Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Phytother Res ; 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38642047

ABSTRACT

Osteoarthritis (OA) is a complicated joint disorder characterized by inflammation that causes joint destruction. Cucurbitacin B (CuB) is a naturally occurring triterpenoid compound derived from plants in the Cucurbitaceae family. The aim of this study is to investigate the potential role and mechanisms of CuB in a mouse model of OA. This study identified the key targets and potential pathways of CuB through network pharmacology analysis. In vivo and in vitro studies confirmed the potential mechanisms of CuB in OA. Through network pharmacology, 54 potential targets for CuB in treating OA were identified. The therapeutic potential of CuB is associated with the nod-like receptor pyrin domain 3 (NLRP3) inflammasome and pyroptosis. Molecular docking results indicate a strong binding affinity of CuB to nuclear factor erythroid 2-related factor 2 (Nrf2) and p65. In vitro experiments demonstrate that CuB effectively inhibits the expression of pro-inflammatory factors induced by interleukin-1ß (IL-1ß), including cyclooxygenase-2, inducible nitric oxide synthase, IL-1ß, and IL-18. CuB inhibits the degradation of type II collagen and aggrecan in the extracellular matrix (ECM), as well as the expression of matrix metalloproteinase-13 and a disintegrin and metalloproteinase with thrombospondin motifs-5. CuB protects cells by activating the Nrf2/hemeoxygenase-1 (HO-1) pathway and inhibiting nuclear factor-κB (NF-κB)/NLRP3 inflammasome-mediated pyroptosis. Moreover, in vivo experiments show that CuB can slow down cartilage degradation in an OA mouse model. CuB effectively prevents the progression of OA by inhibiting inflammation in chondrocytes and ECM degradation. This action is further mediated through the activation of the Nrf2/HO-1 pathway to inhibit NF-κB/NLRP3 inflammasome activation. Thus, CuB is a potential therapeutic agent for OA.

2.
J Endovasc Ther ; : 15266028231158294, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36891634

ABSTRACT

PURPOSE: This study aimed to develop and internally validate nomograms for predicting restenosis after endovascular treatment of lower extremity arterial diseases. MATERIALS AND METHODS: A total of 181 hospitalized patients with lower extremity arterial disease diagnosed for the first time between 2018 and 2019 were retrospectively collected. Patients were randomly divided into a primary cohort (n=127) and a validation cohort (n=54) at a ratio of 7:3. The least absolute shrinkage and selection operator (LASSO) regression was used to optimize the feature selection of the prediction model. Combined with the best characteristics of LASSO regression, the prediction model was established by multivariate Cox regression analysis. The predictive models' identification, calibration, and clinical practicability were evaluated by the C index, calibration curve, and decision curve. The prognosis of patients with different grades was compared by survival analysis. Internal validation of the model used data from the validation cohort. RESULTS: The predictive factors included in the nomogram were lesion site, use of antiplatelet drugs, application of drug coating technology, calibration, coronary heart disease, and international normalized ratio (INR). The prediction model demonstrated good calibration ability, and the C index was 0.762 (95% confidence interval: 0.691-0.823). The C index of the validation cohort was 0.864 (95% confidence interval: 0.801-0.927), which also showed good calibration ability. The decision curve shows that when the threshold probability of the prediction model is more significant than 2.5%, the patients benefit significantly from our prediction model, and the maximum net benefit rate is 30.9%. Patients were graded according to the nomogram. Survival analysis found that there was a significant difference in the postoperative primary patency rate between patients of different classifications (log-rank p<0.001) in both the primary cohort and the validation cohort. CONCLUSION: We developed a nomogram to predict the risk of target vessel restenosis after endovascular treatment by considering information on lesion site, postoperative antiplatelet drugs, calcification, coronary heart disease, drug coating technology, and INR. CLINICAL IMPACT: Clinicians can grade patients after endovascular procedure according to the scores of the nomograms and apply intervention measures of different intensities for people at different risk levels. During the follow-up process, an individualized follow-up plan can be further formulated according to the risk classification. Identifying and analyzing risk factors is essential for making appropriate clinical decisions to prevent restenosis.

3.
J Nanobiotechnology ; 20(1): 422, 2022 Sep 24.
Article in English | MEDLINE | ID: mdl-36153544

ABSTRACT

BACKGROUND: Patients with critical limb ischemia (CLI) are at great risk of major amputation and cardiovascular events. Adipose-derived mesenchymal stem cell (ADSC) therapy is a promising therapeutic strategy for CLI, but the poor engraftment and insufficient angiogenic ability of ADSCs limit their regenerative potential. Herein, we explored the potential of human umbilical vein endothelial cells (HUVECs)-derived small extracellular vesicles (sEVs) for enhancing the therapeutic efficacy of ADSCs in CLI. RESULTS: sEVs derived from hypoxic HUVECs enhanced the resistance of ADSCs to reactive oxygen species (ROS) and further improved the proangiogenic ability of ADSCs in vitro. We found that the hypoxic environment altered the composition of sEVs from HUVECs and that hypoxia increased the level of miR-486-5p in sEVs. Compared to normoxic sEVs (nsEVs), hypoxic sEVs (hsEVs) of HUVECs significantly downregulated the phosphatase and tensin homolog (PTEN) via direct targeting of miR-486-5p, therefore activating the AKT/MTOR/HIF-1α pathway and influencing the survival and pro-angiogenesis ability of ADSCs. In a hindlimb ischemia model, we discovered that hsEVs-primed ADSCs exhibited superior cell engraftment, and resulted in better angiogenesis and tissue repair. CONCLUSION: hsEVs could be used as a therapeutic booster to improve the curative potential of ADSCs in a limb ischemia model. This finding offers new insight for CLI treatment.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Adipose Tissue/metabolism , Animals , Extracellular Vesicles/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia/metabolism , Ischemia/metabolism , Ischemia/therapy , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Pathologic/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Tensins/metabolism
4.
Phytomedicine ; 96: 153908, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35026516

ABSTRACT

BACKGROUND: Therapeutic angiogenesis by transplantation of autologous/allogeneic adipose stem cells (ADSCs) is a potential method for the treatment of critical limb ischemia (CLI). However, the therapeutic efficiency is limited by poor viability, adhesion, migration and differentiation after cell transplantation into the target area. Astragaloside IV (AS-IV), one of the main active components of Astragalus, has been widely used in the treatment of ischemic diseases and can promote cell proliferation and angiogenesis. However, there is no report on the effect of AS-IV on ADSCs and its effect on hindlimb ischemia through cell transplantation. PURPOSE: The purpose of this study was to elucidate that AS-IV pretreatment enhances the therapeutic effect of ADSC on critical limb ischemia, and to characterize the underlying molecular mechanisms. METHODS: ADSCs were obtained and pretreated with the different concentration of AS-IV. In vitro, we analyzed the influence of AS-IV on ADSC proliferation, migration, angiogenesis and recruitment of human umbilical vein endothelial cells (HUVECs) and analyzed the relevant molecular mechanism. In vivo, we injected drug-pretreated ADSCs into a Matrigel or hindlimb ischemia model and evaluated the therapeutic effect by the laser Doppler perfusion index, immunofluorescence, and histopathology. RESULTS: In vitro experiments showed that AS-IV improved ADSC migration, angiogenesis and endothelial recruitment. The molecular mechanism may be related to the upregulation of CXC receptor 2 (CXCR2) to promote the phosphorylation of focal adhesion kinase (FAK). In vivo, Matrigel plug assay showed that ADSCs pretreated with AS-IV have stronger angiogenic potential. The laser Doppler perfusion index of the hindlimbs of mice in the ADSC/AS-IV group was significantly higher than the laser Doppler perfusion index of the hindlimbs of mice of the ADSC group and the control group, and the microvessel density was significantly increased. CONCLUSION: Our results demonstrate that AS-IV pretreatment of ADSC improves their therapeutic efficacy in ameliorating severe limb exclusion symptomology through CXCR2 induced FAK phosphorylation, which will bring new insights into the treatment of severe limb ischemia.


Subject(s)
Mesenchymal Stem Cells , Neovascularization, Physiologic , Adipose Tissue , Animals , Cell Proliferation , Chronic Limb-Threatening Ischemia , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Hindlimb , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/drug therapy , Mice , Phosphorylation , Receptors, Interleukin-8B , Saponins , Triterpenes
5.
Ann Vasc Surg ; 81: 240-248, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34748950

ABSTRACT

OBJECTIVE: To determine the risk for pulmonary embolism (PE) and explore the relationship between the site of thrombosis and PE in patients with acute lower extremity deep vein thrombosis (DVT). METHODS: A total of 1585 hospitalized patients first diagnosed with acute lower extremity DVT were investigated retrospectively. The patients were divided into two groups: the non-PE group (Group 1) and the PE group (Group 2). Then, Group 2 was divided into two subgroups: asymptomatic pulmonary embolism (asPE, Group 2a) and symptomatic pulmonary embolism (sPE, Group 2b). Kaplan-Meier curves and logistic regression analysis were used to explore the relevant risk factors for PE. RESULTS: Among 1585 patients, 458 patients suffered from PE, accounting for 28.9%. 102 (22.3%) of them had the typical clinical manifestations of PE and were defined as sPE, and the remaining 356 (77.7%) patients were classified as asPE. Patients with proximal lower extremity DVT were significantly more predominant in the PE group than in the non-PE group (92.8% vs. 86.2%, P<0.001). Moreover, in Group 2, patients with typical PE manifestations showed a higher proportion of patients with right lower extremity DVT than left lower extremity DVT (26.7% vs. 17.7%, P = 0.035), and bilateral lower extremity DVT than unilateral DVT (44.1% vs. 20.5%, P<0.001). By multivariate analysis, alcohol consumption (OR, 1.824; 95% CI, 1.194-2.787; P = 0.005), heart failure (OR, 2.345; 95% CI, 1.560-3.526; P<0.001), proximal DVT (OR, 2.096; 95% CI,1.407-3.123; P<0.001) were independent risk factors for PE. CONCLUSIONS: Patients with proximal acute lower extremity DVT were more likely to suffer from PE than those with distal DVT. Patients with right acute lower extremity DVT had a higher risk of sPE than patients with left acute lower extremity DVT. Alcohol consumption and heart failure were associated with the occurrence of PE in patients with acute lower extremity DVT.


Subject(s)
Pulmonary Embolism , Venous Thrombosis , Humans , Lower Extremity/blood supply , Pulmonary Embolism/complications , Pulmonary Embolism/diagnosis , Pulmonary Embolism/epidemiology , Retrospective Studies , Risk Factors , Treatment Outcome , Venous Thrombosis/complications , Venous Thrombosis/epidemiology
6.
Injury ; 53(2): 777-783, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34756414

ABSTRACT

BACKGROUND: The objective of this study was to compare the outcomes of vacuum sealing drainage (VSD) and conventional drainage after surgery in the treatment of closed calcaneal fracture. We hypothesize that VSD is superior to conventional drainage in reducing volume of drainage, time of wound drying, time of skin fold, time of wound healing, VAS at day 3 postoperatively, wound complications and increasing wound healing grade. METHODS: 120 patients with closed calcaneal fractures from January 2016 to December 2018 were enrolled in our study. They were divided randomly into VSD group (n = 60) and conventional (n = 60). The volume of drainage, duration of drainage, time of wound drying, time of skin fold, time of wound healing and VAS at day 3 postoperatively were recorded. Furthermore, the wound complications of the two groups were also evaluated. Besides, wound healing grade was used to assess the degree of wound healing. The functional outcome American Orthopaedic Foot and Ankle Society (AOFAS) hindfoot scores and visual analog scale (VAS) pain scores was also evaluated. RESULTS: A total of 10 patients were lost to follow-up for various reasons, VSD group remained 55 cases while conventional group remained 53 cases. Our results showed that VSD group exhibited significantly more volume of drainage (P< 0.0001), longer duration of drainage (P< 0.0001), shorter time of wound drying (P = 0.0086), shorter time of skin fold (P = 0.0158), shorter time of wound healing (P = 0.0240) and lower VAS at day 3 postoperatively (P = 0.0019) compared with conventional group. Moreover, VSD group was demonstrated to have significantly lower wound complications (P = 0.025) and higher rate grade A of wound healing (P = 0.031). However, no significant difference was noted in time of fracture union (P = 0.754), VAS (P = 0.407) and AOFAS score (P = 0.512) at final follow-up between the two groups. CONCLUSIONS: Our hypothesis was confirmed that VSD was superior in terms of some aspects than conventional drainage. Therefore, VSD is a safe and effective postoperative wound drainage method in the treatment of closed calcaneal fracture. However, more and higher evidence needs to be carried to demonstrate the results.


Subject(s)
Fractures, Bone , Fractures, Closed , Negative-Pressure Wound Therapy , Drainage , Fractures, Bone/surgery , Humans , Treatment Outcome , Vacuum
7.
Stem Cell Res Ther ; 12(1): 497, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34503551

ABSTRACT

BACKGROUND: Adipose-derived mesenchymal stem cells (ADSC)-based therapy is an outstanding treatment strategy for ischaemic disease. However, the therapeutic efficacy of this strategy is not ideal due to the poor paracrine function and low survival rate of ADSCs in target regions. Platelet extracellular vesicles (PEVs) are nanoparticles derived from activated platelets that can participate in communication between cells. Accumulating evidence indicates that PEVs can regulate the biological functions of several cell lines. In the present study, we aimed to investigate whether PEVs can modulate the proangiogenic potential of ADSCs in vitro and in vivo. METHODS: PEVs were identified using scanning electron microscope (SEM), flow cytometry (FCM) and nanoparticle tracking analysis (NTA). The CCK8 assay was performed to detect proliferation of cells. Transwell and wound healing assays were performed to verify migration capacity of cells. AnnexinV-FITC/PI apoptosis kit and live/dead assay were performed to assess ADSCs apoptosis under Cocl2-induced hypoxia condition. The underlying mechanisms by which PEVs affected ADSCs were explored using real time-PCR(RT-PCR) and Western blot. In addition, matrigel plug assays were conducted and mouse hindlimb ischaemic models were established to investigate the proangiogenic potential of PEV-treated ADSCs in vivo. RESULTS: We demonstrated that ADSC could internalize PEVs, which lead to a series of biological reactions. In vitro, dose-dependent effects of PEVs on ADSC proliferation, migration and antiapoptotic capacity were observed. Western blotting results suggested that multiple proteins such as ERK, AKT, FAK, Src and PLCγ1 kinase may contribute to these changes. Furthermore, PEVs induced upregulation of several growth factors expression in ADSCs and amplified the proliferation, migration and tube formation of HUVECs induced by ADSC conditioned medium (CM). In in vivo experiments, compared with control ADSCs, the injection of PEV-treated ADSCs resulted in more vascularization in matrigel plugs, attenuated tissue degeneration and increased blood flow and capillary density in ischaemic hindlimb tissues. CONCLUSION: Our data demonstrated that PEVs could enhance the proangiogenic potential of ADSCs in mouse hindlimb ischaemia. The major mechanisms of this effect included the promotion of ADSC proliferation, migration, anti-apoptosis ability and paracrine secretion.


Subject(s)
Blood Platelets , Extracellular Vesicles , Adipocytes , Adipose Tissue , Animals , Cell Proliferation , Mice , Stem Cells
8.
Phytother Res ; 35(6): 2902-2924, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33368709

ABSTRACT

OBJECTIVE: This systematic review and meta-analysis were performed to investigate the efficacy and safety of Tripterygium wilfordii glycosides (TG) for rheumatoid arthritis (RA) from the current literature. METHODS: An electronic search was conducted in eight databases (PubMed, EMBASE, Web of Science, Cochrane Library, Chinese National Knowledge Infrastructure, Chinese Biomedical Literature Database, Chinese VIP Database, and Wanfang Database) from inception until September 2020. Randomized controlled trials (RCTs) with risk of bias (RoB) score ≥ 4 according to the Cochrane RoB tool were included for the analyses. The primary outcome measures were duration of morning stiffness (DMS), tender joint count (TJC), swollen joint count (SJC), visual analog score (VAS), C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), and rheumatoid factor (RF). The secondary outcome measures were the total clinical effective rate and adverse events. All the analyses were used by the random effects models. The meta-analysis was performed using RevMan 5.3 and STATA 14.0. RESULTS: A total of 40 RCTs with 3092 patients met our inclusion criteria. This meta-analysis showed that TG plus DMARDs for RA could decrease the DMS (p < .001), TJC (p < .001), SJC (p < .001), VAS (p < .001), serum CRP (p < .001), ESR (p < .001), and RF (p < .001) and improve total effective rate (p < .001). In addition, TG was generally safe and well tolerated in RA patients. CONCLUSION: Despite the limitations, the present evidence supports, at least to an extent, that TG can be recommended for routine use for RA patients. More large multicenter and high-quality RCTs are required for further research.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Drugs, Chinese Herbal/therapeutic use , Glycosides/therapeutic use , Phytotherapy , Tripterygium/chemistry , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , C-Reactive Protein/metabolism , Drugs, Chinese Herbal/pharmacology , Glycosides/pharmacology , Humans , Randomized Controlled Trials as Topic , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...