Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Cell Mol Med ; 24(17): 10245-10250, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32813323

RESUMEN

Pulmonary fibrosis is a chronic interstitial lung disease characterized by pulmonary epithelial injury, fibroblast activation, extracellular matrix deposition, and tissue structure destruction. However, an effective drug treatment remains unavailable. Therefore, studying the mechanism of pulmonary fibrogenesis and finding effective drugs have become important problems in the field of respiratory diseases. Pulmonary fibrosis is typically characterized by activated fibroblast proliferation and migration. Hence, abnormality in activated fibroblast proliferation and migration is a major concern for treating pulmonary fibrosis. Long noncoding RNA (lncRNA) is an enigmatic subclass of ncRNA that regulates various fundamental biological processes and participates in disease occurrence and development. However, studies on lncRNA as the therapeutic target of drug action are rarely reported. Our group first identified differentially expressed lncRNAs and revealed that lncITPF is a highly upregulated lncRNA in lung fibrosis. In particular, lncITPF is detected in the blood of patients with idiopathic pulmonary fibrosis. Clinical analysis shows that lncITPF is positively correlated with the degree of fibrosis. The receiver operating characteristic (ROC) curve indicates that the specificity and sensitivity values are 95.0 and 64.3, respectively. The area under the ROC curve is 0.804, indicating that lncITPF can be a diagnostic biomarker for IPF. However, whether lncITPF is effective as a therapeutic target of drug action against pulmonary fibrosis remains unclear. In this study, lncITPF acting as the therapeutic target of astaxanthin was explored in depth. The findings elucidated that astaxanthin blocks the activated fibroblast proliferation and migration through lncITPF and mitochondria-mediated signal pathways to alleviate pulmonary fibrogenesis.


Asunto(s)
Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Pulmón/efectos de los fármacos , Mitocondrias/efectos de los fármacos , ARN Largo no Codificante/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Bleomicina/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Ratones , Mitocondrias/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Xantófilas/farmacología
2.
Pulm Pharmacol Ther ; 50: 19-27, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29621624

RESUMEN

BACKGROUND AND OBJECTIVE: The nature of pulmonary fibrosis involves inadequate repair of the epithelial cell barrier accompanied by impaired regulation of the fibroblast. Moreover, pulmonary fibrosis currently lacks an effective therapeutic drug. This study targets the protection of the epithelial cell and fibroblast to identify a novel, potentially therapeutic drug (i.e., astilbin). METHODS: In this study, the cytotoxicity of astilbin was firstly detected using CCK-8. A real-time proliferation/migration analysis system was used to test the inhibitory proliferation and migration of astilbin in vitro. The expression of mesenchymal markers and the loss of epithelial cell markers were analyzed to evaluate the antifibrotic activity of astilbin on TGF-ß1-treated AEC-II and L929 cells and bleomycin-treated mice. Then, in fibrosis-associated signaling pathways, the regulation of astilbin was tested using RNA sequencing and Cignal Finder 45-Pathway system. Rescue and other experiments were used to confirm this pathway regulation further. RESULTS: The data showed that astilbin inhibited proliferation and migration of cell samples. Its treatment resulted in the reduction of pathological score and collagen deposition, with a decrease in α-SMA and Snail and an increase in E-cadherin and SP-C in vivo and in vitro. The fibrosis-associated aberrant genes are some of the most notable components of the Hedgehog signaling pathway. CONCLUSIONS: Astilbin ameliorates pulmonary fibrosis via blockade of Hedgehog signaling pathway and has potential therapeutic value for lung fibrosis treatment.


Asunto(s)
Flavonoles/farmacología , Proteínas Hedgehog/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina/farmacología , Cadherinas/metabolismo , Línea Celular , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Fibroblastos/metabolismo , Humanos , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
3.
BMC Complement Altern Med ; 18(1): 234, 2018 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-30092799

RESUMEN

BACKGROUND: Feifukang (FFK) is a traditional Chinese medicine composed of herbs that protect lung function. However, difficulty arises regarding the clinical application of FFK due to the complex mechanism of Chinese medicines. This study aimed to investigate the efficacy of FFK and explore its targeted genes and pathways. METHODS: Histopathological changes and collagen deposition were measured to evaluate the effect of FFK on bleomycin-induced pulmonary fibrosis in mice. The differentially expressed targeted genes and pathways were first screened using RNA sequencing. Then network pharmacology and other experiments were conducted to confirm RNA sequencing data. RESULTS: FFK treatment reduced the pathological score and collagen deposition, with a decrease in α-SMA and collagen. RNA sequencing and network pharmacology results all showed that FFK can ameliorate pulmonary fibrosis through multi-genes and multi-pathways. The targeted genes in JAK-STAT signaling pathway are some of the most notable components of these multi-genes and multi-pathways. Further experiments illustrated that FFK regulated phosphorylation of SMAD3, STAT3 and JAK1, and their co-expressed lncRNAs, which all are the important genes in JAK-STAT signaling pathway. CONCLUSION: FFK can ameliorate pulmonary fibrosis by inhibiting JAK-STAT signaling pathway and has potential therapeutic value for lung fibrosis treatment. Our study provides a new idea for the study of traditional Chinese medicine.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Quinasas Janus/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Medicamentos Herbarios Chinos/uso terapéutico , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/metabolismo
4.
BMC Pulm Med ; 17(1): 163, 2017 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-29197377

RESUMEN

BACKGROUND: Aging is a known risk factor of idiopathic pulmonary fibrosis (IPF). However, the pathogenic mechanisms underlying the effects of advanced aging remain largely unknown. Telomeric repeat-containing RNA (TERRA) represents a type of long noncoding RNA. In this study, the regulatory roles of TERRA on human telomeres and mitochondria and IPF epithelial injury model were identified. METHODS: Blood samples were collected from patients with IPF (n = 24) and matched control individuals (n = 24). The significance of clinical research on the TERRA expression correlated with pulmonary fibrosis was assessed. The expression levels of TERRA in vivo and in vitro were determined through quantitative real-time polymerase chain reaction analysis. Telomerase activity was observed using a fluorescent quantitative TRAP assay kit. The functions of telomeres, mitochondria, and associated genes were analyzed through RNA interference on TERRA. RESULTS: TERRA expression levels significantly increased in the peripheral blood mononuclear cells of IPF patients. The expression levels also exhibited a direct and significantly inverse correlation with the percentage of predicted force vital capacity, which is a physiological indicator of fibrogenesis during IPF progression. This finding was confirmed in the epithelial injury model of IPF in vitro. RNA interference on TERRA expression can ameliorate the functions of telomeres; mitochondria; associated genes; components associated with telomeres, such as telomerase reverse transcriptase, telomerase, and cell nuclear antigen, cyclin D1; and mitochondria-associated cyclin E genes, including the MMP and Bcl-2 family. The RNA interference on TERRA expression can also improve the functions of oxidative-stress-associated genes, such as reactive oxygen species, superoxide dismutase, and catalase, and apoptosis-related genes, such as cytochrome c, caspase-9, and caspase-3. CONCLUSIONS: In this study, the regulation of TERRA expression on telomeres and mitochondria during IPF pathogenesis was identified for the first time. The results may provide valuable insights for the discovery of a novel biomarker or therapeutic approach for IPF treatment.


Asunto(s)
Envejecimiento/genética , Fibrosis Pulmonar Idiopática/genética , Mitocondrias/enzimología , ARN Largo no Codificante/genética , Telomerasa/metabolismo , Telómero/enzimología , Telómero/genética , Células A549/fisiología , Células A549/ultraestructura , Anciano , Animales , Apoptosis/efectos de los fármacos , Estudios de Casos y Controles , Catalasa/metabolismo , Proliferación Celular , Femenino , Humanos , Peróxido de Hidrógeno/farmacología , Fibrosis Pulmonar Idiopática/sangre , Fibrosis Pulmonar Idiopática/patología , Masculino , Ratones , Persona de Mediana Edad , Mitocondrias/ultraestructura , Interferencia de ARN , ARN Largo no Codificante/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Homeostasis del Telómero , Proteína p53 Supresora de Tumor/genética , Capacidad Vital/genética
5.
Pharm Biol ; 54(10): 1982-90, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26911316

RESUMEN

Context In clinical practice, the promotion of neuron survival is necessary to recover neurological functions after the onset of stroke. Objective This study aimed to investigate the post-ischaemic neuroprotective effect of SMND-309, a novel metabolite of salvianolic acid, on differentiated SH-SY5Y cells. Materials and methods SH-SY5Y cells were differentiated by pre-treating with 5 µM all-trans-retinoic acid for 6 d. The differentiated SH-SY5Y cells were exposed to oxygen-glucose deprivation (OGD) for 2 h and reperfusion (R) for 24 h to induce OGD/R injury. After OGD injury, differentiated SH-SY5Y cells were treated with or without SMND-309 (5, 10, 20 µM) for another 24 h. Cell viability was detected through Cell counting kit-8 assay and lactate dehydrogenase leakage assay. Apoptosis was evaluated through flow cytometry, caspase-3 activity assay. Changes in protein levels were assessed through Western blot. Results SMND-309 ameliorated the degree of injury in the differentiated SH-SY5Y cells by increasing cell viabilities (5 µM, 65.4% ± 4.1%; 10 µM, 69.8% ± 3.7%; 20 µM, 75.3% ± 5.1%) and by reducing LDH activity (20 µM, 2.5 fold) upon OGD/R stimulation. Annexin V-fluorescein isothiocyanate/propidium iodide staining results suggested that apoptotic rate of differentiated SH-SY5Y cells decreased from 43.8% induced by OGD/R injury to 19.2% when the cells were treated with 20 µM SMND-309. SMND-309 significantly increased the Bcl-2 level of the injured differentiated SH-SY5Y cells but decreased the caspase-3 activity of these cells by 1.6-fold. In contrast, SMND-309 did not affect the Bax level of these cells. SMND-309 evidently increased the protein expression of BDNF when Akt and CREB were activated. This function was antagonized by the addition of LY294002. Conclusion SMND-309 can prevent neuronal cell death in vitro. This process may be related to the activation of the PI3K/Akt/CREB-signalling pathway.


Asunto(s)
Ácidos Cafeicos/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glucosa/deficiencia , Humanos , Neuronas/enzimología , Neuronas/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología
6.
J Cell Mol Med ; 19(9): 2215-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26119034

RESUMEN

Promotion of myofibroblast apoptosis is a potential therapeutic strategy for pulmonary fibrosis. This study investigated the antifibrotic effect of astaxanthin on the promotion of myofibroblast apoptosis based on dynamin-related protein-1 (Drp1)-mediated mitochondrial fission in vivo and in vitro. Results showed that astaxanthin can inhibit lung parenchymal distortion and collagen deposition, as well as promote myofibroblast apoptosis. Astaxanthin demonstrated pro-apoptotic function in myofibroblasts by contributing to mitochondrial fission, thereby leading to apoptosis by increasing the Drp1 expression and enhancing Drp1 translocation into the mitochondria. Two specific siRNAs were used to demonstrate that Drp1 is necessary to promote astaxanthin-induced mitochondrial fission and apoptosis in myofibroblasts. Drp1-associated genes, such as Bcl-2-associated X protein, cytochrome c, tumour suppressor gene p53 and p53-up-regulated modulator of apoptosis, were highly up-regulated in the astaxanthin group compared with those in the sham group. This study revealed that astaxanthin can prevent pulmonary fibrosis by promoting myofibroblast apoptosis through a Drp1-dependent molecular pathway. Furthermore, astaxanthin provides a potential therapeutic value in pulmonary fibrosis treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , GTP Fosfohidrolasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Dinámicas Mitocondriales , Proteínas Mitocondriales/metabolismo , Miofibroblastos/patología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/prevención & control , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Recuento de Células , Línea Celular Tumoral , Modelos Animales de Enfermedad , Dinaminas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Dinámicas Mitocondriales/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Miofibroblastos/ultraestructura , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Fibrosis Pulmonar/patología , Ratas Sprague-Dawley , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Xantófilas/farmacología , Xantófilas/uso terapéutico , Proteína X Asociada a bcl-2/metabolismo
7.
Biol Pharm Bull ; 34(3): 343-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21372382

RESUMEN

Hepatic stellate cells (HSCs), activated during liver injury, are defined as the most important target in the therapy of hepatic fibrosis. In the present study, we evaluated the effect of Rosmarinic acid (RosA) on the proliferation and apoptosis in activated hepatic stellate cells (HSC-T6), which is useful to decrease this cell population. The proliferation of HSC-T6 was significantly inhibited after treated with various concentrations of RosA for different times. Flow cytometric analyses and transmission electron microscope (TEM) observations revealed that HSC-T6 treated with RosA underwent apoptosis in a time dependent manner and displayed typical apoptotic features in the cells. The phosphorylation in signal transducer and activator of transcription protein-3 (STAT3), which regulates cell survival, proliferation and differentiation in a variety of tissues, was markedly decreased as the result of Western blot assay and correlated with downregulation of CyclinD1 and B cell lymphoma/leukemia-2 (Bcl-2). In conclusion, these results suggested that RosA was able to inhibit proliferation and induce apoptosis in HSC-T6, partly due to the inhibition of phosphorylation in STAT3, which contributed to the reversal of hepatic fibrosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cinamatos/farmacología , Depsidos/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Cirrosis Hepática/metabolismo , Hígado/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Línea Celular , Cinamatos/uso terapéutico , Ciclina D1/metabolismo , Depsidos/uso terapéutico , Regulación hacia Abajo , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hígado/citología , Hígado/metabolismo , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Fosforilación , Fitoterapia , Extractos Vegetales/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína Letal Asociada a bcl/metabolismo , Ácido Rosmarínico
8.
Aging (Albany NY) ; 13(10): 13807-13821, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33929970

RESUMEN

Pulmonary fibrosis is a common pulmonary interstitial disease of pathogenesis without effective drugs for treatment. Therefore, discovering new and effective drugs is urgently needed. In the present study, we prepared a novel compound named acetyl oxygen benzoate engeletin ester (AOBEE), investigated its effect on experimental pulmonary fibrosis, and proposed a long non-coding RNA (lncRNA)-mediated mechanism of its action. Bleomycin-induced pulmonary fibrosis in mice exhibited that AOBEE improved forced vital capacity (FVC) and alveolar structure and inhibited α-SMA, vimentin, and collagen expression. TGFß1-stimulated fibroblast L929 cells showed that AOBEE reduced these fibrotic proteins expression and inhibited the activated-fibroblast proliferation and migration. Whole transcriptome sequencing was performed to screen out lncRNA-lnc865 and lnc556 with high expression under bleomycin treatment, but AOBEE caused a considerable decrease in lnc865 and lnc556. Mechanistic study elucidated that AOBEE alleviated pulmonary fibrosis through lnc865- and lnc556-mediated mechanism, in which both lnc865 and lnc556 sponged miR-29b-2-5p to target signal transducer and activator of transcription 3 (STAT3). Further signal pathway inhibitors and the Cignal Finder 45-pathway reporter array illustrated that the up- and downstream pathways were TGFß1-smad2/3 and p38MAPK, and Krüppel-like factor 4 (KLF4), respectively. In conclusion, AOBEE promoted KLF4 degradation leading to the attenuation of pulmonary fibrosis by inhibiting TGFß1-smad/p38MAPK-lnc865/lnc556-miR-29b-2-5p-STAT3 signal pathway. We hope this work will provide valuable information to design new drugs and therapeutic targets of lncRNAs for pulmonary fibrosis treatment.


Asunto(s)
Flavonoles/farmacología , Glicósidos/farmacología , Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs/metabolismo , Fibrosis Pulmonar/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Flavonoles/química , Glicósidos/química , Factor 4 Similar a Kruppel , Ratones Endogámicos C57BL , MicroARNs/genética , Modelos Biológicos , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo
9.
Int J Mol Med ; 42(6): 3256-3268, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30272257

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive type of interstitial pneumonia with unknown causes, poor prognosis and no effective therapy available. Circular RNAs (circRNAs), which serve as potential therapeutic targets and diagnostic biomarkers for certain diseases, represent a recent hotspot in the field of RNA research. In the present study, a total of 67 significantly dysregulated circRNAs were identified in the plasma of IPF patients by using a circRNA microarray. Among these circRNAs, 38 were upregulated, whereas 29 were downregulated. Further validation of the results by polymerase chain reaction analysis indicated that Homo sapiens (hsa)_circRNA_100906, hsa_circRNA_102100 and hsa_circRNA_102348 were significantly upregulated, whereas hsa_circRNA_101225, hsa_circRNA_104780 and hsa_circRNA_101242 were downregulated in plasma samples of IPF patients compared with those in samples from healthy controls. The majority of differentially expressed circRNAs were generated from exonic regions. The host genes of the differentially expressed circRNAs were involved in the regulation of the cell cycle, adherens junctions and RNA transport. The competing endogenous RNA (ceRNA) network of the circRNAs/micro(mi)RNAs/mRNAs indicated that circRNA­protected mRNA participated in transforming growth factor­ß1, hypoxia­inducible factor­1, Wnt, Janus kinase, Rho­associated protein kinase, vascular endothelial growth factor, mitogen­activated protein kinase, Hedgehog and nuclear factor κB signalling pathways or functioned as biomarkers for pulmonary fibrosis. Furthermore, luciferase reporter assays confirmed that hsa_circRNA_100906 and hsa_circRNA_102348 directly interact with miR­324­5p and miR­630, respectively, which were downregulated in IPF patients. The present study provided a novel avenue for exploring the underlying molecular mechanisms of IPF disease.


Asunto(s)
Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , MicroARNs/genética , ARN Mensajero/genética , ARN/genética , Anciano , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , ARN Circular , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
10.
Cell Death Dis ; 8(10): e3137, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29072702

RESUMEN

Emerging evidence suggests that microRNA (miRNA) and long noncoding RNA (lncRNA) play important roles in disease development. However, the mechanism underlying mRNA interaction with miRNA and lncRNA in idiopathic pulmonary fibrosis (IPF) remains unknown. This study presents a novel lnc-PCF that promotes the proliferation of TGF-ß1-activated epithelial cells through the regulation of map3k11 by directly targeting miR-344a-5p during pulmonary fibrogenesis. Bioinformatics and in vitro translation assay were performed to confirm whether or not lnc-PCF is an actual lncRNA. RNA fluorescent in situ hybridization (FISH) and nucleocytoplasmic separation showed that lnc-PCF is mainly expressed in the cytoplasm. Knockdown and knockin of lnc-PCF indicated that lnc-PCF could promote fibrogenesis by regulating the proliferation of epithelial cells activated by TGF-ß1 according to the results of xCELLigence real-time cell analysis system, flow cytometry, and western blot analysis. Computational analysis and a dual-luciferase reporter system were used to identify the target gene of miR-344a-5p, whereas RNA pull down, anti-AGO2 RNA immunoprecipitation, and rescue experiments were conducted to confirm the identity of this direct target. Further experiments verified that lnc-PCF promotes the proliferation of activated epithelial cells that were dependent on miR-344a-5p, which exerted its regulatory functions through its target gene map3k11. Finally, adenovirus packaging sh-lnc-PCF was sprayed into rat lung tissues to evaluate the therapeutic effect of lnc-PCF. These findings revealed that lnc-PCF can accelerate pulmonary fibrogenesis by directly targeting miR-344a-5p to regulate map3k11, which may be a potential therapeutic target in IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Proliferación Celular/fisiología , Células Epiteliales , Fibrosis Pulmonar Idiopática/enzimología , Fibrosis Pulmonar Idiopática/patología , ARN Largo no Codificante/genética , Ratas , Ratas Sprague-Dawley , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
11.
Brain Res ; 1263: 114-21, 2009 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-19368834

RESUMEN

(2E)-2-{6-[(E)-2-carboxylvinyl]-2,3-dihydroxyphenyl}-3-(3,4-dihydroxyphenyl) propenoic acid, a novel compound designated SMND-309, is a new degradation product of salvianolic acid B. The present study was conducted to evaluate whether SMND-309 has a protective effect on permanent focal cerebral ischemia in rats. The results showed that SMND-309 at doses higher than 4.0 mg/kg (i.v.) produced a significant neuroprotection in focal ischemia rats when administered 30 min after the onset of ischemia. SMND-309 (25.0 mg/kg, i.v.) demonstrated significant neuroprotective activity even after delayed administration at 1 h, 3 h and 6 h after ischemia. The neuroprotective effect of SMND-309 (25.0 mg/kg, bolus injection intravenous at 30 min after middle cerebral artery occlusion) was still present 7 days after ischemia. Meanwhile, SMND-309 significantly increased the brain ATP content, improved mitochondrial energy metabolism and mitochondrial respiratory chain complex activities and attenuated the elevation of malondialdehyde (MDA) content, the decrease in superoxide dismutase (SOD) and glutathione-peroxidase (GSH-Px) activity in brain mitochondria. All of these findings indicate that SMND-309 exerts potent and long-term neuroprotective effects with a favorable therapeutic time-window in the model of permanent cerebral ischemia, and its protective effects may be due to the amelioration of cerebral mitochondrial energy metabolism and the antioxidant property.


Asunto(s)
Ácidos Cafeicos/administración & dosificación , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Adenosina Trifosfato/metabolismo , Análisis de Varianza , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Ácidos Cafeicos/química , Relación Dosis-Respuesta a Droga , Transporte de Electrón/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Glutatión Peroxidasa/metabolismo , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Malondialdehído/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Mitocondrias/fisiología , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Superóxido Dismutasa/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA