Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Int J Biol Macromol ; 259(Pt 1): 129177, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38176488

RESUMEN

We reported the anti-cervical cancer effect of proprietary saponin content from seeds of Impatiens balsamina L., Hosenkoside A. Our study found that Hosenkoside A significantly promotes cell apoptosis and cell cycle arrest after administration, exhibiting anti-tumor effects. Then the transcriptome sequencing results after administration showed that Hosenkoside A had a significant inhibitory effect on Histone deacetylase 3 (HDAC3). After sufficient administration time, the inhibition of HDAC3 expression level leads to a significant decrease in lysine acetylation at histone 3 sites 4 and 9, blocking the activation of Signal transducer and activator of transcription 3 (STAT3) and achieving anti-tumor effects. In addition, we encapsulated Hosenkoside A into polypeptide metal complexes (PMC) to form slow-release spheres. This material breaks down in the tumor environment, not only does it solve the problem of low drug solubility, but it also achieves targeted sustained-release drug delivery. Under the same concentration of stimulation, the PMC complex group showed better anti-tumor effects in both in vitro and in vivo experiments.


Asunto(s)
Complejos de Coordinación , Neoplasias del Cuello Uterino , Femenino , Humanos , Complejos de Coordinación/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Histonas/metabolismo , Puntos de Control del Ciclo Celular , Péptidos/farmacología , Péptidos/metabolismo , Acetilación , Apoptosis , Línea Celular Tumoral
2.
PeerJ ; 12: e16701, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38188175

RESUMEN

Rapid urbanization has drawn some aquatic environments into the urban texture from the outskirts of cities, and the composition and distribution of plant species in urban wetlands along the urban gradient have changed. Understanding the drivers of these changes will help in the conservation and utilization of urban wetlands. This study investigated the differences in plant diversity and associated influencing factors in three wetlands, Xixi wetland, Tongjian Lake wetland, and Qingshan Lake wetland, which are located in a core area, fringe area, and suburban area of Hangzhou City, respectively. The results showed that a total of 104 families, 254 genera, and 336 species of plants were recorded in the Xixi wetland; 179 species, 150 genera, and 74 families were found in the Qingshan Lake wetland; and 112 species, 96 genera, and 57 families were collected in the Tongjian Lake wetland. The main plant species and flora distribution of the three urban wetlands showed similarities. Indigenous spontaneous vegetation was highest in the Xixi wetland, while cultivated plant species were most abundant in the Tongjian Lake wetland. The introduction of cultivated plants decreased the distance attenuation effect of plant communities, which led to a certain degree of plant diversity convergence among the three wetlands. Eight endangered plants were preserved in the Xixi wetland by planting them in suitable habitats. Ellenberg's indicator values showed that the proportion of heliophilous plants was higher in the Qingshan Lake wetland, while the proportion of thermophilous plants and nitrogen-loving plants in the Tongjian Lake wetland was higher than in the other two wetlands. The importance of artificial interference factors affecting the differences in plant diversity was significantly higher than that of natural environmental factors in urban wetlands. The preservation of spontaneous plants and the introduction of cultivated plants had an importance of 25.73% and 25.38%, respectively. These were the main factors influencing the plant diversity of urban wetlands. The management mode that did not interfere with spontaneous vegetation and confined maintenance to cultivated plants in the Xixi wetland was beneficial for improving wetland plant diversity. Scientific plant reintroduction can also improve wetland plant diversity.


Asunto(s)
Urbanización , Humedales , Humanos , Ciudades , China , Lagos
4.
Oncol Lett ; 21(2): 122, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33552243

RESUMEN

Ovarian cancer (OC) is a common malignant tumor of the female reproductive system. Long non-coding RNAs (lncRNAs) play an important role in OC occurrence and development. Thus, the function and potential mechanism of lncRNA small nucleolar RNA host gene 3 (SNHG3) was explored in the development of OC. The expression of SNHG3, microRNA (miR)-139-5p and Notch homolog 1, translocation-associated (Drosophila) (Notch1) in OC were detected by RT-qPCR or western blot assay. In addition, CCK-8 and wound-healing assays were used to detect OVCAR3 proliferation and migration ability. The targeting relationship of miR-139-5p with SNHG3 or Notch1 was verified through luciferase reporter assay. Rescue experiments were performed to confirm whether SNHG3 could mediate OVCAR3 proliferation and migration through miR-139-5p and Notch1. In OC tissues and cell lines, the expression of SNHG3 and Notch1 were significantly increased, and the expression of miR-139-5p was significantly decreased. SNHG3 inhibition suppressed the proliferation and migration of OVCAR3 cells. Luciferase reporter experiment confirmed that miR-139-5p could target SNHG3 and Notch1. Transfection of miR-139-5p inhibitor significantly reversed the inhibitory effect of SNHG3 knockdown on OVCAR3 proliferation and migration. Moreover, SNHG3 inhibition or miR-139-5p mimic abolished the promotion of Notch1 overexpression on OVCAR3 proliferation and migration. In conclusion, SNHG3 could accelerate the proliferation and migration of OC cells by regulating miR-139-5p and Notch1.

5.
Mol Oncol ; 15(1): 262-278, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33107691

RESUMEN

Increasing evidence demonstrates that Lin28B plays critical roles in numerous biological processes including cell proliferation and stemness maintenance. However, the molecular mechanisms underlying Lin28B nuclear translocation remain poorly understood. Here, we found for the first time that KRAS promoted Lin28B nuclear translocation through PKCß, which directly bound to and phosphorylated Lin28B at S243. Firstly, we observed that Lin28B was upregulated in pancreatic cancer, contributing to cellular migration and proliferation. Furthermore, nuclear Lin28B upregulated TET3 messenger RNA and protein levels by blocking the production of mature let-7i. Subsequently, increased TET3 expression could also promote the expression of Lin28B, thereby forming a Lin28B/let-7i/TET3 feedback loop. Our results suggest that the KRAS/Lin28B axis drives the let-7i/TET3 pathway to maintain the stemness of pancreatic cancer cells. These findings illuminate the distinct mechanism of Lin28B nuclear translocation and its important roles in KRAS-driven pancreatic cancer, and have important implications for development of novel therapeutic strategies for this cancer.


Asunto(s)
Dioxigenasas/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Modelos Biológicos , Células Madre Neoplásicas/patología , Proteína Quinasa C beta/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética
6.
J Cell Mol Med ; 24(2): 1969-1979, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31851779

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) has been implicated in the development and progression of human cancers. However, few studies reveal its role in epithelial-mesenchymal transition (EMT) of pancreatic cancer cells. In this study, we find that PRMT5 is up-regulated in pancreatic cancer, and promotes proliferation, migration and invasion in pancreatic cancer cells, and promotes tumorigenesis. Silencing PRMT5 induces epithelial marker E-cadherin expression and down-regulates expression of mesenchymal markers including Vimentin, collagen I and ß-catenin in PaTu8988 and SW1990 cells, whereas ectopic PRMT5 re-expression partially reverses these changes, indicating that PRMT5 promotes EMT in pancreatic cancer. More importantly, we find that PRMT5 knockdown decreases the phosphorylation level of EGFR at Y1068 and Y1172 and its downstream p-AKT and p-GSK3ß, and then results in down-regulation of ß-catenin. Expectedly, ectopic PRMT5 re-expression also reverses the above changes. It is suggested that PRMT5 promotes EMT probably via EGFR/AKT/ß-catenin pathway. Taken together, our study demonstrates that PRMT5 plays oncogenic roles in the growth of pancreatic cancer cell and provides a potential candidate for pancreatic cancer treatment.


Asunto(s)
Transición Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Proteína-Arginina N-Metiltransferasas/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Pancreáticas
7.
J Cell Biochem ; 121(1): 632-641, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31452251

RESUMEN

Large intergenic noncoding RNA regulator of reprogramming (Linc-RoR) was first identified as a regulator to increase the emergence of induced pluripotent stem cells through reprogramming differentiated cells and is abnormal expression in a variety of malignant tumors. However, the function of Linc-RoR in pancreatic cancer progression needs further clarification. The data from this study demonstrated that Linc-RoR knockdown suppressed cell proliferative capacity and colony formation, while Linc-RoR overexpression promoted these behaviors. In particular, Linc-RoR overexpression promoted the level of mesenchymal markers, inhibited the expression of epithelial markers, as well as enhanced pancreatic cancer cells migration and invasion, whereas Linc-RoR knockdown inhibited the expression of mesenchymal markers, promoted the expression of epithelial markers, as well as weakened pancreatic cancer cells migration and invasion. Further study revealed that Linc-RoR knockdown brought about a significant fall in YAP phosphorylation and a rise in total YAP, while Linc-RoR overexpression produced the opposite results. Specifically, Linc-RoR promoted YAP in the cytoplasm into the nucleus. Taken together, we conjectured that Linc-RoR promoted proliferation, migration, and invasion of pancreatic cancer cells by activating the Hippo/YAP pathway. YAP might be an underlying target of Linc-RoR and mediate epithelial-mesenchymal transition (EMT) in pancreatic cancer (PC); thus, Linc-RoR might be a very meaningful biomarker for PC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Largo no Codificante/genética , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Vía de Señalización Hippo , Humanos , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteínas Señalizadoras YAP
8.
Brain Res ; 1724: 146464, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31536729

RESUMEN

Visceral pain is a complex and common symptom of inflammatory bowel disease (IBD) patients. Developing novel efficient therapeutics is still a common interest for clinicians. Increasing evidence have shown that tumor necrosis factor (TNF) receptor associated factor 6 (TRAF6) contributes to the pathological pain state in some pain models. Resveratrol (RSV) has showed promising potential for the treatment of neuropathic pain and inflammatory pain. However, whether RSV has analgesic effect on visceral pain and the underlying mechanisms remain unclear. In this study, we established the colitis model through intrarectal administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS), and found that TNBS induced colonic inflammation and visceral hypersensitivity. Meanwhile, astroglial marker glial fibrillary acidic protein (GFAP), TRAF6, phosphorylation of NF-κB (pNF-κB), tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) levels were increased in L6-S1 spinal cord after TNBS enema. Then, intrathecal injection of TRAF6 siRNA attenuated visceral pain, blocked the upregulation of pNF-κB, TNF-α and IL-1ß levels in the spinal cord in TNBS mice. Furthermore, spinal administration of NF-κB inhibitor, BAY11-7082 reversed the pain behavior and suppressed spinal TNF-α and IL-1ß expression in TNBS mice. Finally, repeated intrathecal injection of RSV reversed TNBS-induced visceral pain hypersensitivity in a dose-dependent manner. Meanwhile, TNBS-induced enhancement of spinal GFAP, TRAF6, pNF-κB, TNF-α and IL-1ß were reduced by the same treatment of RSV. In conclusion, our results suggest that RSV exerts the effects of antinociception on colitis-induced visceral hyperalgesia through inhibition of spinal TRAF6/NF-κB signaling pathway and the production of inflammatory mediators in the spinal cord, suggesting a new application of RSV for the treatment of visceral pain.


Asunto(s)
Resveratrol/farmacología , Dolor Visceral/tratamiento farmacológico , Dolor Visceral/metabolismo , Analgésicos/farmacología , Animales , Colitis/tratamiento farmacológico , Colitis/fisiopatología , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Neuralgia/metabolismo , Resveratrol/metabolismo , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Am J Cancer Res ; 9(3): 496-510, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30949406

RESUMEN

Emerging evidence indicates that the long noncoding RNA UCA1 is upregulated in multiple cancers, including pancreatic ductal adenocarcinoma (PDAC), and plays a critical role in various complex biological processes. However, the functional roles of UCA1 in PDAC remain to be clarified. In the current study, we showed that UCA1 significantly promoted cell proliferation and tumor growth both in vitro and in vivo, and enhanced stemness maintenance of PDAC cell lines. Moreover, we found that UCA1 overexpression increased the activity and expression of oncogenic KRAS. Mechanistically, upregulated UCA1 increased phospho-KRAS protein levels by interacting with hnRNPA2B1, and KRAS facilitated high cytoplasmic accumulation of hnRNPA2B1. Additionally, we identified that UCA1 functioned as a competing endogenous RNA (ceRNA) to increase the expression of KRAS via sponging miR-590-3p, and in turn, KRAS promoted UCA1 expression. Collectively, these findings suggest that the UCA1-KRAS axis plays a crucial role in PDAC progression and that UCA1 may serve as a target for new PDAC therapies.

10.
Oncol Rep ; 40(2): 1093-1102, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29845201

RESUMEN

Recent studies have revealed that overexpression of long non­coding RNA (lncRNA) PVT1 is correlated with several types of cancer. However, its role in pancreatic cancer development remains to be clarified. In the present study, we found that PVT1 promoted the growth and the epithelial­mesenchymal transition (EMT) of pancreatic cancer cells. We first determined that PVT1 was upregulated in pancreatic cancer tissues compared with adjacent normal tissues. Knockdown of PVT1 inhibited viability, adhesion, migration and invasion. Furthermore, PVT1 knockdown reduced the expression of mesenchymal markers including Snail, Slug, ß­catenin, N­cadherin and vimentin, while it increased epithelial marker expression of E­cadherin. Finally, knockdown of PVT1 inhibited the TGF­ß/Smad signaling, including p­Smad2/3 and TGF­ß1 but enhanced the expression of Smad4. In contrast, overexpression of PVT1 reversed the process. These findings revealed that PVT1 acts as an oncogene in pancreatic cancer, possibly through the regulation of EMT via the TGF­ß/Smad pathway and PVT1 may serve as a potential target for diagnostics and therapeutics in pancreatic cancer.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Neoplasias Pancreáticas/genética , ARN Largo no Codificante/genética , Proteínas Smad/genética , Factor de Crecimiento Transformador beta/genética , Biomarcadores de Tumor/genética , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias Pancreáticas/patología , Regulación hacia Arriba/genética
11.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1770-1782, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29510195

RESUMEN

Although overexpression of the long non-coding RNA (lncRNA) UCA1 has been implicated in several human cancers, its biological function in pancreatic cancer remains to be clarified. In this study, we reported that UCA1 expression was significantly increased in pancreatic cancer tissues and correlated with clinicopathological features, tumor stage, and poorer patient outcome. We further showed that UCA1 promoted cell migration and invasion of pancreatic cancer cells. Importantly, we found that UCA1 overexpression inhibited YAP phosphorylation, and increased YAP expression. Mechanistically, UCA1 interacted with MOB1, Lats1, and YAP, forming shielding composites. Moreover, we demonstrated that UCA1 increased YAP nuclear localization and stabilization, and improved TEAD luciferase activity. In turn, YAP promotes UCA1 expression. Collectively, the present study provides insights into the mechanistic regulation of UCA1 promoting pancreatic cancer progression through the Hippo signaling pathway. UCA1 may serve as a candidate biomarker for poor prognosis and a target for new pancreatic cancer therapies.


Asunto(s)
Movimiento Celular , Neoplasias Pancreáticas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Largo no Codificante/metabolismo , ARN Neoplásico/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Vía de Señalización Hippo , Humanos , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , ARN Largo no Codificante/genética , ARN Neoplásico/genética , Factores de Transcripción , Proteínas Señalizadoras YAP
12.
Brain Res Bull ; 139: 235-242, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29550454

RESUMEN

Visceral pain, observed in inflammatory bowel disease (IBD) patients, is a challenging medical problem and remains poorly understood because the mechanisms underlying it are unclear. Emerging evidence indicates that microRNAs (miRNAs) play a crucial role in the pathogenesis of acute and chronic pain. In this study, we aimed to explore the potential role of miR-146a-5p (the mature form of miR-146a) in a mouse model of colitis induced by intracolonic injection of trinitrobenzene sulfonic acid (TNBS). We found that induction of colitis resulted in visceral hyperalgesia manifested by a decreased pain threshold to colorectal distension and upregulation of miR-146a-5p expression in the lumbosacral spinal cord. In situ hybridization and immunohistochemistry results showed that miR-146a-5p was colocalized with neuronal marker NeuN, but not with astrocytic marker GFAP or microglial marker IBA-1. Dual-luciferase reporter assay showed that miR-146a-5p directly targeted the 3'-untranslated region (UTR) of CCL8, which was previously identified as an important regulator of visceral pain. In cultured Neuro-2a cells, TNF-α-induced CCL8 upregulation was decreased by transfection of miR-146a-5p mimic dose-dependently. In vivo, exogenous supplementation of miR-146a-5p by intrathecal miR-146a-5p agomir significantly alleviated visceral pain and decreased CCL8 expression in colitis mice. Furthermore, inhibition of CCL8 expression by CCL8 siRNA relieved colitis-induced visceral nociception. Finally, in naïve mice intrathecal miR-146a-5p antagomir upregulated CCL8 expression and induced visceral pain hypersensitivity, which could be partially rescued by neutralization of CCL8. Taken together, the present findings indicate that miR-146a-5p may be an endogenous suppressor of visceral pain and exogenous supplementation of miR-146a-5p could exert an analgesic effect at least partly by targeting spinal CCL8 expression. Thus, miR-146a-5p may serve as a novel therapeutic target for visceral pain intervention in the context of colitis.


Asunto(s)
Quimiocina CCL8/metabolismo , Colitis/complicaciones , Regulación de la Expresión Génica/genética , MicroARNs/uso terapéutico , Médula Espinal/metabolismo , Dolor Visceral , Animales , Antagomirs/uso terapéutico , Anticuerpos/uso terapéutico , Células Cultivadas , Quimiocina CCL8/química , Quimiocina CCL8/genética , Quimiocina CCL8/inmunología , Colitis/inducido químicamente , Colitis/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/química , MicroARNs/metabolismo , Peroxidasa/metabolismo , ARN Interferente Pequeño/uso terapéutico , Médula Espinal/efectos de los fármacos , Ácido Trinitrobencenosulfónico/toxicidad , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Dolor Visceral/etiología , Dolor Visceral/patología , Dolor Visceral/terapia
13.
Oncol Lett ; 15(3): 3192-3196, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29435056

RESUMEN

Hexa-D-arginine (D6R), an inhibitor of furin, has potential therapeutic applications in different types of human tumor. However, the function of D6R in targeting pancreatic cancer cells remains to be elucidated. In the present study, the proliferation, invasion and migration abilities of SW1990 and PaTu8988 cells were examined using a Cell Counting Kit-8, and Transwell and wound healing assays. Subsequently, the expression of proteins associated with epithelial-mesenchymal transition (EMT) and the Hippo-yes-associated protein (YAP) pathway were detected using western blot analysis. It was revealed that D6R significantly inhibited the proliferation, migration and invasion abilities of SW1990 and PaTu8988 cells. Additionally, D6R led to the upregulation of E-cadherin (an epithelial marker), and the downregulation of N-cadherin and vimentin (mesenchymal markers) in SW1990 and PaTu8988 cells. Furthermore, the results of the present study revealed that D6R significantly affected the YAP phosphorylation level and the total YAP protein level, indicating that D6R was functionally involved in the Hippo-YAP signaling pathway. It has been suggested that D6R-suppressed EMT in SW1990 and PaTu8988 cells may occur via the Hippo-YAP pathway and that it may be a feasible drug to ameliorate the malignant phenotype of SW1990 and PaTu8988 cells.

14.
Exp Cell Res ; 362(1): 43-50, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29097184

RESUMEN

Protein arginine methyltransferase 1 (PRMT1) is up-regulated and promotes migration, invasion and proliferation in wide range of cancers. However, we for the first time identify that PRMT1 promotes migration and invasion and inhibits proliferation in gastric cancer cells, a phenomenon called "migration-proliferation dichotomy". First, we find that PRMT1 overexpression promotes migration and invasion and inhibits proliferation, whereas PRMT1 knockdown reverses the above abilities. Next, PRMT1 reduces the expression of epithelial marker E-cadherin and increases the expression of mesenchymal markers including N-cadherin, Vimentin, snail and ß-catenin in gastric cancer cells. Furthermore, our studies show that PRMT1 silencing promotes the phosphorylation of LATS1, and then induces YAP phosphorylation, while overexpression of PRMT1 down-regulates the phosphorylation of LATS1 and YAP, indicating that PRMT1 inhibits EMT probably via Hippo signaling. Collectively, the present study reveals important roles of PRMT1 in progression of gastric cancer. Given the dual functions of PRMT1, it is as a potential drug target of gastric cancer with extreme caution.


Asunto(s)
Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Proteína-Arginina N-Metiltransferasas/fisiología , Proteínas Represoras/fisiología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Movimiento Celular/genética , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Células Tumorales Cultivadas
15.
Brain Res Bull ; 135: 170-178, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29037608

RESUMEN

Visceral hypersensitivity induced by inflammatory bowel disease (IBD) is a clinical challenge since the underlying mechanisms remain elusive. Chemokines and their receptors have been suggested to modulate inflammatory pain and neuropathic pain. However, the exact chemokines involved in visceral pain remain to be determined. Here, we investigated the effects of spinal chemokine CCL8 and its major receptor CCR5 on the development of visceral hyperalgesia. We showed that intracolonic injection of 2,4,6-trinitrobenzene sulfonic acid (TNBS) in mice produced significant colonic inflammation and visceral hypersensitivity to colorectal distension. Moreover, the mRNA and protein expression of CCL8 and CCR5 in the lumbosacral spinal cord were significantly upregulated. Both of CCL8 and CCR5 were expressed in spinal neurons. Furthermore, TNBS induced the activation of extracellular signal-regulated kinase (ERK) in the spinal cord. The induction of visceral pain by TNBS was attenuated by injection of ERK upstream kinase (MEK) inhibitor PD98059. Finally, intrathecal CCL8 neutralizing antibody or CCR5 antagonist DAPTA dose-dependently suppressed TNBS-evoked visceral hyperalgesia and spinal ERK activation. Taken together, these data demonstrated that CCL8 and CCR5, expressed and upregulated in spinal neurons after colonic inflammation, are involved in the maintenance of visceral hyperalgesia via the activation of spinal ERK. Targeting CCL8/CCR5/ERK pathway in the spinal cord might provide a novel treatment for the relief of visceral pain.


Asunto(s)
Quimiocina CCL8/metabolismo , Colitis/fisiopatología , Dolor Visceral/metabolismo , Animales , Quimiocina CCL8/fisiología , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuralgia/metabolismo , Neuronas/metabolismo , Receptores CCR5/metabolismo , Receptores CCR5/fisiología , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Regulación hacia Arriba , Dolor Visceral/fisiopatología
16.
Int J Oncol ; 50(4): 1352-1362, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28259973

RESUMEN

Furin, a well-characterized proprotein convertase, plays an important role in many diseases and links to tumor metastasis. However, the role of furin in pancreatic cancer progression remains to be elucidated. In the present study, we found that furin promotes the growth and the epithelial-mesenchymal transition (EMT) of pancreatic cancer cells. First, we found that furin knockdown significantly inhibited proliferation, invasion and migration in BxPC3 and SW1990 cells, while furin overexpression promoted the above behavior in PANC1 and PaTu8988 cells. Further evidence revealed that furin knockdown resulted in the upregulation of E-cadherin (epithelial marker), and the downregulation of N-cadherin and Vimentin (mesenchymal markers) in BxPC3 and SW1990 cells, whereas furin overexpression remarkably led to the opposite effects in PANC1 and PaTu8988 cells. Furthermore, our data showed that Furin knockdown, furin inhibitor D6R or overexpression significantly affected YAP phosphoration level and total YAP protein level, indicating that furin was involved in Hippo-YAP pathway. It is suggested that furin promotes epithelial-mesenchymal transition in pancreatic cancer cells probably via Hippo-YAP pathway and may be a potential target for anti-pancreatic cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transición Epitelial-Mesenquimal , Furina/metabolismo , Neoplasias Pancreáticas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Furina/antagonistas & inhibidores , Furina/genética , Vía de Señalización Hippo , Humanos , Oligopéptidos/farmacología , Neoplasias Pancreáticas/patología , Fosfoproteínas/química , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Transcripción , Regulación hacia Arriba , Vimentina/metabolismo , Proteínas Señalizadoras YAP
17.
Mol Cell Biochem ; 431(1-2): 161-168, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28281184

RESUMEN

Although the function of miR-200a has been discussed in many cancers and fibrotic diseases, its role in pancreatic fibrosis is still poorly understood. In this study, we for the first time confirm that miR-200a attenuates TGF-ß1-induced pancreatic stellate cells activation and extracellular matrix formation. First, we find that TGF-ß1 induces activation and extracellular matrix (ECM) formation in PSCs, and the effects are blocked by the inhibitor of PI3K (LY294002). Furthermore, we identify that miR-200a is down-regulated in TGF-ß1-activated PSCs, and up-regulation of miR-200a inhibits PSCs activation induced by TGF-ß1. Meanwhile, TGF-ß1 inhibits the expression of the epithelial marker E-cadherin, and increases the expression of mesenchymal markers vimentin, and the expression of ECM proteins a-SMA and collagen I, while miR-200a mimic reversed the above effects in PSCs, indicating that miR-200a inhibits TGF-ß1-induced activation and epithelial-mesenchymal transition (EMT). In addition, overexpression of miR-200a promotes the expression of PTEN and decreases the expression of matrix proteins and attenuates phosphorylation of Akt and mTOR. Taken together, our study uncovers a novel mechanism that miR-200a attenuates TGF-ß1-induced pancreatic stellate cells activation and ECM formation through inhibiting PTEN /Akt/mTOR pathway.


Asunto(s)
Transición Epitelial-Mesenquimal , MicroARNs/metabolismo , Fosfohidrolasa PTEN/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Cromonas/farmacología , Matriz Extracelular/metabolismo , Fibrosis , Masculino , Morfolinas/farmacología , Células Estrelladas Pancreáticas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Oncotarget ; 8(15): 24978-24987, 2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28212565

RESUMEN

Pancreatic cancer (PC) is a highly lethal disease due to extensive metastatic lesions. Accumulating evidence suggests that miR-675-5p plays different roles in metastasis through the regulation of epithelial to mesenchymal (EMT) and the mesenchymal to epithelial transitions (MET) in different cancers. ZEB1 promotes the EMT process by controlling the expression of E-cadherin and may have a reciprocal regulation with Ubiquilin1 (UBQLN1) and mir-200 family in cancer progression. In the present study, we showed that decreased expression of miR-675-5p is associated with the enhanced cell proliferation and survival of PC cells, while the increased expression of mir-675-5p shows the opposite one. The mir-675-5p could decrease the expression of mir-200 which is intermediated by ZEB1, and increase the expression of UBQLN1 gene. The mir-675-5p can increase the expression of ZEB1 mRNA, but the ZEB1 protein level was decreased. When mir-675-5p mimics and siUBQLN1 were co-transfected into the pancreatic cancer Patu8988 cells, the expression of ZEB1 protein was increased. It suggests that mir-675-5p may affect ZEB1 in a post-transcriptional level which was verified to be regulated by UBQLN1 protein. Hence, mir-675-5p regulates the progression of pancreatic cancer cells through the UBQLN1-ZEB1-mir200 pathway.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , MicroARNs/metabolismo , Neoplasias Pancreáticas/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Relacionadas con la Autofagia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Progresión de la Enfermedad , Humanos , MicroARNs/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Transfección
19.
Cell Physiol Biochem ; 40(1-2): 277-286, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27855406

RESUMEN

BACKGROUND/AIMS: This study was to investigate the influence of scoparone on pancreatic fibrosis in vitro and in vivo. METHODS: Pancreatic stellate cells (PSCs) were isolated from pancreas tissue blocks, and cultured for 3-5 generations for the experiment. PSCs were treated with scoparone in different doses as experimental groups, salvianolic acid B as a positive control and PBS as a blank group. We measured the effects of scoparone on cellular proliferation, oxidative stress, epithelial-mesenchymal transition (EMT), and pancreatic fibrosis. Cellular oxidative stress was detected by using commercially available kits. The impact of scoparone on EMT and fibrosis was detected through immunofluorescence or western blotting. RESULTS: Compared with the control group, scoparone significantly inhibited stellate cell proliferation, and reduced MDA, the expression of mesenchymal makers, and increased the levels of SOD and the expression of E-cadherin (P < 0.05). Western blot analysis showed that scoparone downregulated the expression of TGF-ß and p-smad2/3, and upregultated the expression of smad7 (P < 0.05). CONCLUSION: Scoparone can reduce the levels of oxidative stress, repress pancreatic stellate cells activation, and alleviate fibrosis by regulating TGF-ß/Smad pathway.


Asunto(s)
Cumarinas/farmacología , Páncreas/metabolismo , Páncreas/patología , Sustancias Protectoras/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Separación Celular , Células Cultivadas , Cumarinas/uso terapéutico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibrosis , Masculino , Malondialdehído/metabolismo , Estrés Oxidativo/efectos de los fármacos , Páncreas/efectos de los fármacos , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Pancreatitis Crónica/tratamiento farmacológico , Pancreatitis Crónica/patología , Sustancias Protectoras/uso terapéutico , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo
20.
J Med Chem ; 59(14): 6772-90, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27396490

RESUMEN

Starting from the lead isodaphnetin, a natural product inhibitor of DPP-4 discovered through a target fishing docking based approach, a series of novel 2-phenyl-3,4-dihydro-2H-benzo[f]chromen-3-amine derivatives as potent DPP-4 inhibitors are rationally designed utilizing highly efficient 3D molecular similarity based scaffold hopping as well as electrostatic complementary methods. Those ingenious drug design strategies bring us approximate 7400-fold boost in potency. Compounds 22a and 24a are the most potent ones (IC50 ≈ 2.0 nM) with good pharmacokinetic profiles. Compound 22a demonstrated stable pharmacological effect. A 3 mg/kg oral dose provided >80% inhibition of DPP-4 activity within 24 h, which is comparable to the performance of the long-acting control omarigliptin. Moreover, the efficacy of 22a in improving the glucose tolerance is also comparable with omarigliptin. In this study, not only promising DPP-4 inhibitors as long acting antidiabetic that are clinically on demand are identified, but the target fish docking and medicinal chemistry strategies were successfully implemented.


Asunto(s)
Productos Biológicos/farmacología , Cromanos/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Descubrimiento de Drogas , Animales , Productos Biológicos/síntesis química , Productos Biológicos/química , Cromanos/síntesis química , Cromanos/química , Diabetes Mellitus Tipo 2/enzimología , Inhibidores de la Dipeptidil-Peptidasa IV/síntesis química , Inhibidores de la Dipeptidil-Peptidasa IV/química , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Obesos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA