Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Environ Manage ; 355: 120424, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38430878

RESUMEN

As digital economy develops, its impact on green innovation and energy efficiency has become the focus of current research. To explore the impact of the current development of the digital economy on the energy industry, this paper selects the parameter of green innovation resilience, analyzes the impact mechanism of green innovation resilience on energy efficiency under the shock of digital economic development, and uses relevant data from 284 cities in China from 2011 to 2019 for empirical testing. It is found that: green innovation resilience promotes energy efficiency; low level of green innovation resilience inhibits the improvement of energy efficiency, while high level of resilience promotes energy efficiency; the initial stage of digital economic development generates resource grabbing and the effect of technological constraints, which weakens the role of green innovation resilience in promoting energy efficiency. The results indicate that the resilience of the green innovation system should be strengthened in order to fully tap the potential for promoting energy efficiency; the policy orientation of "digital greening-energy efficient" should be pursued in the development of digital economy; the rational allocation of resources and the implementation of green standards should be strengthened in the process of digital economic development; and the constraints on energy efficiency improvement in the early stage of digital economic development should be broken through by accelerating the digitalization process.


Asunto(s)
Conservación de los Recursos Energéticos , Resiliencia Psicológica , Desarrollo Económico , China , Ciudades , Eficiencia
2.
Immunopharmacol Immunotoxicol ; 45(6): 663-671, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37358084

RESUMEN

OBJECTIVE: Breast cancer (BC) causes cancer-related death in women. Sufentanil is used for cancer pain and postoperative analgesia. This study aimed to explore the role of sufentanil in BC. METHODS: BC cells were treated with sufentanil, and cell viability was evaluated using the cell counting kit-8 (CCK-8) assay. Biological behaviors were analyzed using EDU assay, flow cytometry, transwell assay, western blotting, and ELISA. The levels of NF-κB pathway-related factors were examined using western blotting. A xenograft tumor model was established to assess the effects of sufentanil on tumor growth in vivo. RESULTS: Sufentanil at the concentration of 20, 40, 80, and 160 nM suppressed cell viability (IC50 = 39.84 in MDA-MB-231 cells, and IC50 = 47.46 in BT549 cells). Sufentanil inhibited the proliferation, invasion, epithelial-mesenchymal transition (EMT), and inflammation, but induced apoptosis of BC cells. Mechanically, sufentanil suppressed the activation of the NF-κB pathway. Rescue experiments showed that RANKL (NF-κB receptor agonist) abrogated the effects induced by sufentanil. Moreover, sufentanil inhibited tumor growth, inflammatory response, but promoted apoptosis via the NF-κB pathway in vivo. CONCLUSIONS: Sufentanil decelerated the progression of BC by regulating the NF-κB pathway, suggesting sufentanil may be used in BC therapy.


Sufentanil treatment inhibited BC cell proliferation, invasion, epithelial-mesenchymal transition (EMT), immune response, but induced apoptosis.Sufentanil suppressed the activation of the NF-κB pathway.RANKL (NF-κB receptor agonist) abrogated the effects induced by sufentanil.Sufentanil inhibited tumor growth, proliferation, immune response and promoted apoptosis via the NF-κB pathway.


Asunto(s)
Neoplasias de la Mama , FN-kappa B , Humanos , Femenino , FN-kappa B/metabolismo , Neoplasias de la Mama/patología , Sufentanilo/farmacología , Transducción de Señal , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Inmunidad , Proliferación Celular , Movimiento Celular , Apoptosis
3.
Toxicol Appl Pharmacol ; 416: 115458, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33607128

RESUMEN

Morphine is an opioid agonist and a nonselective mu, kappa and delta receptor agonist. It is a commonly used analgesic drug for the treatment of acute and chronic pain as well as cancer pain. Morphine is particularly important to address the problem of morphine tolerance. Tcf7l2, known as a risk gene for schizophrenia and autism, encodes a member of the LEF1/TCF transcription factor family. TCF7L2 is an important transcription factor that is upregulated in neuropathic pain models. However, the relationship between TCF7L2 and morphine tolerance has not been reported. In this study, we found that morphine tolerance led to the upregulation of TCF7L2 in the spinal cord, and also led to the upregulation of TCF7L2 expression in glial cells, which promoted inflammation related signal, and activated TLR4 / NF-κB/NLRP3 pathway. In addition, TCF7L2 regulated microglial cell activation induced by chronic morphine treatment. Mechanically, we found that TCF7L2 transcriptionally regulated TLR4 expression, and the depletion of TCF7L2 alleviated morphine tolerance induced by chronic morphine treatment, and further alleviated pain hypersensitivity induced by chronic morphine treatment. We therefore suggested that TCF7L2 regulates the activation of TLR4/ NF-κB/NLRP3 pathway in microglia, and is involved in the formation of morphine tolerance. Our results provide a new idea for the regulation mechanism of morphine tolerance.


Asunto(s)
Analgésicos Opioides/toxicidad , Tolerancia a Medicamentos , Hiperalgesia/inducido químicamente , Microglía/efectos de los fármacos , Morfina/toxicidad , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Dolor Nociceptivo/prevención & control , Receptor Toll-Like 4/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Ratones , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/fisiopatología , Umbral del Dolor/efectos de los fármacos , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Receptor Toll-Like 4/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Regulación hacia Arriba
4.
Cancer Cell Int ; 21(1): 401, 2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321010

RESUMEN

BACKGROUNDS: As previously reported, midazolam anesthesia exerts tumor-suppressing effects in non-small cell lung cancer (NSCLC), but the regulating effects of this drug on cisplatin-resistance in NSCLC have not been studied. Thus, we designed this study to investigate this issue and preliminarily delineate the potential molecular mechanisms. METHODS: We performed MTT assay and trypan blue staining assay to measure cell proliferation and viability. Cell apoptosis was examined by FCM. qRT-PCR and immunoblotting were performed to determine the expression levels of genes. The targeting sites between genes were predicted by bioinformatics analysis and were validated by dual-luciferase reporter gene system assay. Mice tumor-bearing models were established and the tumorigenesis was evaluated by measuring tumor weight and volume. Immunohistochemistry (IHC) was used to examine the pro-proliferative Ki67 protein expressions in mice tumor tissues. RESULTS: The cisplatin-resistant NSCLC (CR-NSCLC) cells were treated with high-dose cisplatin (50 µg/ml) and low-dose midazolam (10 µg/ml), and the results showed that midazolam suppressed cell proliferation and viability, and promoted cell apoptosis in cisplatin-treated CR-NSCLC cells. In addition, midazolam enhanced cisplatin-sensitivity in CR-NSCLC cell via modulating the miR-194-5p/hook microtubule-tethering protein 3 (HOOK3) axis. Specifically, midazolam upregulated miR-194-5p, but downregulated HOOK3 in the CR-NSCLC cells, and further results validated that miR-194-5p bound to the 3' untranslated region (3'UTR) of HOOK3 mRNA for its inhibition. Also, midazolam downregulated HOOK3 in CR-NSCLC cells by upregulating miR-194-5p. Functional experiments validated that both miR-194-5p downregulation and HOOK3 upregulation abrogated the promoting effects of midazolam on cisplatin-sensitivity in CR-NSCLC cells. CONCLUSIONS: Taken together, this study found that midazolam anesthesia reduced cisplatin-resistance in CR-NSCLC cells by regulating the miR-194-5p/HOOK3 axis, implying that midazolam could be used as adjuvant drug for NSCLC treatment in clinical practices.

5.
Anesth Analg ; 131(4): 1270-1280, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32925348

RESUMEN

BACKGROUND: Propofol is a common sedative-hypnotic drug traditionally used for inducing and maintaining general anesthesia. Recent studies have drawn attention to the nonanesthetic effects of propofol, but the potential mechanism by which propofol suppresses non-small-cell lung cancer (NSCLC) progression has not been fully elucidated. METHODS: For the in vitro experiments, we used propofol (0, 2, 5, and 10 µg/mL) to treat A549 cells for 1, 4, and 12 hours and Cell Counting Kit-8 (CCK-8) to detect proliferation. Apoptosis was measured with flow cytometry. We also transfected A549 cells with an microribonucleic acid-21 (miR-21) mimic or negative control ribonucleic acid (RNA) duplex and phosphatase and tensin homolog, deleted on chromosome 10 (PTEN) small interfering ribonucleic acid (siRNA) or negative control. PTEN, phosphorylated protein kinase B (pAKT), and protein kinase B (AKT) expression were detected using Western blotting, whereas miR-21 expression was examined by real-time polymerase chain reaction (RT-PCR). In vivo, nude mice were given injections of A549 cells to grow xenograft tumors; 8 days later, the mice were intraperitoneally injected with propofol (35 mg/kg) or soybean oil. Tumors were then collected from mice and analyzed by immunohistochemistry and Western blotting. RESULTS: Propofol inhibited growth (1 hour, P = .001; 4 hours, P ≤ .0001; 12 hours, P = .0004) and miR-21 expression (P ≤ .0001) and induced apoptosis (1 hour, P = .0022; 4 hours, P = .0005; 12 hours, P ≤ .0001) in A549 cells in a time and concentration-dependent manner. MiR-21 mimic and PTEN siRNA transfection antagonized the suppressive effects of propofol on A549 cells by decreasing PTEN protein expression (mean differences [MD] [95% confidence interval {CI}], -0.51 [-0.86 to 0.16], P = .0058; MD [95% CI], 0.81 [0.07-1.55], P = .0349, respectively), resulting in an increase in pAKT levels (MD [95% CI] = -0.82 [-1.46 to -0.18], P = .0133) following propofol exposure. In vivo, propofol treatment reduced NSCLC tumor growth (MD [95% CI] = -109.47 [-167.03 to -51.91], P ≤ .0001) and promoted apoptosis (MD [95% CI] = 38.53 [11.69-65.36], P = .0093). CONCLUSIONS: Our study indicated that propofol inhibited A549 cell growth, accelerated apoptosis via the miR-21/PTEN/AKT pathway in vitro, suppressed NSCLC tumor cell growth, and promoted apoptosis in vivo. Our findings provide new implications for propofol in cancer therapy and indicate that propofol is extremely advantageous in surgical treatment.


Asunto(s)
Anestésicos Intravenosos/farmacología , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Propofol/farmacología , Transducción de Señal/efectos de los fármacos , Células A549 , Anestésicos Intravenosos/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/genética , Humanos , Neoplasias Pulmonares/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/efectos de los fármacos , Fosfohidrolasa PTEN/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Ann Surg Oncol ; 26(3): 884-893, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30565043

RESUMEN

BACKGROUND: The programmed cell death-1/programmed cell death-ligand 1 (PD-1/PD-L1) pathway has been shown to be involved in trauma-induced immunosuppression and to influence CD4+ T cell differentiation. MicroRNA (miR)-21 is a critical player in immune responses. However, it remains largely unknown whether miR-21 is regulated by PD-1 and influences CD4+ T-cell lineage choice after gastric cancer resection. METHODS: In the present study, we analyzed the percentages of T-helper (Th)-17/regulatory T (Treg) cells and PD-1/PD-L1 expression on peripheral blood mononuclear cells (PBMCs) during the perioperative period. We also detected the secretion of interleukin (IL)-17 and transforming growth factor (TGF)-ß1 using enzyme-linked immunosorbent assays (ELISAs). Furthermore, PBMCs isolated from patients were transfected with or without adenovirus-short hairpin-PD-1 (Ad-sh-PD1), pre-miR-21 or adenovirus-green fluorescent protein (Ad-GFP), and the percentages of Th17/Treg cells and related transcription factors were measured. RESULTS: In patients who underwent gastric cancer resection, the number of Th17 cells decreased, whereas the number of Treg cells increased, accompanied by an increased expression of PD-1/PD-L1. In addition, the expression of RORγt and IL-17 decreased, whereas the expression of Foxp3 and TGF-ß1 increased. In vitro, silencing PD-1 via Ad-sh-PD1 promoted the expression of miR-21 and increased the percentage of Th17 cells, but decreased the percentage of Treg cells. The overexpression of miR-21 increased the percentage of Th17 cells but decreased the percentage of Treg cells. CONCLUSIONS: Our study demonstrated that gastric cancer resection altered the balance of Th17/Treg cells and increased PD-1/PD-L1 expression. In the in vitro experiments, the transfection of Ad-sh-PD1 ameliorated Th17/Treg cell imbalance partially by increasing the expression of miR-21.


Asunto(s)
Antígeno B7-H1/metabolismo , Gastrectomía/mortalidad , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Receptor de Muerte Celular Programada 1/metabolismo , Neoplasias Gástricas/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Biomarcadores de Tumor/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Interleucina-17/metabolismo , Masculino , Persona de Mediana Edad , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Pronóstico , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/cirugía , Tasa de Supervivencia , Factor de Crecimiento Transformador beta1/metabolismo
7.
Microvasc Res ; 123: 1-6, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30179598

RESUMEN

BACKGROUND: Dexmedetomidine (DEX) is an α2-adrenergic receptor agonist commonly used during perioperative periods due to its sedation and analgesia effect. It is confirmed that DEX has cardioprotective effects against ischemia/reperfusion (I/R) injury. We investigated whether DEX administration is beneficial to type 2 diabetic rats subjected to I/R injury. METHODS: The diabetes model was established by providing a high-fat diet for 2 weeks followed by injecting 35 mg/kg streptozotocin (STZ). The myocardial I/R model consisted of left anterior descending coronary artery occlusion for 30 min followed by reperfusion for two-hours. DEX was administered before ischemia; alternatively, yohimbine was administered with or without DEX before ischemia. At the end of reperfusion, the rats were sacrificed, and hearts were isolated for histology. The levels of glycogen synthase kinase-3ß (GSK-3ß) and phosphorylated GSK-3ß (p-GSK-3ß) were quantitatively analyzed. The infarct size was measured via Evans Blue and 2,3,5­triphenyltetrazolium chloride (TTC) staining. Plasma samples were collected to measure the levels of cardiac Troponin T (cTnT). Arrhythmia scores were recorded during the first few minutes of reperfusion. RESULTS: DEX preconditioning significantly reduced myocardial infarct size, arrhythmia scores and the plasma cTnT levels, and increased the p-GSK-3ß levels. All of these protective effects of DEX were reversed by co-administration of yohimbine. CONCLUSIONS: These results suggested that DEX preconditioning exerted a cardioprotective effect against regional I/R injury in diabetic rats.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Arritmias Cardíacas/prevención & control , Dexmedetomidina/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Citoprotección , Diabetes Mellitus Experimental/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Fosforilación , Ratas Sprague-Dawley , Troponina T/sangre
8.
Br J Anaesth ; 123(6): 777-794, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31668347

RESUMEN

BACKGROUND: Dexmedetomidine (DEX) is a highly selective alpha2 adrenoceptor agonist with broad pharmacological effects, including sedation, analgesia, anxiolysis, and sympathetic tone inhibition. Here we report a systematic review and meta-analysis of its effects on stress, inflammation, and immunity in surgical patients during the perioperative period. METHODS: We searched MEDLINE, METSTR, Embase, and Web of Science for clinical studies or trials to analyse the effects of DEX on perioperative stress, inflammation, and immune function. RESULTS: Sixty-seven studies (including randomised controlled trials and eight cohort studies) with 4842 patients were assessed, of which 2454 patients were in DEX groups and 2388 patients were in control (without DEX) groups. DEX infusion during the perioperative period inhibited release of epinephrine, norepinephrine, and cortisol; decreased blood glucose, interleukin (IL)-6, tumour necrosis factor-α, and C-reactive protein; and increased interleukin-10 in surgical patients. In addition, the numbers of natural killer cells, B cells, and CD4+ T cells, and the ratios of CD4+:CD8+ and Th1:Th2 were significantly increased; CD8+ T-cells were decreased in the DEX group when compared with the control group. CONCLUSIONS: DEX, an anaesthesia adjuvant, can attenuate perioperative stress and inflammation, and protect the immune function of surgical patients, all of which may contribute to decreased postoperative complications and improved clinical outcomes.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Dexmedetomidina/farmacología , Inflamación/tratamiento farmacológico , Complicaciones Intraoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/tratamiento farmacológico , Estrés Fisiológico/efectos de los fármacos , Humanos , Inmunidad , Periodo Preoperatorio
9.
Anesth Analg ; 122(4): 1031-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26720619

RESUMEN

BACKGROUND: Morphine is widely used in patients with moderate and severe cancer pain, whereas the development of drug tolerance remains a major problem associated with opioid use. Previous studies have shown that cannabinoid type 2 (CB2) receptor agonists induce morphine analgesia, attenuate morphine tolerance in normal and neuropathic pain animals, induce transcription of the µ-opioid receptor (MOR) gene in Jurkat T cells, and increase morphine analgesia in cancer pain animals. However, no studies of the effects of CB2 receptor agonists on morphine tolerance in cancer pain have been performed. Therefore, we investigated the effect of repeated intrathecal (IT) injection of the low-dose CB2 receptor agonist AM1241 on the development of morphine tolerance in walker 256 tumor-bearing rats. We also tested the influence of the CB2 receptor agonist AM1241 on MOR protein and messenger ribonucleic acid (mRNA) expression in the rat spinal cord and dorsal root ganglia (DRG). METHODS: Walker 256 cells were implanted into the plantar region of each rat's right hindpaw. Tumor-bearing rats received IT injection of the CB2 receptor agonist AM1241 or antagonist AM630 with or without morphine subcutaneously twice daily for 8 days. Rats receiving drug vehicle only served as the control group. Mechanical paw withdrawal threshold and thermal paw withdrawal latency were assessed by a von Frey test and hot plate test 30 minutes after drug administration every day. MOR protein and mRNA expression in the spinal cord and DRG were detected after the last day (day 8) of drug administration via Western blot and real-time reverse transcription polymerase chain reaction. The data were analyzed via analysis of variance followed by Student t test with Bonferroni correction for multiple comparisons. RESULTS: Repeated morphine treatments reduced the mechanical withdrawal threshold and thermal latency. Coadministration of a nonanalgetic dose of the CB2 receptor agonist AM1241 with morphine significantly inhibited the development of morphine tolerance and increased the MOR protein expression in the spinal cord and DRG and mRNA expression in the spinal cord in tumor-bearing rats. CONCLUSIONS: Our findings indicate that IT injection of a nonanalgetic dose of a CB2 receptor agonist increased the analgesia effect and alleviated tolerance to morphine in tumor-bearing rats, potentially by regulating MOR expression in the spinal cord and DRG. This receptor may be a new target for prevention of the development of opioid tolerance in cancer pain.


Asunto(s)
Carcinoma 256 de Walker/metabolismo , Regulación Neoplásica de la Expresión Génica , Morfina/administración & dosificación , Receptor Cannabinoide CB2/agonistas , Receptores Opioides mu/biosíntesis , Animales , Cannabinoides/administración & dosificación , Tolerancia a Medicamentos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Ratas , Ratas Wistar , Receptores Opioides mu/genética
10.
BMC Pulm Med ; 16(1): 90, 2016 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-27260506

RESUMEN

BACKGROUND: Ventilation-induced lung injury (VILI) is a health problem for patients with acute respiratory dysfunction syndrome. The aim of this study was to investigate the effectiveness of budesonide in treating VILI. METHODS: Twenty-four rats were randomized to three groups: a ventilation group, ventilation/budesonide group, and sham group were ventilated with 30 ml/kg tidal volume or only anesthesia for 4 hor saline or budesonide airway instillation immediately after ventilation. The PaO2/FiO2and wet-to-dry weight ratios, protein concentration, neutrophil count, and neutrophil elastase levels in bronchoalveolar lavage fluid (BALF) and the levels of inflammation-related factors were examined. Histological evaluation of and apoptosis measurement inthe lung were conducted. RESULTS: Compared with that in the ventilation group, the PaO2/FiO2 ratio was significantly increased by treatment with budesonide. The lung wet-to-dry weight ratio, total protein, neutrophil elastase level, and neutrophilcount in BALF were decreased in the budesonide group. The BALF and plasma tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, intercellular adhesion molecule (ICAM)-1, and macrophage inflammatory protein (MIP)-2 levels were decreased, whereas the IL-10 level was increased in the budesonide group. The phosphorylated nuclear factor (NF)-kBlevels in lung tissue were inhibited by budesonide. The histological changes in the lung and apoptosis were reduced by budesonide treatment. Bax, caspase-3, and cleaved caspase-3 were down-regulated, and Bcl-2 was up-regulated by budesonide. CONCLUSIONS: Budesonide ameliorated lung injury induced by large volume ventilation, likely by improving epithelial permeability, decreasing edema, inhibiting local and systemic inflammation, and reducing apoptosis in VILI.


Asunto(s)
Budesonida/uso terapéutico , Glucocorticoides/uso terapéutico , Pulmón/fisiopatología , Respiración Artificial/efectos adversos , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Caspasa 3/sangre , Caspasa 3/química , Quimiocina CXCL2/sangre , Quimiocina CXCL2/química , Molécula 1 de Adhesión Intercelular/sangre , Molécula 1 de Adhesión Intercelular/química , Interleucina-10/sangre , Interleucina-10/química , Interleucina-1beta/sangre , Interleucina-1beta/química , Interleucina-6/sangre , Interleucina-6/química , Recuento de Leucocitos , Masculino , FN-kappa B/química , Proteínas Proto-Oncogénicas c-bcl-2/química , Distribución Aleatoria , Ratas , Ratas Wistar , Volumen de Ventilación Pulmonar , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/química , Proteína X Asociada a bcl-2/química
11.
Cell Physiol Biochem ; 35(4): 1454-66, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25791376

RESUMEN

BACKGROUND: The accumulation of cytokines in the plasma after trauma can induce myocyte apoptosis. We aimed to identify which cytokine(s) present in the plasma responsible for myocyte apoptosis, and delineated the signal transduction mechanism in rats subjected to surgical trauma. METHODS: Rats were randomized into two groups: control and trauma groups, which was divided into five subgroups: posttraumatic 0, 3, 6, 12, and 24 h subgroups. Cardiomyocytes isolated from traumatized rats were incubated with one of the factors for 12 h (normal plasma; Cytomix; TNF-α; IL-1ß; IFN-γ; trauma plasma; anti-TNF-α antibody; SB203580). Myocyte apoptosis, cytokine levels, and MAPKs activation, as the primary experimental outcomes, were measured by TUNEL, flow cytometry, ELISA and Western blot, respectively. RESULTS: Myocyte apoptosis was induced by surgical trauma during the early stage after trauma. Accompanying this change, plasma TNF-α, IL-1ß, and IFN-γ levels were elevated in traumatized rats. Incubation of traumatized cardiomyocytes with cytomix or TNF-α alone induced myocyte apoptosis, and increased the activation of p38 and ERK1/2. Myocyte apoptosis and p38 activation were elevated in traumatized cardiomyocytes with trauma plasma, and these increases were partly abolished by anti-TNF-α antibody or SB203580. CONCLUSION: Our study demonstrated that there exists the TNF-α-mediated-p38-dependent signaling pathway that contributed to posttraumatic myocyte apoptosis of rats undergoing surgical trauma.


Asunto(s)
Apoptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Heridas y Lesiones/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Anticuerpos/inmunología , Western Blotting , Caspasas/metabolismo , Células Cultivadas , Citocinas/análisis , Ensayo de Inmunoadsorción Enzimática , Imidazoles/farmacología , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/inmunología , Heridas y Lesiones/etiología , Heridas y Lesiones/metabolismo
12.
Cell Physiol Biochem ; 35(1): 126-36, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25591756

RESUMEN

BACKGROUND: There is an increasing interest in the role of astrocytes contributing to the intrinsic bioremediation of ischemic brain injury. The purpose of this study was to disclose the effects and mechanism of midazolam (MDZ) on the proliferation and apoptosis of astrocytes under oxygen glucose deprivation (OGD) condition. METHODS: The astrocytes were assigned randomly into four groups: control group, OGD group, OGD+MDZ group, and OGD+MDZ+IL-6 group. The astrocytes were treated with MDZ at dose of 10 µmol/L in OGD+MDZ group. And in OGD+MDZ+IL-6 group, the astrocytes were treated with MDZ at dose of 10 µmol/L and IL-6 at dose of 50 ng/mL. MTT assay was used to assess cell proliferation, and cell apoptosis was analyzed by TUNEL apoptosis assay kit and flow cytometry. Furthermore, the expression of JAK2, p-JAK2, STAT3, p-STAT3, Bcl-2, Bax and Caspase-3 proteins were determined by western blotting assay. RESULTS: Astrocytes proliferation was decreased obviously in OGD group, while MDZ could increase astrocytes proliferation under OGD condition. Moreover, OGD could induce apoptosis in astrocytes and MDZ could play an anti-apoptotic role. However, IL-6, a JAK2 activator, could attenuate cell proliferation and anti-apoptotic effects of MDZ in astrocytes. In addition, the expression of Bcl-2 protein in MDZ group increased markedly, while the JAK2/STAT3 signal proteins, Bax and Caspase-3 proteins decreased relative to OGD group. But IL-6 could counteract the anti-apoptotic effects of MDZ. CONCLUSION: Midazolam has protective effects on the proliferation and apoptosis of astrocytes via JAK2/STAT3 signal pathway in vitro. We firstly disclose the beneficial roles of midazolam in astrocytes under ischemic condition, which may be a rational treatment selection for ischemic cerebral protection.


Asunto(s)
Ansiolíticos/farmacología , Apoptosis/efectos de los fármacos , Midazolam/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Caspasa 3/metabolismo , Hipoxia de la Célula , Células Cultivadas , Glucosa/farmacología , Interleucina-6/farmacología , Janus Quinasa 2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Proteína X Asociada a bcl-2/metabolismo
13.
Cell Physiol Biochem ; 36(4): 1527-38, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26159236

RESUMEN

BACKGROUND/AIMS: The effects of H2S on cerebral inflammatory reaction after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) remain poorly understood. In this study, we investigated the effects of exogenous 40 ppm and 80 ppm H2S gas on inflammatory reaction and neurological outcome after CA/CPR. METHODS: CA was induced by ventricular fibrillation and followed by CPR. Forty or 80 ppm H2S was inhaled for 1 h immediately following CPR. The levels of IL-1ß, IL-6 and TNF-α, the myeloperoxidase (MPO) activity, the expression of iNOS and ICAM-1, and the phosphorylation and translocation of NF-κB were evaluated at 24 h after CA/ CPR. The tape removal test, survival rate and hippocampal neuronal counts were investigated at 14 d after CA/CPR. RESULTS: CA/CPR induced significant increases in IL-1ß, IL-6, TNF-α and MPO activity. The phosphorylation and translocation of NF-κB, and the expression of iNOS and ICAM-1 were increased significantly. Inhalation of 40 or 80 ppm H2S gas decreased these inflammatory cytokines. Furthermore, 40 or 80 ppm H2S inhibited the activation of NF-κB and the downstream proinflammatory mediators iNOS and ICAM-1. H2S inhalation also improved neurological function, 14-d survival rate, and reduced hippocampal neuronal loss. CONCLUSION: These results indicated that inhalation of H2S protected against brain injury after CA/CPR. The mechanisms underlying protective effects of H2S were associated with the inhibition of CA/ CPR-induced inflammation reactions by reducing IL-1ß, IL-6 and TNF-α, and concomitantly inhibiting the activation and infiltration of neutrophils. The beneficial effects of H2S might be mediated by downregulation of NF-κB and the downstream proinflammatory signaling pathway.


Asunto(s)
Lesiones Encefálicas/etiología , Lesiones Encefálicas/prevención & control , Gasotransmisores/uso terapéutico , Paro Cardíaco/complicaciones , Sulfuro de Hidrógeno/uso terapéutico , FN-kappa B/inmunología , Fármacos Neuroprotectores/uso terapéutico , Administración por Inhalación , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/patología , Reanimación Cardiopulmonar , Supervivencia Celular/efectos de los fármacos , Citocinas/inmunología , Gasotransmisores/administración & dosificación , Sulfuro de Hidrógeno/administración & dosificación , Masculino , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
14.
Cell Physiol Biochem ; 34(4): 1227-40, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25277843

RESUMEN

BACKGROUND: Cervical sympathetic blockade has been found to reduce cerebral vascular resistance and improve focal cerebral ischemia/reperfusion injury. In this study, we tested the hypothesis that the sympathetic blockade of high thoracic epidural anesthesia (HTEA) would reduce hippocampal apoptosis after global cerebral ischemia (GCI) injury. METHODS: Fifteen-minute global ischemia was established by 4-vessel occlusion in adult male Wistar rats. And 0.5% bupivacaine or 0.9% saline (20 µl//h) was infused continuously to the thoracic epidural space through the T4-5 intervertebral space from 15 minutes before ischemia to 24 hours or 72 hours after ischemia. Cerebral blood flow (CBF), Mortality, neurodeficit scores (NDS), Nissl and TUNEL staining, hippocampal superoxide dismutase (SOD) activity, malondialdehyde (MDA) concentrations, western blot of poly (ADP-ribose) polymerase (PARP) and immunohistochemical staining (PARP, Bax and Bcl-2) were determined. RESULTS: Both the hyperpefusion and hypoperfusion after reperfusion were improved by HTEA. HTEA decreased the number of apoptotic neurons in cornu ammonis area 1 (CA1), reduced PARP and Bax expressions with a decrease of Bax/Bcl-2 ratio induced by ischemic injury. The upregulation of SOD activity and the downregulation of MDA were obvious in the HTEA group compared with the GCI group. HTEA also improved NDS but not the mortality rate. CONCLUSION: Our study demonstrated that continuous HTEA attenuates hippocampal apoptosis and a behavioral deficit after global cerebral ischemia, and that these protective effects are associated with the improved microcirculation, reduced oxidative stress and the less activation of PARP.


Asunto(s)
Anestésicos Locales/farmacología , Apoptosis/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Bupivacaína/farmacología , Hipocampo/efectos de los fármacos , Anestesia Epidural/métodos , Animales , Apoptosis/genética , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Hipocampo/metabolismo , Masculino , Malondialdehído/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
15.
Can J Physiol Pharmacol ; 92(2): 155-61, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24502639

RESUMEN

Myocardial toxicity is one of the major side effects of many chemotherapeutics. It has been shown that propofol can ameliorate the cardiotoxicity of chemotherapeutic agents. In this study, we intend to investigate the role of the PI3K-Akt-Bad signaling pathway in propofol relief of doxorubicin-induced oxidative stress and apoptosis in rat cardiomyocytes. Cultured neonatal rat cardiomyocytes were treated with vehicle, doxorubicin, propofol, or propofol plus doxorubicin in the presence or absence of the PI3K inhibitor LY294002. Cells were harvested 20 h post-exposure to doxorubicin followed by analysis of their cellular taurine content, oxidative/nitrative stresses, and cellular apoptosis. The activation of the PI3K-Akt pathway was analyzed by immunoblotting. FACS, TUNEL, and LDH assays showed that the viability of cardiomyocytes was markedly reduced by doxorubicin, but was improved by propofol. Doxorubicin treatment significantly elevated cellular reactive oxygen and nitrogen contents while lowering the levels of taurine, Akt, and phosphorylated Akt and Bad. The abovementioned doxorubicin-induced changes were reversed by propofol. The protective effects of propofol were abrogated by simultaneous treatment with LY294002. In conclusion, the PI3K-Akt-Bad pathway plays a critical role in conferring the protective effects of propofol against myocardial toxicity from doxorubicin.


Asunto(s)
Anestésicos Intravenosos/farmacología , Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Propofol/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Taurina/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Humanos , Morfolinas/farmacología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Ratas , Ratas Sprague-Dawley
16.
Sci Rep ; 14(1): 6769, 2024 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-38514720

RESUMEN

Breast cancer is a serious threat to human health. The transforming growth factor-ß signaling pathway is an important pathway involved in the occurrence and development of cancer. The SMAD family genes are responsible for the TGF-ß signaling pathway. However, the mechanism by which genes of the SMAD family are involved in breast cancer is still unclear. Therefore, it is necessary to investigate the biological roles of the SMAD family genes in breast cancer. We downloaded the gene expression data, gene mutation data, and clinical pathological data of breast cancer patients from the UCSC Xena database. We used the Wilcox test to estimate the expression of genes of the SMAD family in cancers. And the biological functions of SMAD family genes using the DAVID website. The Pearson correlation method was used to explore the immune cell infiltration and drug response of SMAD family genes. We conducted in biological experiments vitro and vivo. In this study, we integrated the multi-omics data from TCGA breast cancer patients for analysis. The expression of genes of SMAD family was significantly dysregulated in patients with breast cancer. Except for SMAD6, the expression of other SMAD family genes was positively correlated. We also found that genes of the SMAD family were significantly enriched in the TGF-ß signaling pathway, Hippo signaling pathway, cell cycle, and cancer-related pathways. In addition, SMAD3, SMAD6, and SMAD7 were lowly expressed in stage II breast cancer, while SMAD4 and SMAD2 were lowly expressed in stage III cancer. Furthermore, the expression of genes of the SMAD family was significantly correlated with immune cell infiltration scores. Constructing a xenograft tumor mouse model, we found that SMAD3 knockdown significantly inhibited tumorigenesis. Finally, we analyzed the association between these genes and the IC50 value of drugs. Interestingly, patients with high expression of SMAD3 exhibited significant resistance to dasatinib and staurosporine, while high sensitivity to tamoxifen and auranofin. In addition, SMAD3 knockdown promoted the apoptosis of BT-549 cells and decreased cell activity, and BAY-1161909 and XK-469 increased drug efficacy. In conclusion, genes of the SMAD family play a crucial role in the development of breast cancer.


Asunto(s)
Neoplasias de la Mama , Transactivadores , Humanos , Animales , Ratones , Femenino , Transactivadores/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Transducción de Señal , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Proteínas Smad/genética , Proteínas Smad/metabolismo
17.
Exp Anim ; 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38797667

RESUMEN

Ischemia/reperfusion (I/R) is a pathological process that occurs in numerous organs and is often associated with severe cellular damage and death. Ectodysplasin-A2 receptor (EDA2R) is a member of the TNF receptor family that has anti-inflammatory and antioxidant effects. However, to the best of our knowledge, its role in the progression of myocardial I/R injury remains unclear. The present study aimed to investigate the role of EDA2R during myocardial I/R injury and the molecular mechanisms involved. In vitro, dexmedetomidine (DEX) exhibited a protective effect on hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury and downregulated EDA2R expression. Subsequently, EDA2R silencing enhanced cell viability and reduced the apoptosis of cardiomyocytes. Furthermore, knockdown of EDA2R led to an elevated mitochondrial membrane potential (MMP), repressed the release of Cytochrome C and upregulated Bcl-2 expression. EDA2R knockdown also resulted in downregulated expression of Bax, and decreased activity of Caspase-3 and Caspase-9 in cardiomyocytes, reversing the effects of H/R on mitochondria-mediated apoptosis. In addition, knockdown of EDA2R suppressed H/R-induced oxidative stress. Mechanistically, EDA2R knockdown inactivated the NF-κB signaling pathway. Additionally, downregulation of EDA2R weakened myocardial I/R injury in mice, as reflected by improved left ventricular function and reduced infarct size, as well as suppressed apoptosis and oxidative stress. Additionally, EDA2R knockdown repressed the activation of NF-κB signal in vivo. Collectively, knockdown of EDA2R exerted anti-apoptotic and antioxidant effects against I/R injury in vivo and in vitro by suppressing the NF-κB signaling pathway.

18.
JAMA Dermatol ; 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39320907

RESUMEN

Importance: China carries a heavy burden of postherpetic neuralgia, with an unmet need for novel drugs with greater efficacy and less prominent neurotoxic effects than existing calcium channel ligands. Objective: To investigate the efficacy and safety of crisugabalin, an oral calcium channel α2δ-1 subunit ligand, for postherpetic neuralgia. Design, Setting, and Participants: This randomized clinical trial, carried out between November 9, 2021, and January 5, 2023, at 48 tertiary care centers across China had 2 parts. Part 1 was a phase 3, multicenter, randomized, double-blind, placebo-controlled, parallel-group study consisting of a 2-week screening period, a 7-day run-in period, and a 12-week double-blind treatment period. Part 2 was a 14-week open-label extension study. Investigators, statisticians, trial clinicians, and patients were blinded to trial group assignments. Participants included adults with postherpetic neuralgia with an average daily pain score (ADPS) of at least 4 on the 11-point Numeric Pain Rating Scale over the preceding week, with the exclusion of patients with pain not controlled by prior therapy with pregabalin (≥300 mg/d) or gabapentin (≥1200 mg/d). Interventions: Patients were randomized 1:1:1 to receive crisugabalin, 20 mg twice daily (ie, 40 mg/d), and crisugabalin, 40 mg twice daily (ie, 80 mg/d), or placebo for 12 weeks. Eligible patients received crisugabalin, 40 mg, twice daily during extension. Main Outcome and Measure: The primary efficacy end point was the change from baseline in ADPS at week 12. Results: The study enrolled 366 patients (121 patients receiving crisugabalin, 40 mg/d; 121 patients receiving crisugabalin, 80 mg/d; 124 patients receiving placebo; median [IQR] age, 63.0 [56.0-69.0] years; 193 men [52.7%]). At week 12, the least squares mean (SD) change from baseline in ADPS was -2.2 (0.2) for crisugabalin, 40 mg/d, and -2.6 (0.2) for crisugabalin, 80 mg/d, vs -1.1 (0.2) for placebo, with a least squares mean difference of -1.1 (95% CI, -1.6 to -0.7; P < .001) and -1.5 (-95% CI, -2.0 to -1.0; P < .001) vs placebo, respectively. No new safety concerns emerged. Conclusions and Relevance: Crisugabalin, 40 mg/d, or crisugabalin, 80 mg/d, was well tolerated and demonstrated a statistically significant improvement in ADPS over placebo. Trial Registration: ClinicalTrials.gov Identifier: NCT05140863.

19.
Int J Med Sci ; 10(5): 599-606, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23569422

RESUMEN

Propofol (2, 6-diisopropylphenol), is an anesthetic and routinely used for the humans sedation during surgery. The potent inducers of phase II detoxifying and antioxidant stress responsive to propofol were investigated. First, a dose of 25-100 µM propofol showed no significant cytotoxicity on SH-SY5Y cells and pre-treatment of SH-SY5Y cells with propofol (25-100 µM) for 8h prevented cell death and maintained cell integrity following exposure to 1 mM hydrogen peroxide by MTT assays. Then, an increase in the generation of ROS following hydrogen peroxide treatment was significantly attenuated by 8 h pre-treatment with propofol. Additionally, the potential roles of ERK, p 38 MAPK and JNK in the regulation of propofol-induced endogenous HO-1 expression in SH-SY5Y cells were estimated by Western blotting assays. Results showed that propofol significantly increased the phosphorylation levels of ERK, p 38 MAPK and JNK and antioxidant stress responsive to propofol was attenuated by the inhibition of ERK signaling biochemical inhibitors. These results suggest that the ERK pathway plays an important role in the regulation of propofol-mediated antioxidant effects in SH-SY5Y cells.


Asunto(s)
Hemo-Oxigenasa 1/genética , Peróxido de Hidrógeno/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fase II de la Desintoxicación Metabólica/genética , Propofol/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
20.
Artículo en Inglés | MEDLINE | ID: mdl-37905205

RESUMEN

Purpose: Programmed cell death ligand 1 (PDL1) has the predictive and prognostic value in a great deal of cancers. This study aims to explore the expression of PDL1 in stage III breast cancer (BC) and its correlation with clinical outcome. Methods: The protein expression of PDL1 in tumor tissues was determined by immunohistochemistry (IHC). The correlations between PDL1 and clinicopathological variables were performed by χ²-tests or Fisher's exact tests. The Cox proportional hazards model was used for univariate and multivariate analysis of the potential prognostic factors. Survival curves were estimated based on Kaplan-Meier analyses, and Log Rank test was used to contrast factors influencing the survival outcome. Results: On the basis of the semiquantitative scoring method for PDL1 expression, the patients were divided into low PDL1 expression group (109 cases) and high PDL1 expression group (107 cases). PDL1 expression was correlated with positive lymph nodes, positive axillary lymph nodes, postoperative radiotherapy, and CK5/6 expression (P < 0.05). The PDL1 expression in tumor tissues was discovered to be a potential prognostic risk factor with the disease-free survival (DFS) and overall survival (OS) for stage III BC. Moreover, patients with high PDL1 expression showed longer lifetime (DFS and OS) compared to those with low PDL1 expression in total patient population (P < 0.05). Moreover, the nomogram showed that the prediction line is in good agreement with the reference line for postoperative 1-, 3-, and 5-year lifetime. The DCA curve showed that the 3- and 5-year lifetime by nomogram had so much better divination of the clinical application than only by PDL1. Conclusion: PDL1 is a latent prognostic factor in stage III BC and is closely related to some clinicopathological features. PDL1 expression in tumor tissues is significantly associated with better lifetime rate in stage III BC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA