Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Acta Orthop Belg ; 83(3): 396-404, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30423640

RESUMEN

The objective of this retrospective study was to compare open reduction and internal fixation (ORIF) with primary partial arthrodesis for the treatment of Lisfranc injuries accompanied by comminution of the second metatarsal base. Thirty-four patients were treated with ORIF or primary partial arthrodesis from 2007 to 2013. The patients were followed for an average of 28.5 months. Evaluation was performed with clinical examination, radiography, Visual Analogue Scale (VAS), the American Orthopedic Foot and Ankle Society (AOFAS) Midfoot Score, and the Short Form 36 (SF-36). Fifteen patients were treated with ORIF, and nineteen patients were treated with primary partial arthrodesis. Anatomical reduction was obtained in all patients. At two years postoperatively, the mean AOFAS Midfoot score was 84.33 points in the ORIF group and 85.05 points in the primary partial arthrodesis group (P> 0.05). Also, no significant differences were seen in the VAS for pain (1.20 vs 1.05 points), SF-36 physical component (79.60 vs 79.89 points) or SF-36 mental component (77.07 vs 79.21 points). With longer and conservative postoperative management, ORIF as well as primary partial arthrodesis for Lisfranc injuries accompanied by comminution of the second metatarsal base led to similar medium-term outcome.


Asunto(s)
Artrodesis , Fijación Interna de Fracturas , Fracturas Conminutas/cirugía , Huesos Metatarsianos/cirugía , Reducción Abierta , Articulaciones Tarsianas/cirugía , Adulto , Artrodesis/efectos adversos , Femenino , Estudios de Seguimiento , Fijación Interna de Fracturas/efectos adversos , Fracturas Conminutas/diagnóstico por imagen , Humanos , Masculino , Huesos Metatarsianos/efectos de los fármacos , Huesos Metatarsianos/lesiones , Persona de Mediana Edad , Reducción Abierta/efectos adversos , Dolor Postoperatorio/etiología , Estudios Retrospectivos , Articulaciones Tarsianas/diagnóstico por imagen , Resultado del Tratamiento , Adulto Joven
2.
Chin J Integr Med ; 2023 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-37930511

RESUMEN

OBJECTIVE: To investigate the therapeutic effect of Sanhuang Xiexin Decoction (SXD) on triple-negative breast cancer (TNBC) in mice and its underlying mechanism. METHODS: The high-performance liquid chromatography (HPLC) was used to quantitate and qualify SXD. A total of 15 female BALB/c mice were inoculated subcutaneously on the right hypogastrium with 3×105 of 4T1-Luc cells to establish TNBC mouse model. All mice were divided randomly into 3 groups, including phosphate buffered solution (PBS), SXD and doxorubicin (DOX) groups (positive drug). Additionally, tumor growth, pathological changes, serum lipid profiles, expression of Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signaling pathway and its key targets including inflammatory factors, cell cycle and epithelial-mesenchymal transition (EMT) markers were investigated. Besides, the biosafety of SXD was also evaluated in mice. RESULTS: Rhein, coptisine, berberine hydrochloride and baicalin were all found in SXD, and the concentrations of these 4 components were 0.57, 2.61, 2.93, and 46.04 mg/g, respectively. The mouse experiment showed that SXD could notably suppress the development of tumors and reduce the density of tumor cells (P<0.01). The serum lipid analysis and Oil-Red-O staining both showed the differences, SXD group exhibited higher serum adiponectin and HDL-C levels with lower TC and LDL-C levels compared to the PBS and DOX groups (P<0.05 or P<0.01), respectively. SXD also decreased the levels of phospho-JAK2 (p-JAK2), phospho-STAT3 (p-STAT3) expressions and its downstream factors, including mostly inflammatory cytokine, EMT markers, S phase of tumor cells and vascular endothelial growth factor (VEGF) expression (P<0.05 or P<0.01), respectively. The biosafety assessment of SXD revealed low levels of toxicity in mice. CONCLUSION: SXD could inhibit TNBC by suppressing JAK2-STAT3 phosphorylation which may be associated with modulation of lipid metabolism.

3.
Chin J Integr Med ; 24(3): 200-206, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28432529

RESUMEN

OBJECTIVE: To investigate the potential efficacy of panaxadiol saponins component (PDS-C), a biologically active fraction isolated from total ginsenosides, to reverse chemotherapy-induced myelosuppression and pancytopenia caused by cyclophamide (CTX). METHODS: Mice with myelosuppression induced by CTX were treated with PDS-C at a low- (20 mg/kg), moderate- (40 mg/kg), or high-dose (80 mg/kg) for 7 consecutive days. The level of peripheral white blood cell (WBC), neutrophil (NEU) and platelet (PLT) were measured, the histopathology and colony formation were observed, the protein kinase and transcription factors in hematopoietic cells were determined by immunohistochemical staining and Western blot. RESULTS: In response to PDS-C therapy, the peripheral WBC, NEU and PLT counts of CTX-induced myelosuppressed mice were significantly increased in a dose-dependent manner. Similarly, bone marrow histopathology examination showed reversal of CTX-induced myelosuppression with increase in overall bone marrow cellularity and the number of hematopoietic cells (P<0.01). PDS-C also promoted proliferation of granulocytic and megakaryocyte progenitor cells in CTX-treated mice, as evidenced by significantly increase in colony formation units-granulocytes/monocytes and -megakaryocytes (P<0.01). The enhancement of hematopoiesis by PDS-C appears to be mediated by an intracellular signaling pathway, this was evidenced by the up-regulation of phosphorylated mitogen-activated protein kinase (p-MEK) and extracellular signal-regulated kinases (p-ERK), and receptor tyrosine kinase (C-kit) and globin transcription factor 1 (GATA-1) in hematopoietic cells of CTX-treated mice (P<0.05). CONCLUSIONS: PDS-C possesses hematopoietic growth factor-like activities that promote proliferation and also possibly differentiation of hematopoietic progenitor cells in myelosuppressed mice, probably mediated by a mechanism involving MEK and ERK protein kinases, and C-kit and GATA-1 transcription factors. PDS-C may potentially be a novel treatment of myelosuppression and pancytopenia caused by chemotherapy.


Asunto(s)
Antineoplásicos/efectos adversos , Ciclofosfamida/efectos adversos , Ginsenósidos/uso terapéutico , Hematopoyesis/efectos de los fármacos , Células Mieloides/patología , Panax/química , Pancitopenia/tratamiento farmacológico , Saponinas/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor de Transcripción GATA1/metabolismo , Ginsenósidos/farmacología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Células Mieloides/efectos de los fármacos , Pancitopenia/inducido químicamente , Pancitopenia/patología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regulación hacia Arriba/efectos de los fármacos
4.
Chin J Integr Med ; 23(4): 288-294, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28364352

RESUMEN

OBJECTIVE: To explore the effects of bufalin on inhibiting proliferation, up-regulating methylation of Wilm' tumor 1 gene (WT1) as well as its possible mechanisms in human erythroid leukemic (HEL) cells. METHODS: The HEL cells were treated with bufalin at various concentrations to observe cellular morphology, proliferation assay and cell cycle. The mRNA and protein expression levels of WT1 were detected by reverse transcription polymerase chain reaction (RT-PCR), Western blot and immunocytochemistry, DNA methylation of WT1 and protein expression levels of DNA methyltransferase 3a (DNMT3a) and DNMT3b were analyzed by methylation-specific PCR, and Western blot respectively. RESULTS: The bufalin was effective to inhibit proliferation of HEL cells in a dose-dependent manner, their suppression rates were from 23.4%±2.1% to 87.2%±5.4% with an half maximal inhibit concentration (IC50) of 0.046 µmol/L. Typical apoptosis morphology was observed in bufalin-treated HEL cells. The proliferation index of cell cycle decreased from 76.4%±1.9% to 49.7%±1.3%. The expression levels of WT1 mRNA and its protein reduced gradually with increasing doses of bufalin, meanwhile, the methylation status of WT1 gene changed from unmethylated into partially or totally methylated. While, the expression levels of DNMT3a and DNMT3b protein gradually increased by bufalin treatment in a dose-dependent manner. CONCLUSIONS: Bufalin can not only significantly inhibit the proliferation of HEL cells and arrest cell cycle at G0/G1 phase, but also induce cellular apoptosis and down-regulate the expression level of WT1. Our results provide the evidence of bufalin for anti-leukemia, its mechanism may involve in increasing WT1 methylation status which is related to the up-regulation of DNMT3a and DNMT3b proteins in erythroid leukemic HEL cells.


Asunto(s)
Bufanólidos/farmacología , Metilación de ADN/efectos de los fármacos , Leucemia Eritroblástica Aguda/genética , Regulación hacia Arriba/efectos de los fármacos , Proteínas WT1/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , ADN Metiltransferasa 3A , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Eritroblástica Aguda/enzimología , Leucemia Eritroblástica Aguda/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba/genética , Proteínas WT1/metabolismo , ADN Metiltransferasa 3B
5.
Chin J Integr Med ; 22(12): 910-917, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26712211

RESUMEN

OBJECTIVE: To explore the mechanism of the protective effects of Panax notoginseng saponins (PNS) on kidney in diabetic rats. METHODS: Diabetic rat model was obtained by intravenous injection of alloxan, and the rats were divided into model, PNS-100 mg/(kg day) and PNS-200 mg/(kg day) groups, 10 each. Another 10 rats injected with saline were served as control. Periodic acid-Schiff staining and immunological histological chemistry were used to observe histomorphology and tissue expression of bone morphogenetic protein-7 (BMP-7). Silent information regulator 1 (SIRT1) was silenced in rat mesangial cells by RNA interference. The mRNA expressions of SIRT-1, monocyte chemoattractant protein-1 (MCP-1), transforming growth factor ß1 (TGF-ß1) and plasminogen activator inhibitor-1 (PAI-1) were analyzed by reverse transcription polymerase chain reaction. The protein expressions of SIRT1 and the acetylation of nuclear factor κB (NF-κB) P65 were determined by western blotting. The concentration of MCP-1, TGF-ß1 and malondialdehyde (MDA) in culture supernatant were detected by enzyme-linked immuno sorbent assay. The activity of superoxide dismutase (SOD) was detected by the classical method of nitrogen and blue four. RESULTS: In diabetic model rats, PNS could not only reduce blood glucose and lipid (P<0.01), but also increase protein level of BMP-7 and inhibit PAI-1 expression for suppressing fibrosis of the kidney. In rat mesangial cells, PNS could up-regulate the expression of SIRT1 (P<0.01) and in turn suppress the transcription of TGF-ß1 (P<0.05) and MCP-1 (P<0.05). PNS could also reverse the increased acetylation of NF-κB p65 by high glucose. In addition, redox regulation factor MDA was down-regulated (P<0.05) and SOD was up-regulated (P<0.01), which were both induced by SIRT1 up-regulation. CONCLUSIONS: PNS could protect kidney from diabetes with the possible mechanism of up-regulating SIRT1, therefore inhibiting inflammation through decreasing the induction of inflammatory cytokines and TGF-ß1, as well as activating antioxidant proteins.


Asunto(s)
Antioxidantes/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Riñón/patología , Panax notoginseng/química , Sustancias Protectoras/uso terapéutico , Saponinas/uso terapéutico , Sirtuina 1/genética , Regulación hacia Arriba/efectos de los fármacos , Acetilación/efectos de los fármacos , Animales , Glucemia/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Quimiocina CCL2/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatología , Técnicas de Silenciamiento del Gen , Inmunohistoquímica , Riñón/efectos de los fármacos , Pruebas de Función Renal , Lípidos/sangre , Masculino , Malondialdehído/metabolismo , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Sustancias Protectoras/farmacología , Ratas Sprague-Dawley , Saponinas/farmacología , Superóxido Dismutasa/metabolismo , Factor de Transcripción ReIA/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA