and the tissue inhibitor of metalloproteinases (TIMPs) in different segments from VGSV walls to determine the relationship between MMPs, TIMPs expression, and expansion of the venous wall. METHODS: Twenty-one VGSV and 12 normal great saphenous vein (GSV) specimens were collected. Venous walls in the two groups, expression of MMP-2, MMP-9, TIMP-1, and TIMP-2 proteins, protein-positive expression ratios, mRNA expression, and protein content were determined by immunohistochemistry, PCR, and western blot. RESULTS: The MMP-2, MMP-9, TIMP-1, and TIMP-2 protein-positive expression ratios, mRNA expression in the upper, middle, and lower segments in the VGSV group were significantly higher than the corresponding regions in the GSV group, respectively. The MMP-2, MMP-9, TIMP-1, and TIMP-2 protein-positive expression ratios, mRNA expression, and protein concentrations in the lower segments in the VGSV group were also significantly higher than the upper and middle segments in the VGSV group and the corresponding regions in the GSV group, respectively. CONCLUSIONS: Under high hemodynamics, disequilibrium of MMPs and TIMPs from VGSVs exists within the upper, middle, and lower segments of VGSVs. These results suggested that MMPs and TIMPs participate in the process of venous wall remodeling and may be one of the mechanisms associated with the formation and development in varicose veins.


Asunto(s)
Metaloproteinasas de la Matriz/metabolismo , Vena Safena/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Várices/metabolismo , Anciano , Femenino , Humanos , Masculino , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Persona de Mediana Edad , ARN Mensajero/metabolismo , Vena Safena/patología , Inhibidor Tisular de Metaloproteinasa-1 , Inhibidor Tisular de Metaloproteinasa-2 , Várices/patología
15.
Scand J Surg ; 108(1): 67-75, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30319041

RESUMEN

BACKGROUND:: The saphenous vein is the most frequently used bypass conduit for vascular reconstructions, which may need to be stored for a prolonged time. The aim of this study was to compare the effect of different preservation solutions on the morphology of saphenous veins during the long-term cold storage. DESIGN:: An in vitro study. MATERIAL AND METHODS:: Saphenous vein samples, collected from 29 patients undergoing varicose vein surgery, were stored at +4°C in (1) 10% formalin, (2) isotonic saline with heparin and antibiotic, (3) phosphate-buffered saline, (4) 2.5% glutaraldehyde + phosphate-buffered saline, and (5) Custodiol (histidine-tryptophan-ketoglutarate). Changes in the vein wall were histologically investigated up to day 35. Possible retention of the capacity of endothelial function was evaluated by immunohistochemical detection of endothelial nitric oxide synthase. RESULTS:: Formalin as the control medium well preserved the vein wall morphology, but endothelial nitric oxide synthase immunostaining was very weak. Phosphate-buffered saline and isotonic saline with heparin and antibiotic poorly preserved vein wall morphology. Phosphate-buffered saline endothelial nitric oxide synthase staining decreased dramatically throughout the study period. Compared to phosphate-buffered saline, stronger isotonic saline with heparin and antibiotic endothelial nitric oxide synthase staining was noted at day 35 (p < 0.001). Custodiol and glutaraldehyde better preserved vein morphology compared to ISHA and PBS at day 5 and later (p < 0.001), but compared to stronger isotonic saline with heparin and antibiotic their endothelial nitric oxide synthase staining was weaker. CONCLUSION:: In terms of preserving the morphology of saphenous veins, phosphate-buffered saline and isotonic saline with heparin and antibiotic were the poorest, while Custodiol and glutaraldehyde were the best. Demonstrating good retention of endothelial nitric oxide synthase staining throughout the study period, isotonic saline with heparin and antibiotic seems to have the best potential to retain vein wall functionality, despite relatively poor morphological preservation.


Asunto(s)
Aloinjertos , Fármacos Cardiovasculares/administración & dosificación , Soluciones Farmacéuticas/administración & dosificación , Vena Safena , Conservación de Tejido/métodos , Aloinjertos/efectos de los fármacos , Aloinjertos/metabolismo , Aloinjertos/patología , Frío , Humanos , Inmunohistoquímica , Técnicas In Vitro , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Distribución Aleatoria , Vena Safena/efectos de los fármacos , Vena Safena/metabolismo , Vena Safena/patología , Factores de Tiempo , Várices/metabolismo , Várices/patología , Várices/cirugía
16.
J Cell Biochem ; 120(6): 10031-10042, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30556158

RESUMEN

In varicose veins, vascular smooth muscle cells (VSMCs) often show abnormal proliferative and migratory rates and phenotypic transition. This study aimed to investigate whether microRNA (miR)-202 and its potential target, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), were involved in VSMC phenotypic transition. miR-202 expression was analyzed in varicose veins and in VSMCs conditioned with platelet-derived growth factor. The effect of miR-202 on cell proliferation and migration was assessed. Furthermore, contractile marker SM-22α, synthetic markers vimentin and collagen I, and PGC-1α were analyzed by Western blot analysis. The modulation of PGC-1α expression by miR-202 was also evaluated. In varicose veins and proliferative VSMCs, miR-202 expression was upregulated, with decreased SM-22α expression and increased vimentin and collagen I expression. Transfection with a miR-202 mimic induced VSMC proliferation and migration, whereas a miR-202 inhibitor reduced cell proliferation and migration. miR-202 mimic constrained luciferase activity in HEK293 cells that were cotransfected with the PGC-1α 3'-untranslated region (3'-UTR) but not those with mutated 3'-UTR. miR-202 suppressed PGC-1α protein expression, with no influence on its messenger RNA expression. PGC-1α mediated VSMC phenotypic transition and was correlated with reactive oxygen species production. In conclusion, miR-202 affects VSMC phenotypic transition by targeting PGC-1α expression, providing a novel target for varicose vein therapy.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/biosíntesis , Várices/metabolismo , Regiones no Traducidas 3' , Anciano , Anciano de 80 o más Años , Femenino , Células HEK293 , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Várices/genética , Várices/patología
17.
Oxid Med Cell Longev ; 2018: 3974026, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30250632

RESUMEN

In chronic venous insufficiency of the lower limbs, data show that the clinical manifestation is varicose veins (VVs), and VV epidemiology suggests that sex hormones directly influence disease development through intracellular receptors. This study aimed to determine the presence and localization of oestrogen receptors (ERs), progesterone receptors (PRs), and androgen receptors (ARs) in both healthy and VV wall cells and their relationship with gender. In this study, samples from patients without a history of venous disease (CV) (n = 18) and with VV (n = 40) were used. The samples were divided by gender: CV women (CVw) = 6, CV men (CVm) = 12, VV women (VVw) = 25, and VV men (VVm) = 15. RT-qPCR and immunohistochemical techniques were performed, and increased ER and PR protein expression was found in VVw in all tunica layers. ARs were localized to the adventitial layer in the CV and were found in the neointima in VVs. mRNA expression was increased for ER and PR in VVw. AR gene expression was significantly decreased in VVm. The increase in the number of these receptors and their redistribution through the wall reinforces the role of sex hormones in varicose vein development.


Asunto(s)
Premenopausia/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Várices/metabolismo , Várices/fisiopatología , Remodelación Vascular , Adulto , Recuento de Células , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/genética , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Várices/genética
18.
Adv Clin Exp Med ; 27(5): 695-701, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29616754

RESUMEN

BACKGROUND: Venous insufficiency is still a serious clinical problem. The exact cause and molecular mechanisms of this disease are still unknown. In this study, we try to identify whether there is a difference in the level of trace elements between healthy and pathological veins. Our results show that insufficient veins have different levels of some trace elements: magnesium, calcium, manganese, and silicon compared to control samples. This study could lead to a better understanding of the molecular causes of venous insufficiency and may help to develop better methods of treatment. OBJECTIVES: Nowadays, venous diseases are a very common clinical phenomenon. Venous insufficiency is thought to be one of the most common vein diseases. The exact mechanisms of its etiology are still unknown, although from a clinical point of view some risk factors include gender, age, changing hormone levels, heredity, and standing or sitting for long periods. An imbalance in trace elements could also play a crucial role in the development and/or progression of venous insufficiency. MATERIAL AND METHODS: The trace element content in varicose vein walls and in normal vein walls was measured using an inductively coupled plasma-optical emission spectrometer (ICP-OES) after sample mineralization. Statistical analysis (the Mann-Whitney U test and the Friedman ANOVA) was performed to compare insufficient veins to controls (healthy veins). RESULTS: This study found statistically significant higher magnesium (Mg) ion levels in varicose veins compared to controls (p = 0.0067) and differences close to statistical significance in calcium (Ca), manganese (Mn), and silicon (Si) ion levels. CONCLUSIONS: The results obtained could indicate oxidative stress occurring in chronic venous insufficiency as well as free radical neutralization pathways due to superoxide dismutase (SOD) activity with Mg, Mn and copper (Cu) ion involvement. Our results are consistent with literature data and are preliminary in nature.


Asunto(s)
Oligoelementos/metabolismo , Várices/metabolismo , Insuficiencia Venosa/etiología , Cobre/metabolismo , Humanos , Magnesio/metabolismo , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Insuficiencia Venosa/metabolismo , Zinc/metabolismo
19.
Ann Vasc Surg ; 46: 380-393, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28688874

RESUMEN

Chronic venous disease is a potentially prevalent and debilitating condition affecting millions of individuals, mostly in Western world. Predisposing genetic and environmental factors contribute to its development. However, the main etiology remains to be elucidated. An extensive literature search was conducted in Medline using the following key words algorithm: ("Chronic venous disease" OR "Chronic venous insufficiency" OR "varicose veins") AND ("endothelial dysfunction" OR "inflammation"). Besides being a multifactorial disease, it is now recognized that the hallmark of chronic venous disease pathophysiology likely remains in inflammation, possibly triggered by sustained venous hypertension and valvular incompetence. Shear stress changes are directly sensed by endothelial cells, leading to its activation and subsequent recruitment of leukocytes and release of proinflammatory agents. Dysfunctional endothelium has a pivotal role perpetuating the inflammatory cascade, with consequent pathological venous changes and chronic venous disease worsening. Endothelial dysfunction may be the central player in the link between varicose veins and deep vein thrombosis. In this article, we aim to analyze the crucial role of endothelial activation in the persistent inflammatory cycle that characterizes chronic venous disease.


Asunto(s)
Endotelio Vascular/fisiopatología , Hemodinámica , Inflamación/fisiopatología , Várices/fisiopatología , Remodelación Vascular , Insuficiencia Venosa/fisiopatología , Animales , Enfermedad Crónica , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Glicocálix/inmunología , Glicocálix/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Rodamiento de Leucocito , Leucocitos/inmunología , Leucocitos/metabolismo , Factores de Riesgo , Várices/inmunología , Várices/metabolismo , Insuficiencia Venosa/inmunología , Insuficiencia Venosa/metabolismo
20.
Br J Surg ; 105(1): 59-67, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29143961

RESUMEN

BACKGROUND: Leg ulceration is a feared complication of venous insufficiency. It is not known whether varicose veins predispose skin to poor wound healing. The expression pattern of gap junctional protein connexin, a known marker of poor wound healing, was investigated across various stages of venous disease. METHODS: Patients undergoing intervention for varicose veins were assessed according to the Clinical Etiologic Anatomic Pathophysiologic (CEAP) classification of varicose veins. Paired 4-mm punch biopsies were taken from above the ankle (pathological) and above the knee (control). Tissues were stained with haematoxylin and eosin, and for connexin 43, connexin 30 and connexin 26. RESULTS: Forty-eight paired biopsies were taken (12 each for CEAP class C0, C2, C4 and C6). The pathological skin showed progressive epithelial hyperthickening, an increase in the number and depth of rete ridges, increased inflammation and loss of dermal architecture with disease progression from C4 onwards. The overall absolute connexin expression and mean connexin expression per cell in the pathological skin similarly increased across the CEAP classes from as early as C2. Increasing levels of connexin in control skin were also noted, indicating progression of the disease proximally. Connexin 43 expression showed the strongest positive correlation between pathological and control skin. CONCLUSION: Connexins were overexpressed in patients with simple varicose veins, with a stepwise increased expression through venous eczema to ulceration. Connexin 43 is a potential biomarker for venous disease. This finding suggests that varicose veins predispose skin to poor wound healing. Surgical relevance The overexpression of connexins, a family of gap junctional proteins, is known to cause poor healing in venous leg ulceration. It is not known whether there is any association with superficial venous disease. Here, connexin proteins were overexpressed in patients with uncomplicated varicose veins, before histological skin changes. Connexin could be a biomarker of venous disease progression.


Asunto(s)
Conexinas/metabolismo , Piel/metabolismo , Regulación hacia Arriba , Várices/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Biopsia , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Microscopía Confocal , Persona de Mediana Edad , Piel/patología , Várices/patología , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...