Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Scand Cardiovasc J ; 55(5): 287-296, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34296634

RESUMEN

OBJECTIVE: The present study aimed to evaluate the protective role of cilengitide (CGT), an integrin αvß3 and αvß5 inhibitor, on doxorubicin (DOX)-induced myocardial fibrosis and cardiac dysfunction in a rat model. Methods. Forty male rats were randomly divided into four groups: DOX (n = 12), intraperitoneal (i.p.) injection of DOX 0.8 ∼ 1.0 mg/kg three times a week for up to 6 weeks, then saline i.p. three times a week for another 3 weeks; CGT (n = 8), CGT 10 mg/kg, i.p. three times a week for 9 weeks; DOX + CGT (n = 12), DOX and CGT co-administration as above for 6 weeks, then CGT alone for another 3 weeks; Control (n = 8), saline i.p. three times a week for 9 weeks. Echocardiography, serum procollagen I C-terminal propeptide (PICP) procollagen III N-terminal propeptide (PIIINP) and C telopeptide type I (CTX-I) were evaluated at baseline and 3, 6 and 9 weeks after initial DOX administration for all surviving rats. The heart tissues were then harvested for myocardial hydroxyproline (HYP) evaluation, qRT-PCR, and western blotting. Results. CGT attenuated DOX-induced eccentric remodeling by improving relative wall thickness at the 9th week. CGT also improved systolic function at the 9th week and diastolic function at the 6th and 9th week. CGT reduced myocardial HYP and serum PICP, PIIINP, CTX-I, and the PICP/PIIINP ratio. RT-PCR and western blot showed that CGT blocked the TGF-ß1/SMAD3 pathway and mitigating extracellular matrix turnover. Conclusions. CGT exerted a cardioprotective effect against doxorubicin-induced fibrosis and improved cardiac function.


Asunto(s)
Cardiomiopatías , Integrina alfaVbeta3 , Receptores de Vitronectina , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/epidemiología , Modelos Animales de Enfermedad , Doxorrubicina/toxicidad , Fibrosis/prevención & control , Integrina alfaVbeta3/antagonistas & inhibidores , Masculino , Ratas , Receptores de Vitronectina/antagonistas & inhibidores
2.
Biotechnol Lett ; 38(11): 1839-1849, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27447467

RESUMEN

OBJECTIVES: To determine the effect of NF-κB on cell proliferation and apoptosis, we investigate the expression of inflammation and apoptosis-related factors in the bovine mammary epithelial cell line, MAC-T. RESULTS: MAC-T cells were cultured in vitro and MTT and LDH assays used to determine the effects of lipopolysaccharide (LPS) on proliferation and cytotoxicity respectively. RT-PCR and western blotting were used to evaluate the effect of LPS and NF-κB inhibition [pyrrolidine dithiocarbamate (PDTC) treatment] on the expression of inflammation and apoptosis-related factors. LPS significantly inhibited MAC-T cell proliferation in a dose- and time-dependent manner. Furthermore, LPS promoted apoptosis while the NF-кB inhibitor PDTC attenuated this effect. After LPS treatment, the NF-кB signaling pathway was activated, and the expression of inflammation and apoptosis-related factors increased. When PDTC blocked NF-кB signaling, the expression of inflammation and apoptosis-related factors were decreased in MAC-T cells. CONCLUSIONS: LPS activates the TLR4/NF-κB signaling pathway, inhibits proliferation and promotes apoptosis in MAC-T cells. NF-кB inhibition attenuates MAC-T cell apoptosis and TLR4/NF-κB signaling pathway. NF-кB inhibitor alleviating MAC-T cell apoptosis is presumably modulated by NF-кB.


Asunto(s)
Células Epiteliales/citología , Lipopolisacáridos/farmacología , FN-kappa B/genética , FN-kappa B/metabolismo , Pirrolidinas/farmacología , Tiocarbamatos/farmacología , Acidosis/genética , Acidosis/metabolismo , Acidosis/patología , Animales , Apoptosis/efectos de los fármacos , Bovinos , Enfermedades de los Bovinos/genética , Enfermedades de los Bovinos/metabolismo , Enfermedades de los Bovinos/patología , Línea Celular , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos
3.
Tumour Biol ; 36(12): 9807-12, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26159853

RESUMEN

Eosinophil granule ontogeny transcript (EGOT) is a long noncoding RNA involved in the regulation of eosinophil granule protein transcript expression. However, little is known about the role of EGOT in malignant disease. This study aimed to assess the potential role of EGOT in the pathogenesis of breast cancer. Quantitative real-time polymerase chain reaction was performed to detect the expression levels of EGOT in 250 breast cancerous tissues and 50 adjacent noncancerous tissues. The correlation of EGOT expression with clinicopathological features and prognosis was also analyzed. EGOT expression was lower in breast cancer compared with the adjacent noncancerous tissues (P < 0.001), and low levels of EGOT expression were significantly correlated with larger tumor size (P = 0.022), more lymph node metastasis (P = 0.020), and higher Ki-67 expression (P = 0.017). Moreover, patients with low levels of EGOT expression showed significantly worse prognosis for overall survival (P = 0.040), and this result was further validated in a larger cohort from a public database. Multivariate analysis suggested that low levels of EGOT were a poor independent prognostic predictor for breast cancer patients (HR = 1.857, 95 % CI = 1.032-3.340, P = 0.039). In conclusion, EGOT may play an important role in breast cancer progression and prognosis and may serve as a new potential prognostic target in breast cancer patients.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias de la Mama/genética , Carcinoma de Células Escamosas/genética , ARN Largo no Codificante/biosíntesis , Adulto , Biomarcadores de Tumor/genética , Neoplasias de la Mama/patología , Carcinoma de Células Escamosas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Pronóstico , ARN Largo no Codificante/genética
4.
Nuklearmedizin ; 60(4): 289-298, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33638136

RESUMEN

The present study evaluated interstitial alterations in doxorubicin-induced cardiomyopathy using a radiolabeled RGD peptide 99mTc-3PRGD2 specific for integrin αvß3 that targets myofibroblasts.Cardiomyopathy was induced in 20 Sprague-Dawley rats by intraperitoneal doxorubicin injections (2.5 mg/kg/week) for up to six weeks. 99mTc-3PRGD2 scintigraphy was performed in control rats (n = 6) at baseline and three, six, and nine weeks after first doxorubicin administration (n = 6, 6, and 5 for each time point). For another three rats of 6-week modeling, cold c(RGDyK) was co-injected with 99mTc-3PRGD2 to evaluate specific radiotracer binding. Semi-quantitative parameters were acquired to compare radiotracer uptake among all groups. The biodistribution of 99mTc-3PRGD2 was evaluated by a γ-counter after scintigraphy. Haematoxylin and eosin, and Masson's staining were used to evaluate myocardial injury and fibrosis, while western blotting and immunofluorescence co-localization were used to analyze integrin αvß3 expression in the myocardium.The 99mTc-3PRGD2 half-life in the cardiac region (Heartt 1/2) of the 9-week model and heart radioactivity percentage (%Heart20 min, %Heart40 min and %Heart60 min) of the 6 and 9-week models were significantly increased compared to the control. Heart-to-background ratio (HBR20 min, HBR40 min and HBR60 min) increase began in the third week, continued until the sixth week, and was reversed in the ninth week, which paralleled the changing trend of cardiac integrin αvß3 expression. The myocardial biodistribution of 99mTc-3PRGD2 was significantly correlated with integrin ß3 expression.The 99mTc-3PRGD2 scintigraphy allows for non-invasive visualization of interstitial alterations during doxorubicin-induced cardiomyopathy.


Asunto(s)
Cardiomiopatías , Integrina alfaVbeta3 , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/diagnóstico por imagen , Doxorrubicina , Integrina alfaVbeta3/metabolismo , Compuestos de Organotecnecio , Cintigrafía , Ratas , Ratas Sprague-Dawley , Distribución Tisular
5.
Life Sci ; 253: 117694, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32325132

RESUMEN

AIMS: Chondrocyte degeneration is the main cause of osteoarthritis (OA) and increased evidence suggests that miRNAs could have vital roles in the pathology of various cartilage illnesses. miR-1236 has been found to contribute to inflammation in diseases such as pneumonia. However, the exact role of miR-1236 in OA is poorly understood. MATERIALS AND METHODS: H&E staining and saffron fixation experiments were employed to determine OA tissues. qRT-PCR and immunohistochemistry were used to detect the expression levels of miR-1236 and PIK3R3. Western blot was performed to detect the expression levels of proteins. Luciferase reporter assays were utilized to investigate the interaction between miR-1236 and PIK3R3. Cell counting assays and AO/EB were used to quantify cell growth and apoptosis. KEY FINDINGS: miR-1236 was up-regulated in OA knee cartilage compared to normal cartilage. Up-regulated expression of miR-1236 suppressed cell proliferation as well as induced apoptosis in chondrocytes. Bioinformatics identified PIK3R3 as a target of miR-1236. Co-transfection with miR-1236 and PIK3R3 could reverse cell apoptosis induced by the miR-1236 mimic. SIGNIFICANCE: These data enhance our understanding on the role of miR-1236 in OA and identifies miR-1236 as a potential biomarker or possible treatment target within OA.


Asunto(s)
Apoptosis/genética , Condrocitos/patología , MicroARNs/genética , Osteoartritis de la Rodilla/patología , Fosfatidilinositol 3-Quinasas/genética , Cartílago/patología , Proliferación Celular/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Osteoartritis de la Rodilla/genética , Regulación hacia Arriba
6.
Exp Ther Med ; 18(3): 1953-1960, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31410157

RESUMEN

Remote ischemic preconditioning (IPreC) is an effective strategy to defend against cerebral ischemia/reperfusion (IR) injury; however, its mechanisms remain to be elucidated. The aim of the present study was to investigate the effect of IPreC on brain tissue following cerebral ischemia, as well as the underlying mechanisms. Adult male Sprague-Dawley rats were treated with IPreC for 72 h prior to the induction of transient cerebral ischemia and reperfusion. The results demonstrated that IPreC reduced the area of cerebral infarction in the IR rats by 2,3,5-triphenyl-tetrazolium chloride staining. In addition, cell apoptosis was markedly suppressed by IPreC with an increased expression of B-cell lymphoma 2 (Bcl-2)/Bcl-2-associatd X protein using Terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling assay and western blot analysis. IR induced a decrease in the level of superoxide dismutase, and IPreC significantly suppressed increased levels of malondialdehyde, lactate dehydrogenase and nitric oxide. The expression of CD11b and CD18 was markedly inhibited by IpreC unsing flow cytometry. Furthermore, IPreC markedly decreased the release of pro-inflammatory factors interleukin (IL)-6 and IL-1ß, and enhanced the level of anti-inflammatory factors (IL-10 and IL-1 receptor antagonist) by ELISA assay. Finally, IPreC reduced the levels of transforming growth factor-ß-activated kinase 1, phosphorylated-P65/P65, and tumor necrosis factor-α, indicating that the nuclear factor-κB pathway was involved in IPreC-mediated protection against cerebral ischemia. Taken together, the results suggested that IPreC decreased ischemic brain injury through alleviating free radical injury and the inflammatory response in cerebral IR rats.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA