Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Endovasc Ther ; : 15266028241245325, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38616626

RESUMEN

PURPOSE: The purpose was to evaluate the clinical outcomes of a dedicated venous stent with the tripartite composite segments for the treatment of iliofemoral venous obstruction (IVO) in a mixed cohort of nonthrombotic iliac vein lesion (NIVL) and post-thrombotic syndrome (PTS) over a period of 12 months. METHODS: The Grency Trial is a prospective, multicenter, single-arm, open-label, pivotal study, which was conducted at 18 large tertiary hospitals in China from August 2019 to October 2020. A total of 133 hospitalized patients were screened and 110 patients with clinical, etiology, anatomical, and pathophysiology clinical class (CEAP) clinical grade C>3 and iliac vein stenosis >50% or occlusion, including 72 patients with NIVL and 38 patients with PTS, were implanted with Grency venous stents. Primary endpoint was stent patency at 12 months follow-up, and secondary outcomes were technical success; improvement in venous clinical severity score (VCSS) at 3, 6, and 12 month follow-up; and rates of clinical adverse events. RESULTS: Among 110 patients who were implanted with Grency venous stents, 107 patients completed the 12 month follow-up. All 129 stents were successfully implanted in 110 limbs. Twelve-month primary patency rate was 94.39% [95% confidence interval [CI]=88.19%-97.91%] overall, and 100% [94.94%-100%] and 83.33% [67.19%-93.63%] in the NIVL and PTS subgroups, respectively. Venous clinical severity score after iliac vein stenting improved significantly up to 12 months follow-up. There were 3 early major adverse events (1 intracerebral hemorrhage and 2 stent thrombosis events related to anticoagulation therapy), and 7 late major adverse events (1 cardiovascular death, 1 intracranial hemorrhage with uncontrolled hypertension, and 5 in-stent restenosis cases without stent fractures or migration). CONCLUSIONS: The Grency venous stent system appeared excellent preliminary safe and effective for IVO treatment. Further large-scale studies with longer-term follow-up are needed to evaluate long-term patency and durability of stent. CLINICAL IMPACT: The design of venous stents for iliofemoral venous obstruction (IVO) must address engineering challenges distinct from those encountered in arterial stenting. The Grency venous stent, a nitinol self-expanding stent specifically tailored for IVO, features a composite structure designed to meet the stent requirements of various iliac vein segments. The Grency Trial is a prospective, multicenter, single-arm, open-label pivotal study aimed at evaluating the efficacy and safety of the Grency stent system. Following a 12-month follow-up period, the Grency venous stent system has demonstrated both safety and efficacy in treating iliofemoral venous outflow obstruction.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38294627

RESUMEN

In this study, we investigated the protective role of Mzb1 in atherosclerotic plaque vulnerability. To explore the impact of Mzb1, we analyzed Mzb1 expression, assessed apoptosis, and evaluated mitochondrial function in atherosclerosis (AS) mouse models and human vascular smooth muscle cells (HVSMCs). We observed a significant decrease in Mzb1 expression in AS mouse models and ox-LDL-treated HVSMCs. Downregulation of Mzb1 increased ox-LDL-induced apoptosis and cholesterol levels of HVSMCs, while Mzb1 overexpression alleviated these effect. Mzb1 was found to enhance mitochondrial function, as evidenced by restored ATP synthesis, mitochondrial membrane potential, and reduced mtROS production. Moreover, Mzb1 overexpression attenuated atherosclerotic plaque vulnerability in ApoE-/- mice. Our findings suggest that Mzb1 overexpression regulates the AMPK/SIRT1 signaling pathway, leading to the attenuation of atherosclerotic plaque vulnerability. This study provides compelling evidence for the protective effect of Mzb1 on atherosclerotic plaques by alleviating apoptosis and modulating mitochondrial function in ApoE-/- mice.

3.
Pathol Res Pract ; 253: 154969, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38029715

RESUMEN

Endoplasmic reticulum (ER) stress refers to a condition where the normal functioning of the ER is disrupted due to a variety of cellular stress factors. As a result, there is an accumulation of unfolded and misfolded proteins within the ER. Numerous studies have shown that ER stress can exacerbate inflammatory reactions and contribute to the development of various inflammatory diseases. However, the role of ER stress in the stability of atherosclerotic plaques remains poorly understood. In this study, we aimed to explore the potential impact of a specific ER stress inhibitor known as 4-phenyl butyric acid (4-PBA) on atherosclerosis in mice. The mice were fed a high-fat diet, and treatment with 4-PBA significantly improved the stability of the atherosclerotic plaques. This was evidenced by a reduction in oxidative stress and an increase in circadian locomotor output cycles kaput (CLOCK) protein and mRNA expression within the plaques. Additionally, 4-PBA reduced the expression of ER stress-related proteins and decreased apoptosis in the atherosclerotic plaques. In vitro investigation, we observed the effect of 4-PBA on vascular smooth muscle cells (VSMCs) that were exposed to oxidized low-density lipoprotein (ox-LDL), a significant contributor to the development of atherosclerosis. 4-PBA reduced reactive oxygen species (ROS) production and attenuated apoptosis, GRP78 and CHOP protein expression in ox-LDL-Induced VSMCs via up-regulating CLOCK expression. However, when the short hairpin RNA against CLOCK (sh-CLOCK) was introduced to the VSMCs, the protective effect of 4-PBA was abolished. This suggests that the up-regulation of CLOCK expression is crucial for the beneficial effects of 4-PBA on atherosclerotic plaque stability. This finding suggests that targeting ER stress and modulating CLOCK protein levels might be a promising way to enhance the stability of atherosclerotic plaques.


Asunto(s)
Aterosclerosis , Butilaminas , Placa Aterosclerótica , Animales , Ratones , Proteínas CLOCK/farmacología , Aterosclerosis/metabolismo , Apoptosis , Estrés del Retículo Endoplásmico
4.
Exp Cell Res ; 427(2): 113587, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37044315

RESUMEN

BACKGROUND: Vascular smooth muscle cell (VSMC) phenotypic switching contributes to VSMC proliferation and migration in atherosclerosis (AS). Nevertheless, the regulatory mechanism of VSMC phenotypic switching during AS progression is unclear. Here, the role and regulatory mechanism of UCHL5 in VSMC phenotypic switching during AS progression were investigated. METHODS: ApoE-/- mice were fed with high fat diet to establish AS model in vivo. VSMCs stimulated by ox-LDL were used as AS cellular model. VSMC proliferation and migration were examined by CCK8 assay and transwell assay, respectively. The levels of pro-inflammatory cytokines were assessed using ELISA. The interactions between METTL14/YTHDF1, UCHL5 and NLRP3 were analyzed using RIP and/or dual-luciferase reporter gene and/or Co-IP assays. NLRP3 ubiquitination was analyzed by ubiquitination analysis. RESULTS: UCHL5 was significantly upregulated in AS patients and ox-LDL-treated VSMCs. UCHL5 silencing ameliorated plaque formation and vascular remodeling in vivo and suppressed ox-LDL-induced VSMC proliferation, migration, inflammation and phenotypic switching in vitro. Moreover, METTL14 could increase UCHL5 mRNA m6A level and promoted UCHL5 expression by recruiting YTHDF1. Moreover, UCHL5 overexpression enhanced protein stability by deubiquitinating NLRP3. Rescue studies revealed that NLRP3 overexpression abrogated UCHL5 silencing-mediated biological effects in ox-LDL-treated VSMCs. CONCLUSION: UCHL5 modified by METTL14/YTHDF1 axis could facilitate the inflammation and vascular remodeling in atherosclerosis by activating the NLRP3 inflammasome.


Asunto(s)
Aterosclerosis , MicroARNs , Animales , Ratones , Aterosclerosis/genética , Aterosclerosis/metabolismo , Movimiento Celular , Proliferación Celular , Inflamasomas/metabolismo , Lipoproteínas LDL/metabolismo , MicroARNs/genética , Miocitos del Músculo Liso/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Remodelación Vascular
5.
Biotechnol Genet Eng Rev ; : 1-16, 2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36946412

RESUMEN

Atherosclerosis (AS) is a chronic inflammatory disease which gives rise to life-threatening complications like ischemic stroke. Rupture of carotid atherosclerotic plaque is the main cause of ischemic stroke. Emerging evidence has demonstrated that disturbed circadian rhythms could accelerate the progression of atherosclerosis by regulating endothelial function. Moreover, our previous study implicated the circadian gene circadian locomotor output cycles kaput (CLOCK) in the pathogenesis of unstable plaques. In this study, we explored the underlying mechanism that CLOCK mediates endothelial cell autophagy involved in the progression of AS. Circadian and autophagy gene expression was analyzed in the GSE41571 dataset and human carotid atherosclerotic plaque samples. Then we used ox-LDL to treat HUVECs, and analyzed CLOCK and autophagy gene in endothelial cells. Besides that, we comprehensively analyzed in vivo experiments to explore the function of CLOCK in autophagy and atherosclerosis using different staining including HE, MT and IF staining. In the dataset and patient samples, CLOCK expression and autophagy were decreased in the unstable plaque group compared with the stable group. Decreased Beclin1, ATG5, LC3, and CLOCK were also observed in HUVECs under oxidative stress condition which also enhances cell proliferation. In vivo, we also found decreasing level of CLOCK, Beclin1, LC3 and ATG5 in ApoE-/- mice compared with WT mice. Silencing of CLOCK in ApoE-/- mice may further aggravate atherosclerosis including decreased cap thickness and collagens. Our findings implicated that downregulation CLOCK would impair endothelial cell autophagy and accelerate atherosclerotic plaque, which provides a novel strategy for treatment of progression in AS.

6.
Int J Low Extrem Wounds ; : 15347346221131844, 2022 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-36221954

RESUMEN

Aims: Diabetic foot results in frequent amputation and quality-of-life reduction in diabetes population. These lesions are featured by a prolonged and exaggerated inflammation with a significant impairment in local bacterial invasion. Negative pressure wound therapy (NPWT) attenuates hyperinflammation in the healing of diabetic foot wounds, but the potential mechanism of NPWT down-regulated inflammatory reaction still remains elusive. This study aims to explore the inflammatory signaling involved in the effect of NPWT on diabetic ulcer. Methods: Thirty patients with diabetic foot ulceration were divided into NPWT group (treated with NPWT, n = 10), NPWT + FK565 group (treated with NPWT combined with FK565 which is NOD1 receptor ligand, n = 10) and control group (n = 10). After two weeks treatment, samples were harvested and analyzed by histochemistry for infiltration of inflammatory cells, immunofluorescence stain for NOD1, western blotting for NOD1, RIP2 (Receptor interacting protein 2), IL-1ß, TAK1 (Transforming growth factor-ß-activated kinase1), p65 and real time-PCR for expression of NOD1 and RIP2. Results: NPWT could notably accelerate the diabetic wound healing through alleviating inflammatory reaction. The immunofluorescence analysis results revealed that NOD1 was mainly expressed in the cytoplasm and noticeably decreased after the NPWT treatment. And NPWT obviously decreased both the mRNA and protein level of NOD1 and RIP2. Moreover, The protein expression of IL-1ß, TAK1 and p65 in the NPWT-group were significant decreased. Conclusion: NPWT effectively promotes wound healing by suppressing the wound inflammation in diabetic foot, which is mediated at least in part by suppression of NOD1 receptor.

7.
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
8.
BMC Cardiovasc Disord ; 22(1): 317, 2022 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-35842583

RESUMEN

BACKGROUND: The efficacy and validity of excimer laser ablation (ELA) in the in-stent restenosis (ISR) has been confirmed. However, its application in de novo atherosclerotic lesions of lower extremity artery disease (LEAD) has not been clearly defined and its procedure has not been standardized. METHODS: ELABORATE is a prospective, multicenter, real-world study designed to evaluate the efficacy and safety between ELA combined with drug-coated balloon (DCB) and DCB alone in de novo atherosclerotic lesions of LEAD. DISCUSSION: ELABORATE is a prospective, multicenter, real-world study designed to assess the efficacy and safety between ELA combined with drug-coated balloon (DCB) and DCB alone in patients with de novo atherosclerotic lesions of LEAD. According to the real-world situation, eligible patients will be allocated to ELA + DCB group (group E) and DCB group (group C). Baseline and follow-up information (at 3, 6, and 12 months) will be collected. The primary efficacy point is primary patency at 12-months, and the secondary efficacy points include clinically driven target lesion reintervention (CD-TLR), change of Rutherford class, ankle-brachial index and ulcer healing rate. These indexes will be assessed and recorded at 3, 6, and 12-month follow-up. Also, safety evaluation, including major adverse event, all-cause mortality through 30-day follow-up, unplanned major amputation, bailout stent and distal embolization, will also be evaluated by an independent core laboratory. All the data will be collected and recorded by the electric data capture system. This study will be finished in 3 years and the 12-month results will be available in 2023. All the patients will be followed for 5 years. Trial registration number Chinese Clinical Trial Registry (ChiCTR2100051263). Registered 17 September 2019. http://www.chictr.org.cn/listbycreater.aspx .


Asunto(s)
Angioplastia de Balón , Terapia por Láser , Enfermedad Arterial Periférica , Angioplastia de Balón/efectos adversos , Terapia Combinada/efectos adversos , Constricción Patológica/etiología , Constricción Patológica/cirugía , Humanos , Extremidad Inferior , Estudios Multicéntricos como Asunto , Enfermedad Arterial Periférica/terapia , Estudios Prospectivos , Resultado del Tratamiento , Grado de Desobstrucción Vascular
9.
Vasc Med ; 27(4): 358-364, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35361027

RESUMEN

INTRODUCTION: Venous ulceration is a multifactorial disease, and whether hyperhomocysteinemia (HHcy) promotes deterioration from primary varicose veins to venous ulcers remains unproven. METHODS: This study retrospectively analyzed clinical data from 717 patients to investigate the potential correlation between HHcy and primary varicose veins ulcer formation, including 611 patients without ulcers (control group) and 106 with ulcers (case group). RESULTS: In this study, 46.2% (49/106) of patients in the case group and 17.5% (107/611, p < 0.001) in the control group suffered from HHcy. Multivariate logistic analysis revealed that HHcy was closely associated with the incidence of venous ulceration in patients with primary varicose veins (p < 0.001). Propensity score matching created 101 matched pairs of patients with and without ulcers, and the analysis pointed to a potential link between HHcy and ulcer formation in the context of primary varicose veins (p < 0.001). Additional experiments showed that HHcy could induce endothelial dysfunction and phenotypic switching of vascular smooth muscle cells. CONCLUSION: Both clinical and experimental findings implicated HHcy as a key factor in the development of venous ulceration. Further research is needed to appraise the effectiveness of HHcy-lowering therapy in the prevention of venous ulcers in patients with varicose veins.


Asunto(s)
Úlcera Varicosa , Várices , Homocisteína , Humanos , Estudios Retrospectivos , Úlcera/complicaciones , Úlcera Varicosa/diagnóstico , Úlcera Varicosa/epidemiología , Várices/complicaciones
10.
Surgery ; 171(5): 1427-1433, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34823897

RESUMEN

BACKGROUND: This study aimed to investigate the clinical results of endovenous thermal ablation combined with stab phlebectomy for unilateral varicose veins based on diabetic management. METHODS: The study reviewed 501 patients who underwent endovenous thermal ablation combined with stab phlebectomy for unilateral varicose veins, including 337 nondiabetics (control group) and 164 diabetics. Diabetics with hemoglobin A1c ≥7% were classified as the poor glycemic control group, and hemoglobin A1c <7% as the good glycemic control group. Surgical outcomes were assessed by Venous Clinical Severity Score. The Chronic Venous disease quality of life Questionnaire was used to assess the quality of life. RESULTS: Lower limb varicose veins can be treated successfully with endovenous thermal ablation combined with stab phlebectomy in patients with or without poor glycemic control, accompanied by a significant improvement in health status. For patients with initial varicose veins (preoperative Venous Clinical Severity Score <10), the results revealed satisfactory improvements in Venous Clinical Severity Score and quality of life among the control, poor glycemic control, and good glycemic control groups. Patients with advanced varicose veins (preoperative Venous Clinical Severity Score ≥10) also showed an obvious amelioration concerning venous symptoms and quality of life. However, the extent of improvement varied among the 3 groups. Patients subjected to advanced varicose veins with the condition of poor glycemic control exhibited a less desirable improvement in postoperative health conditions compared with the control and good glycemic control groups, especially in edema relief and ulcer healing. CONCLUSION: Endovenous thermal ablation combined with stab phlebectomy is safe and effective in the treatment of varicose veins with or without poor glycemic control. Clinical attempts at hemoglobin A1c management may contribute to improved clinical outcomes in patients with advanced varicose veins.


Asunto(s)
Várices , Insuficiencia Venosa , Hemoglobina Glucada , Control Glucémico , Humanos , Extremidad Inferior , Calidad de Vida , Vena Safena/cirugía , Resultado del Tratamiento , Várices/cirugía , Insuficiencia Venosa/cirugía
11.
J Mol Histol ; 52(4): 799-808, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34131827

RESUMEN

Defective autophagy in vascular smooth muscle cells (VSMCs) in response to oxidative stress can lead to cellular apoptosis and plaque instability. Previous studies have revealed that the circadian clock system is involved in autophagic regulation and plaque progression. However, the mechanism by which circadian rhythmicity influences VSMC autophagy and plaque stability remains unclear. Our study described the circadian profiles in atheromatous plaques and verified the role of circadian misalignment in VSMC autophagy and apoptosis. We found that the mRNA expression levels of circadian locomotor output cycles protein kaput (CLOCK) and Beclin 1 were significantly decreased in unstable plaques compared with stable plaques. No significant differences were observed in other circadian rhythm genes. VSMCs treated with oxidized low-density lipoprotein (ox-LDL, 80 µg/ml) exhibited abnormal circadian rhythmicity and impaired autophagy, as evidenced by consistent decreases in CLOCK and Beclin 1 expression, suggesting a correlation between CLOCK and autophagy. CLOCK protein expression was inhibited by ox-LDL, accompanied by defective autophagy and an increased apoptosis rates (P < 0.05). Administration of rapamycin (10 nM) reversed the effect of ox-LDL on VSMC autophagy and apoptosis. Finally, CLOCK silencing led to a considerable decrease in autophagy. VSMCs with stable CLOCK silencing also showed an increased apoptosis rate. In addition, gene silencing of CLOCK in VSMCs counteracted the effects of moderate rapamycin concentrations on autophagy and apoptosis. In conclusion, these findings suggested that the CLOCK-dependent rapamycin signaling pathway is a critical mediator in ox-LDL-induced VSMCs with defective autophagy that exacerbates plaque destabilization.


Asunto(s)
Apoptosis , Autofagia , Beclina-1/genética , Proteínas CLOCK/genética , Trastornos Cronobiológicos/complicaciones , Regulación de la Expresión Génica/fisiología , Músculo Liso Vascular/patología , Aorta/citología , Western Blotting , Cadaverina/análogos & derivados , Cadaverina/metabolismo , Células Cultivadas , Humanos , Lipoproteínas LDL/farmacología , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
12.
J Vasc Surg Venous Lymphat Disord ; 8(5): 905-906, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32800262
13.
Front Neurosci ; 14: 651, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655362

RESUMEN

BACKGROUND: Selective loss of dopaminergic neurons and diminished putamen gray matter volume (GMV) represents a central feature of Parkinson's disease (PD). Recent studies have reported specific effects of kinectin 1 gene (KTN1) variants on the putamen GMV. OBJECTIVE: To examine the relationship of KTN1 variants, KTN1 mRNA expression in the putamen and substantia nigra pars compacta (SNc), putamen GMV, and PD. METHODS: We examined the associations between PD and a total of 1847 imputed KTN1 single nucleotide polymorphisms (SNPs) in one discovery sample [2,000 subjects with PD vs. 1,986 healthy controls (HC)], and confirmed the nominally significant associations (p < 0.05) in two replication samples (900 PD vs. 867 HC, and 940 PD vs. 801 HC, respectively). The regulatory effects of risk variants on the KTN1 mRNA expression in putamen and SNc and the putamen GMV were tested. We also quantified the expression levels of KTN1 mRNA in the putamen and/or SNc for comparison between PD and HC in five independent cohorts. RESULTS: Six replicable and two non-replicable KTN1-PD associations were identified (0.009 ≤ p ≤ 0.049). The major alleles of five SNPs, including rs12880292, rs8017172, rs17253792, rs945270, and rs4144657, significantly increased risk for PD (0.020 ≤ p ≤ 0.049) and putamen GMVs (19.08 ≤ ß ≤ 60.38; 2.82 ≤ Z ≤ 15.03; 5.0 × 10-51 ≤ p ≤ 0.018). The risk alleles of five SNPs, including rs8017172, rs17253792, rs945270, rs4144657, and rs1188184 also significantly increased the KTN1 mRNA expression in the putamen or SNc (0.021 ≤ p ≤ 0.046). The KTN1 mRNA was abundant in the putamen and/or SNc across five independent cohorts and differentially expressed in the SNc between PD and HC in one cohort (p = 0.047). CONCLUSION: There was a consistent, significant, replicable, and robust positive relationship among the KTN1 variants, PD risk, KTN1 mRNA expression in putamen, and putamen volumes, and a modest relation between PD risk and KTN1 mRNA expression in SNc, suggesting that KTN1 may play a functional role in the development of PD.

14.
J Vasc Surg Venous Lymphat Disord ; 8(1): 74-83, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31699666

RESUMEN

BACKGROUND: Lower limb chronic venous disease (CVD), resulting from iliac vein compression syndrome (IVCS), manifests as a series of symptoms ranging from varicose veins to venous ulcerations. Stent implantation has been considered an effective treatment method; however, the management of CVD has rarely been reported. In the present study, we evaluated the treatment and outcomes of patients with CVD. METHODS: We performed a retrospective cohort study of patients with severe iliac vein stenosis with lower limb CVD. The patients were divided into two groups: group 1 had received stenting alone (n = 42), and group 2 had received stenting and high ligation/endovenous laser treatment (n = 29). We evaluated the clinical outcomes using the Venous Clinical Severity Score and visual analog scale, and assessed the quality of life (QoL) using the Chronic Venous Disease QoL questionnaire at a median follow-up point of 15 months (range, 6-25 months). RESULTS: In our cohort, the prevalence rate of nonthrombotic IVCS (NIVCS) was 11.7% (98 of 838 patients). The technical success rate was 100%, without severe complications. During the study period, three group 1 patients and two group 2 patients were lost to follow-up. The overall patency rate in the patients with NIVCS during a mean follow-up period of 15.0 months (range, 6-25 months) was 94.4%. For patients with a Clinical, Etiology, Anatomy, Pathophysiology (CEAP) clinical class of <4, all parameters showed similar improvements in the two groups, except for the disappearance of varicose veins. However, in patients with a CEAP clinical class of ≥4, the combination therapy significantly improved their QoL. The Venous Clinical Severity Score reduction was 4.64 ± 1.72 in group 1 and 11.89 ± 1.82 in group 2 (P < .01). Pain, scored using the visual analog scale, demonstrated a decrease from 4.41 to 2.52 (P < .05) in group 1 and 4.71 to 0.53 (P < .01) in group 2. The relief rate of stasis dermatitis in groups 1 and 2 was 26.9% and 90.5%, respectively (P < .05), and the venous ulceration healing rate was 16.7% and 87.5%, respectively (P < .05). CONCLUSIONS: The prevalence of NIVCS should not be overlooked. The proposed combination treatment is an effective therapeutic strategy for patients with NIVCS and advanced CVD (CEAP clinical class, ≥4) during short-term follow-up.


Asunto(s)
Procedimientos Endovasculares/instrumentación , Vena Ilíaca , Terapia por Láser , Síndrome de May-Thurner/terapia , Vena Safena/cirugía , Stents , Várices/cirugía , Anciano , Enfermedad Crónica , Terapia Combinada , Procedimientos Endovasculares/efectos adversos , Femenino , Humanos , Vena Ilíaca/diagnóstico por imagen , Vena Ilíaca/fisiopatología , Terapia por Láser/efectos adversos , Ligadura , Masculino , Síndrome de May-Thurner/diagnóstico por imagen , Síndrome de May-Thurner/fisiopatología , Persona de Mediana Edad , Calidad de Vida , Estudios Retrospectivos , Vena Safena/diagnóstico por imagen , Vena Safena/fisiopatología , Factores de Tiempo , Resultado del Tratamiento , Várices/diagnóstico por imagen , Várices/fisiopatología , Cicatrización de Heridas
15.
Diabetes Res Clin Pract ; 150: 81-89, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30825563

RESUMEN

AIMS: Negative pressure wound therapy displayed significant clinical benefits in the healing of diabetic foot wounds. In the present study, we investigated the mechanism of regulation of MAPK-JNK (Mitogen-activated protein kinase- c-Jun N-terminal kinase) signaling pathway by negative pressure wound therapy on these wounds. METHODS: Twenty-six type 2 diabetes patients with foot ulceration were randomly assigned to the two groups, thirteen treated with negative pressure wound therapy and the others treated with traditional debridement therapy. Skin samples were harvested and histologically and immunohistochemical analyzed in both groups. Immunofluorescence stain, Enzyme-linked immunosorbent assay and Western blotting were performed for inducible nitric oxide synthase, inter leukin-6, tumor necrosis factor-α, P-c-Jun N-terminal kinase and c-Jun N-terminal kinase. Real time-polymerase chain reaction was performed to evaluate expression of c-Jun N-terminal kinase, extracellular signal regulated kinase1/2 and p38. RESULTS: Negative pressure wound therapy could effectively alleviate inflammatory reaction and reduce inter leukin-6 and inducible nitric oxide synthase production after 7 days treatment. The level of tumor necrosis factor-α, inter leukin-6 and P-c-Jun N-terminal kinase were significantly decreased. However, there was no statistical difference in messenger ribonucleic acid expression of p38, extracellular signal regulated kinase1 and 2. CONCLUSIONS: Negative pressure wound therapy possibly suppress the wound inflammation by inhibiting inter leukin-6, tumor necrosis factor-α and inducible nitric oxide synthase in diabetic foot patients. This effect is maybe mediated at least in part by suppression of Mitogen-activated protein kinase- c-Jun N-terminal kinase signaling pathway.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Pie Diabético/terapia , Inflamación/prevención & control , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Terapia de Presión Negativa para Heridas/métodos , Cicatrización de Heridas , Pie Diabético/etiología , Pie Diabético/patología , Regulación hacia Abajo , Femenino , Humanos , Inflamación/etiología , Inflamación/patología , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Masculino , Persona de Mediana Edad , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transducción de Señal
16.
Jacobs J Genet ; 4(1)2019.
Artículo en Inglés | MEDLINE | ID: mdl-32149191

RESUMEN

OBJECTIVE: Piwi-interacting RNAs (piRNAs) represent a molecular feature shared by all nonaging biological systems, including the germline and somatic cancer stem cells, which display an indefinite renewal capacity and lifespan-stable genomic integrity and are potentially immortal. Here, we tested the hypothesis that piRNA is a critical genetic determinant of aging in humans. METHODS: Expression of transcriptome-wide piRNAs (n=24k) was profiled in the human prefrontal cortex of 12 subjects (84.9±9.5, range 68-100, years of age) using microarray technology. We examined the correlation between these piRNAs' expression levels and age, adjusting for covariates including disease status. RESULTS: A total of 9,453 piRNAs were detected in brain. Including seven intergenic and three intronic piRNAs, ten piRNAs were significantly associated with age after correction for multiple testing (|r|=0.9; 1.9×10-5≤p≤9.9×10-5). CONCLUSION: We conclude that piRNAs might play a potential role in determining the years of survival of humans. The underlying mechanisms might involve the suppression of transposable elements (TEs) and expression regulation of aging-associated genes.

17.
Oxid Med Cell Longev ; 2018: 4169575, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29849894

RESUMEN

Vascular endothelial dysfunction is considered critical development in the progression of cardiovascular events and is associated with vascular damage and oxidative stress. Previous studies have shown that profilin-1 could be induced by advanced glycation end products (AGEs) and contributes to vascular hyperpermeability; however, the mechanisms are not fully understood. In this study, we sought to assess whether reactive oxygen species (ROS) were involved in profilin-1-mediated RhoA/ROCK1 signaling. Treatment with AGEs significantly induced the expression of profilin-1 and ROS production in human umbilical vein endothelial cells (HUVECs), whereas knockdown of profilin-1 diminished AGE-induced RhoA and ROCK1 activation and ROS production. Moreover, ectopic overexpression of profilin-1 also increased RhoA and ROCK1 activation and ROS production under low AGE concentration. Furthermore, blockage of RhoA/ROCK1 with the inhibitors CT04 and Y27632 significantly attenuated the profilin-1-mediated cell damage and ROS production. Additionally, ROS inhibition resulted in a significant decrease in profilin-1-mediated RhoA/ROCK1 expression, suggesting that the regulation of RhoA/ROCK1 signaling was partly independent of ROS. Taken together, these results suggested that the RhoA/ROCK1 pathway activated by excessive ROS is responsible for profilin-1-mediated endothelial damage.


Asunto(s)
Células Endoteliales/metabolismo , Estrés Oxidativo/fisiología , Profilinas/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , ADN Complementario/administración & dosificación , ADN Complementario/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Retroalimentación , Productos Finales de Glicación Avanzada/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Estrés Oxidativo/efectos de los fármacos , Profilinas/genética , Especies Reactivas de Oxígeno/metabolismo , Transfección , Regulación hacia Arriba/efectos de los fármacos
18.
Mol Med Rep ; 15(3): 1401-1406, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28098888

RESUMEN

The expression and secretion of infla-mmation­associated cytokines are induced by hypoxia. Circadian locomotor output cycles protein kaput (CLOCK) has previously been shown to activate the nuclear factor­κB (NF­κB) pathway, which is a key transcription factor during hypoxia. The present study evaluated the role of the NF­κB pathway in the CLOCK­induced inflammatory response. Under hypoxic conditions, the expression levels of NF­κB and proinflammatory cytokines, including interleukin (IL)­1, IL­1ß, IL­6, intercellular adhesion molecule 1, cyclooxygenase 2 and tumor necrosis factor alpha, were significantly increased compared with under control conditions. Conversely, human umbilical vein endothelial cells (HUVECs) that were transfected with small hairpin RNA against human CLOCK exhibited reversed effects. Furthermore, inhibition of NF­κB with pyrrolidine dithiocarbamate (PDTC) reduced the expression of proinflammatory cytokines in HUVECs treated under hypoxic conditions. In addition, the CLOCK­induced inflammatory response was abolished with PDTC treatment. These findings suggest that the mechanism by which CLOCK induces inflammation mainly involves activation of the NF­κB signaling pathway.


Asunto(s)
Proteínas CLOCK/metabolismo , Hipoxia/metabolismo , Inflamación/metabolismo , Transducción de Señal , Proteínas CLOCK/genética , Citocinas/metabolismo , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipoxia/genética , Inflamación/genética , Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , Interferencia de ARN
19.
Pak J Med Sci ; 32(1): 55-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27022345

RESUMEN

OBJECTIVE: To evaluate the clinical effect of endovenous laser treatment (EVLT) for patients with varicose veins. METHODS: Our series included 117 patients who underwent EVLT combined with high ligation and stripping since the introduction of the technique in our institution. All EVLT procedures were performed with local skin cooling to prevent skin burns, as well as stripping after exsanguinations to prevent thrombotic phlebitis. RESULTS: A total of 146 limbs in 117 patients were ablated by EVLT. Bilateral EVLT was performed in 29 patients, with the remaining 88 patients undergoing unilateral EVLT. The mean age of the patients was 57 years ± 12 years (range: 21 years to 80 years), and 56 were male and 61 were female. Follow-up for all patients lasted three to six months. The most common complication was induration and swelling, which was observed in 64 patients, followed by paraesthesia in 27, and skin burns in 12. CONCLUSION: The treatment with endovenous laser treatment for patients with varicose veins is safe and effective.

20.
Oxid Med Cell Longev ; 2016: 9591482, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28058089

RESUMEN

A number of recent studies have implicated that autophagy was activated by reactive oxygen species (ROS). Our previous report indicated that CLOCK increased the accumulation of ROS under hypoxic conditions. In this study, we investigated the mechanisms by which CLOCK mediated endothelial damage, focusing on the involvement of oxidative damage and autophagy. Overexpression of CLOCK in human umbilical vein endothelial cells (HUVECs) showed inhibition of cell proliferation and higher autophagosome with an increased expression of Beclin1 and LC3-I/II under hypoxic conditions. In contrast, CLOCK silencing reversed these effects. Interestingly, pretreatment with 3-methyladenine (3-MA) resulted in the attenuation of CLOCK-induced cell autophagy and but did not influence the production of intracellular reactive oxygen species (ROS). Furthermore, Tiron (4,5-dihydroxy-1,3-benzene disulfonic acid-disodium salt), a ROS scavenger, significantly attenuated CLOCK-induced cell autophagy. In addition, we found that overexpression of CLOCK had no significant effects on the production of ROS and expression of Beclin1 and LC3-I/II under normoxic conditions in HUVEC. In this present investigation, our results suggested a novel mechanism of action of CLOCK in HUVECs, opening up the possibility of targeting CLOCK for the treatment of vascular diseases.


Asunto(s)
Autofagia , Proteínas CLOCK/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Especies Reactivas de Oxígeno/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Autofagia/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA