Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biol Chem ; 299(11): 105335, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37827291

RESUMEN

Hepatoma-derived growth factor (HDGF) overexpression and uncontrolled reactive oxygen species (ROS) accumulation are involved in malignant transformation and poor prognosis in various types of cancer. However, the interplay between HDGF and ROS generation has not been elucidated in hepatocellular carcinoma. Here, we first analyzed the profile of HDGF expression and ROS production in newly generated orthotopic hepatomas by ultrasound-guided implantation. In situ superoxide detection showed that HDGF-overexpressing hepatomas had significantly elevated ROS levels compared with adjacent nontumor tissues. Consistently, liver tissues from HDGF-deficient mice exhibited lower ROS fluorescence than those from age- and sex-matched WT mice. ROS-detecting fluorescent dyes and flow cytometry revealed that recombinant HDGF (rHDGF) stimulated the production of superoxide anion, hydrogen peroxide, and mitochondrial ROS generation in cultured hepatoma cells in a dose-dependent manner. In contrast, the inactive Ser103Ala rHDGF mutant failed to promote ROS generation or oncogenic behaviors. Seahorse metabolic flux assays revealed that rHDGF dose dependently upregulated bioenergetics through enhanced basal and total oxygen consumption rate, extracellular acidification rate, and oxidative phosphorylation in hepatoma cells. Moreover, antioxidants of N-acetyl cysteine and MitoQ treatment significantly inhibited HDGF-mediated cell proliferation and invasive capacity. Genetic silencing of superoxide dismutase 2 augmented the HDGF-induced ROS generation and oncogenic behaviors of hepatoma cells. Finally, genetic knockdown nucleolin (NCL) and antibody neutralization of surface NCL, the HDGF receptor, abolished the HDGF-induced increase in ROS and mitochondrial energetics. In conclusion, this study has demonstrated for the first time that the HDGF/NCL signaling axis induces ROS generation by elevating ROS generation in mitochondria, thereby stimulating liver carcinogenesis.


Asunto(s)
Carcinoma Hepatocelular , Animales , Ratones , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Especies Reactivas de Oxígeno , Carcinogénesis/genética
2.
J Biol Chem ; 298(10): 102442, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36055405

RESUMEN

Leukocyte cell-derived chemotaxin 2 (LECT2) acts as a tumor suppressor in hepatocellular carcinoma (HCC). However, the antineoplastic mechanism of LECT2, especially its influence on hepatic cancer stem cells (CSCs), remains largely unknown. In The Cancer Genome Atlas cohort, LECT2 mRNA expression was shown to be associated with stage, grade, recurrence, and overall survival in human HCC patients, and LECT2 expression was downregulated in hepatoma tissues compared with the adjacent nontumoral liver. Here, we show by immunofluorescence and immunoblot analyses that LECT2 was expressed at lower levels in tumors and in poorly differentiated HCC cell lines. Using functional assays, we also found LECT2 was capable of suppressing oncogenic behaviors such as cell proliferation, anchorage-independent growth, migration, invasiveness, and epithelial-mesenchymal transition in hepatoma cells. Moreover, we show exogenous LECT2 treatment inhibited CSC functions such as tumor sphere formation and drug efflux. Simultaneously, hepatic CSC marker expression was also downregulated, including expression of CD133 and CD44. This was supported by infection with adenovirus encoding LECT2 (Ad-LECT2) in HCC cells. Furthermore, in animal experiments, Ad-LECT2 gene therapy showed potent efficacy in treating HCC. We demonstrate LECT2 overexpression significantly promoted cell apoptosis and reduced neovascularization/CSC expansion in rat hepatoma tissues. Mechanistically, we showed using immunoblot and immunofluorescence analyses that LECT2 inhibited ß-catenin signaling via the suppression of the hepatocyte growth factor/c-MET axis to diminish CSC properties in HCC cells. In summary, we reveal novel functions of LECT2 in the suppression of hepatic CSCs, suggesting a potential alternative strategy for HCC therapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Humanos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/terapia , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Neoplasias Hepáticas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Ratas , Terapia Genética
4.
FASEB J ; 34(12): 16163-16178, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33063394

RESUMEN

Hepatitis is an important health problem worldwide. Novel molecular targets are in demand for detection and management of hepatitis. Hepatoma-derived growth factor (HDGF) has been delineated to participate in hepatic fibrosis and liver carcinogenesis. However, the relationship between hepatitis and HDGF remains unclear. This study aimed to elucidate the role of HDGF during hepatitis using concanavalin A (ConA)-induced hepatitis model. In cultured hepatocytes, ConA treatment-elicited HDGF upregulation at transcriptional level and promoted HDGF secretion while reducing intracellular HDGF protein level and cellular viability. Similarly, mice receiving ConA administration exhibited reduced hepatic HDGF expression and elevated circulating HDGF level, which was positively correlated with serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. By using HDGF knockout (KO) mice, it was found the ConA-evoked cell death was prominently alleviated in KO compared with control. Besides, it was delineated HDGF ablation conferred protection by suppressing the ConA-induced neutrophils recruitment in livers. Above all, the ConA-mediated activation of tumor necrosis factor-α (TNF-α)/interleukin-1ß (IL-1ß)/interleukin-6 (IL-6)/cyclooxygenase-2 (COX-2) inflammatory signaling was significantly abrogated in KO mice. Treatment with recombinant HDGF (rHDGF) dose-dependently stimulated the expression of TNF-α/IL-1ß/IL-6/COX-2 in hepatocytes, further supporting the pro-inflammatory function of HDGF. Finally, application of HDGF antibody not only attenuated the ConA-mediated inflammatory cascade in hepatocytes, but also ameliorated the ConA-induced hepatic necrosis and AST elevation in mice. In summary, HDGF participates in ConA-induced hepatitis via neutrophils recruitment and may constitute a therapeutic target for acute hepatitis.


Asunto(s)
Concanavalina A/farmacología , Hepatitis Animal/inducido químicamente , Hepatitis Animal/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Alanina Transaminasa/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Células Cultivadas , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34502171

RESUMEN

The engineering of vascular regeneration still involves barriers that need to be conquered. In the current study, a novel nanocomposite comprising of fibronectin (denoted as FN) and a small amount of silver nanoparticles (AgNP, ~15.1, ~30.2 or ~75.5 ppm) was developed and its biological function and biocompatibility in Wharton's jelly-derived mesenchymal stem cells (MSCs) and rat models was investigated. The surface morphology as well as chemical composition for pure FN and the FN-AgNP nanocomposites incorporating various amounts of AgNP were firstly characterized by atomic force microscopy (AFM), UV-Visible spectroscopy (UV-Vis), and Fourier-transform infrared spectroscopy (FTIR). Among the nanocomposites, FN-AgNP with 30.2 ppm silver nanoparticles demonstrated the best biocompatibility as assessed through intracellular ROS production, proliferation of MSCs, and monocytes activation. The expression levels of pro-inflammatory cytokines, TNF-α, IL-1ß, and IL-6, were also examined. FN-AgNP 30.2 ppm significantly inhibited pro-inflammatory cytokine expression compared to other materials, indicating superior performance of anti-immune response. Mechanistically, FN-AgNP 30.2 ppm significantly induced greater expression of vascular endothelial growth factor (VEGF) and stromal-cell derived factor-1 alpha (SDF-1α) and promoted the migration of MSCs through matrix metalloproteinase (MMP) signaling pathway. Besides, in vitro and in vivo studies indicated that FN-AgNP 30.2 ppm stimulated greater protein expressions of CD31 and von Willebrand Factor (vWF) as well as facilitated better endothelialization capacity than other materials. Furthermore, the histological tissue examination revealed the lowest capsule formation and collagen deposition in rat subcutaneous implantation of FN-AgNP 30.2 ppm. In conclusion, FN-AgNP nanocomposites may facilitate the migration and proliferation of MSCs, induce endothelial cell differentiation, and attenuate immune response. These finding also suggests that FN-AgNP may be a potential anti-inflammatory surface modification strategy for vascular biomaterials.


Asunto(s)
Antiinflamatorios/administración & dosificación , Diferenciación Celular/efectos de los fármacos , Fibronectinas/administración & dosificación , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Nanopartículas del Metal , Plata , Animales , Proliferación Celular , Células Cultivadas , Citoesqueleto , Células Endoteliales/metabolismo , Inmunohistoquímica , Metaloproteinasas de la Matriz/metabolismo , Células Madre Mesenquimatosas/citología , Nanopartículas del Metal/ultraestructura , Tamaño de la Partícula , Ratas , Especies Reactivas de Oxígeno/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier
6.
Toxicol Appl Pharmacol ; 402: 115129, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32673656

RESUMEN

Urothelial carcinoma (UC) is one of the highest incidence cancers that rank the fourth commonly diagnosed tumors worldwide. The unresectable lesions that are resistant to therapeutic interventions is the major cause leading to death. Previous studies had shown that the resistance and metastatic consequence may arise from cancer stem-like cells population. The phytochemical flavonoids have promised bioactivity and potent anti-carcinogenic effects, and trap great attentions for cancer chemoprevention and/or adjuvant chemotherapy. However, the mechanisms of flavonoids on cancer stemness is still obscured. In this study, we analyzed the biofunctional effects of as-prepared flavonoid derivative-WYC0209 on T24, BFTC905 and BFTC909 human UC cell lines. Our results demonstrated that WYC0209 significantly induced anti-cell viability on UC cells through decreased Akt/NFkB signaling. Moreover, WYC0209 enhanced the cell apoptosis through activated the caspase-3 activity and inactivated Bcl-xL expression. Interestingly, WYC0209 dramatically inhibited the cancer stem cells (CSCs) traits, including attenuation of side population and tumorsphere formation in which were through declined EMT-CSCs markers including MDR1, ABCG2 and BMI-1. We further validated the effects of WYC0209 on several CSC surface markers including CD133, CD44, SOX-2 and Nanog. Our results showed that WYC0209 markedly inhibited CD133 expressions in both transcriptional and translational levels. High expression levels of CD133 was also demonstrated in human upper tract UC specimens. In summary, our study showed that WYC0209 may potentially as an adjuvant agent to against CD133-driven UC CSCs and provide a beneficial strategy to against UC cancer therapeutics resistant.


Asunto(s)
Antígeno AC133/metabolismo , Ciclohexanonas/farmacología , Flavonas/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Urotelio/citología , Antígeno AC133/genética , Biomarcadores de Tumor , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quimioterapia Adyuvante , Ciclohexanonas/química , Flavonas/química , Humanos , Estructura Molecular , Estudios Retrospectivos , Neoplasias de la Vejiga Urinaria
7.
Int J Mol Sci ; 21(21)2020 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-33114317

RESUMEN

Melanoma is notoriously resistant to current cancer therapy. However, the chemoresistance mechanism of melanoma remains unclear. The present study unveiled that chemotherapy drug cisplatin induced the formation of giant cells, which exhibited enlargement in cell diameter and nucleus in mice and human melanoma cells. Giant cells were positive with melanoma maker S100 and cancer stem cell markers including ABCB5 and CD133 in vitro and in vivo. Moreover, giant cells retained the mitotic ability with expression of proliferation marker Ki-67 and exhibited multiple drug resistance to doxorubicin and actinomycin D. The mitochondria genesis/activities and cellular ATP level were significantly elevated in giant cells, implicating the demand for energy supply. Application of metabolic blockers such as sodium azide or 2-deoxy glucose abolished the cisplatin-induced giant cells formation and expression of cancer stemness markers. The present study unveils a novel chemoresistance mechanism of melanoma cells via size alteration and the anti-neoplastic strategy by targeting giant cells.


Asunto(s)
Adenosina Trifosfato/metabolismo , Cisplatino/administración & dosificación , Resistencia a Antineoplásicos , Células Gigantes/patología , Melanoma/tratamiento farmacológico , Antígeno AC133/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular Tumoral , Cisplatino/farmacología , Desoxiglucosa/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Gigantes/efectos de los fármacos , Células Gigantes/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Melanoma/metabolismo , Melanoma/patología , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Proteínas S100/metabolismo , Azida Sódica/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Angiogenesis ; 21(4): 901, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29748783

RESUMEN

In the original publication of the article, there is an error in one of the citations in the Discussion section.

9.
Angiogenesis ; 21(2): 299-312, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29383634

RESUMEN

AIM: Delta-like 1 homolog (DLK1) is a non-canonical ligand of Notch signaling, which plays a pivotal role in vascular development and tumor angiogenesis. This study aimed to elucidate the function and mechanism of DLK1 in angiogenesis. METHODS AND RESULTS: By using in situ hybridization and immunohistochemical studies, expression analysis revealed a unique vascular tropism of DLK1 in vasculature of neuroblastoma and vascular tumors. Thus, it was hypothesized that DLK1 may be cleaved and then bound to endothelial cells, thereby regulating the endothelial function. To test such hypothesis, soluble DLK1 encompassing DLK1 extracellular domain (DLK1-EC) was generated and validated by its inhibitory function in adipogenesis assay. Recombinant DLK1-EC exhibited the preferential binding capability toward endothelial cells and stimulated the microvessels sprouting in aorta rings. Above all, implantation of DLK1-EC dose-dependently elicited the cornea neovascularization in rats. By using various angiogenesis assays, it was delineated that DLK1-EC stimulated the angiogenesis by promoting the proliferation, motility and tube formation of endothelial cells. By immunoblot and luciferase analysis, it was elucidated that DLK1-EC enhanced the expression and activities of Notch1/Akt/eNOS/Hes-1 signaling in dose- and time-dependent manners. Pharmaceutical blockage of Notch signaling using γ-secretase inhibitor DAPT abrogated the DLK1-EC-induced endothelial migration and Hes-1-driven luciferase activities. Furthermore, Notch1 inactivation by neutralizing antibodies or RNA interference reversed the DLK1-EC-induced angiogenesis. CONCLUSIONS: The present study unveils the pro-angiogenic function and mechanism of soluble DLK1 through activation of Notch1 signaling in endothelial cells.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Patológica/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Notch1/metabolismo , Animales , Proteínas de Unión al Calcio , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Neovascularización Patológica/patología , Ratas , Ratas Sprague-Dawley
10.
Nanomedicine ; 14(3): 699-711, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29325741

RESUMEN

Vascularization of engineered tissues remains one of the key problems. Here, we described a novel approach to promote vascularization of engineered tissues using fibronectin (FN) incorporated gold nanoparticles (AuNP) coated onto catheters with mesenchymal stem cells (MSCs) for tissue engineering. We found that the FN-AuNP composite with 43.5 ppm of AuNP exhibited better biomechanical properties and thermal stability than pure FN. FN-AuNP composites promoted MSC proliferation and increased the biocompatibility. Mechanistically, vascular endothelial growth factor (VEGF) promoted MSC migration on FN-AuNP through the endothelial oxide synthase (eNOS)/metalloproteinase (MMP) signaling pathway. Vascular femoral artery tissues isolated from the implanted FN-AuNP-coated catheters with MSCs expressed substantial CD31 and alpha-smooth muscle actin (α-SMA), displayed higher antithrombotic activity, as well as better endothelialization ability than those coated with all other materials. These data suggested that the implantation of FN-AuNP-coated catheter with MSCs could be a novel strategy for vascular biomaterials applications.


Asunto(s)
Arteria Femoral/citología , Fibronectinas/química , Oro/química , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Nanopartículas del Metal/administración & dosificación , Ingeniería de Tejidos/métodos , Catéteres , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Arteria Femoral/fisiología , Humanos , Ensayo de Materiales , Nanopartículas del Metal/química , Regeneración , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas
11.
Nanotechnology ; 27(41): 415702, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27585675

RESUMEN

Accumulation of amyloid fibrils is one of the likely key factors leading to the development of Alzheimer's disease and other amyloidosis associated diseases. Magnetic nanoparticles (NPs) have been developed as promising medical materials for many medical applications. In this study, we have explored the effects of Fe3O4 NPs on the fibrillogenesis process of insulin fibrils. When Fe3O4 NPs were co-incubated with insulin, Fe3O4 NPs had no effect on the structural transformation into amyloid-like fibrils but had higher affinity toward insulin fibrils. We demonstrated that the zeta potential of insulin fibrils and Fe3O4 NPs were both positive, suggesting the binding forces between Fe3O4 NPs and insulin fibrils were van der Waals forces but not surface charge. Moreover, a different amount of Fe3O4 NPs added had no effect on secondary structural changes of insulin fibrils. These results propose the potential use of Fe3O4 NPs as therapeutic agents against diseases related to protein aggregation or contrast agents for magnetic resonance imaging.


Asunto(s)
Nanopartículas de Magnetita , Amiloide , Insulina
12.
Phys Chem Chem Phys ; 18(10): 7124-31, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26886764

RESUMEN

Cells reorganize their membrane biomechanical dynamics in response to environmental stimuli or inhibitors associated with their physiological/pathological processes, and disease therapeutics. To validate the biophysical dynamics during cell exposure to anti-cancer drugs, we investigate the nanoscale biological characterization in melanoma cells undergoing cisplatin treatment. Using atomic force microscopy, we demonstrate that the cellular morphology and membrane ultrastructure are altered after exposure to cisplatin. In contrast to their normal spindle-like shape, cisplatin causes cell deformation rendering cells flat and enlarged, which increases the cell area by 3-4 fold. Additionally, cisplatin decreases the topography height values for both the cytoplasmic and nuclear regions (by 40-80% and 60%, respectively). Furthermore, cisplatin increases the cytoplasmic root mean square roughness by 110-240% in correlation with the drug concentration and attenuates the nuclear RMS by 60%. Moreover, the cellular adhesion force was enhanced, while the Young's modulus elasticity was attenuated by ∼2 and ∼2.3 fold, respectively. F-actin phalloidin staining revealed that cisplatin enlarges the cell size through enhanced stress fiber formation and promotes cytoskeletal reorganization. Immunoblot analyses further revealed that the activities of focal adhesion proteins, such as FAK and c-Src, are upregulated by cisplatin through phosphorylation at tyrosine 397 and 530, respectively. Collectively, these results show that cisplatin-treated melanoma cells not only exhibit the upregulation of FAK-mediated signaling to enhance the cytoskeleton mechanical stretch, but also promote the cytoskeletal rearrangement resulting in 43% decrease in the cell modulus. These mechanisms thus promote the malignancy and invasiveness of the melanoma cells.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Melanoma Experimental/patología , Nanotecnología , Animales , Fenómenos Biomecánicos , Adhesión Celular , Línea Celular Tumoral , Tamaño de la Célula , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Melanoma Experimental/metabolismo , Ratones , Microscopía de Fuerza Atómica , Transducción de Señal , Familia-src Quinasas/metabolismo
13.
J Biotechnol ; 392: 48-58, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38906221

RESUMEN

Combining phytochemicals and nanotechnology to improve the unfavorable innate properties of phytochemicals and develop them into potent nanomedicines to enhance antitumor efficacy has become a novel strategy for cancer chemoprevention. Melanoma is the most aggressive, metastatic, and deadly disease of the primary cutaneous neoplasms. In this study, we fabricated phytoconstituent-derived zingerone nanoparticles (NPs) and validated their effects on cell adhesion and motility in melanoma B16F10 cells. Our data indicated that zingerone NPs significantly induced cytotoxicity and anti-colony formation and inhibited cell migration and invasion. Moreover, zingerone NPs dramatically interfered with the cytoskeletal reorganization and markedly delayed the period of cell adhesion. Our results also revealed that zingerone NPs-mediated downregulation of MMPs (matrix metalloproteinases) activity is associated with inhibiting cell adhesion and motility. We further evaluated the effects of zingerone NPs on Src/FAK /Paxillin signaling, our data showed that zingerone NPs significantly inhibited the protein activities of Src, FAK, and Paxillin, indicating that they play important roles in zingerone NP-mediated anti-motility and anti-invasion in melanoma cells. Accordingly, the phytoconstituent-zingerone NPs can strengthen the inhibition of tumor growth, invasion, and metastasis in malignant melanoma. Altogether, these multi-pharmacological benefits of zingerone NPs will effectively achieve the purpose of melanoma prevention and invasion inhibition.


Asunto(s)
Adhesión Celular , Movimiento Celular , Guayacol , Melanoma Experimental , Nanopartículas , Animales , Movimiento Celular/efectos de los fármacos , Nanopartículas/química , Ratones , Guayacol/análogos & derivados , Guayacol/farmacología , Guayacol/química , Línea Celular Tumoral , Adhesión Celular/efectos de los fármacos , Melanoma Experimental/patología , Melanoma Experimental/tratamiento farmacológico , Paxillin/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Fitoquímicos/farmacología , Fitoquímicos/química , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo
14.
Commun Biol ; 7(1): 594, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760406

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is a chronic disease caused by hepatic steatosis. Adenosine deaminases acting on RNA (ADARs) catalyze adenosine to inosine RNA editing. However, the functional role of ADAR2 in NAFLD is unclear. ADAR2+/+/GluR-BR/R mice (wild type, WT) and ADAR2-/-/GluR-BR/R mice (ADAR2 KO) mice are fed with standard chow or high-fat diet (HFD) for 12 weeks. ADAR2 KO mice exhibit protection against HFD-induced glucose intolerance, insulin resistance, and dyslipidemia. Moreover, ADAR2 KO mice display reduced liver lipid droplets in concert with decreased hepatic TG content, improved hepatic insulin signaling, better pyruvate tolerance, and increased glycogen synthesis. Mechanistically, ADAR2 KO effectively mitigates excessive lipid production via AMPK/Sirt1 pathway. ADAR2 KO inhibits hepatic gluconeogenesis via the AMPK/CREB pathway and promotes glycogen synthesis by activating the AMPK/GSK3ß pathway. These results provide evidence that ADAR2 KO protects against NAFLD progression through the activation of AMPK signaling pathways.


Asunto(s)
Adenosina Desaminasa , Dieta Alta en Grasa , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico , Proteínas de Unión al ARN , Transducción de Señal , Animales , Ratones , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/genética , Adenosina Desaminasa/deficiencia , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/etiología , Obesidad/metabolismo , Obesidad/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética
15.
J Cachexia Sarcopenia Muscle ; 15(3): 949-962, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38533529

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide. Sarcopenia is a syndrome characterized by progressive and generalized loss of skeletal muscle mass and strength, which is commonly associated with NAFLD. Adenosine-to-inosine editing, catalysed by adenosine deaminase acting on RNA (ADAR), is an important post-transcriptional modification of genome-encoded RNA transcripts. Three ADAR gene family members, including ADAR1, ADAR2 and ADAR3, have been identified. However, the functional role of ADAR2 in obesity-associated NAFLD and sarcopenia remains unclear. METHODS: ADAR2+/+/GluR-BR/R mice (wild type [WT]) and ADAR2-/-/GluR-BR/R mice (ADAR2 knockout [KO]) were subjected to feeding with standard chow or high-fat diet (HFD) for 20 weeks at the age of 5 weeks. The metabolic parameters, hepatic lipid droplet, grip strength test, rotarod test, muscle weight, fibre cross-sectional area (CSA), fibre types and protein associated with protein degradation were examined. Systemic and local tissues serum amyloid A1 (SAA1) were measured. The effects of SAA1 on C2C12 myotube atrophy were investigated. RESULTS: ADAR2 KO mice fed with HFD exhibited lower body weight (-7.7%, P < 0.05), lower liver tissue weight (-20%, P < 0.05), reduced liver lipid droplets in concert with a decrease in hepatic triglyceride content (-24%, P < 0.001) and liver injury (P < 0.01). ADAR2 KO mice displayed protection against HFD-induced glucose intolerance, insulin resistance and dyslipidaemia. Skeletal muscle mass (P < 0.01), muscle strength (P < 0.05), muscle endurance (P < 0.001) and fibre size (CSA; P < 0.0001) were improved in ADAR2 KO mice fed with HFD compared with WT mice fed with HFD. Muscle atrophy-associated transcripts, such as forkhead box protein O1, muscle atrophy F-box/atrogin-1 and muscle RING finger 1/tripartite motif-containing 63, were decreased in ADAR2 KO mice fed with HFD compared with WT mice fed with HFD. ADAR2 deficiency attenuates HFD-induced local liver and skeletal muscle tissue inflammation. ADAR2 deficiency abolished HFD-induced systemic (P < 0.01), hepatic (P < 0.0001) and muscular (P < 0.001) SAA1 levels. C2C12 myotubes treated with recombinant SAA1 displayed a decrease in myotube length (-37%, P < 0.001), diameter (-20%, P < 0.01), number (-39%, P < 0.001) and fusion index (-46%, P < 0.01). Myogenic markers (myosin heavy chain and myogenin) were decreased in SAA1-treated myoblast C2C12 cells. CONCLUSIONS: These results provide novel evidence that ADAR2 deficiency may be important in obesity-associated sarcopenia and NAFLD. Increased SAA1 might be involved as a regulatory factor in developing sarcopenia in NAFLD.


Asunto(s)
Adenosina Desaminasa , Ratones Noqueados , Atrofia Muscular , Enfermedad del Hígado Graso no Alcohólico , Proteínas de Unión al ARN , Proteína Amiloide A Sérica , Animales , Adenosina Desaminasa/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ratones , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Atrofia Muscular/metabolismo , Proteína Amiloide A Sérica/metabolismo , Modelos Animales de Enfermedad , Masculino , Dieta Alta en Grasa , Músculo Esquelético/patología , Músculo Esquelético/metabolismo
16.
J Pathol ; 228(2): 158-69, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22247069

RESUMEN

Hepatoma-derived growth factor (HDGF) participates in tumourigenesis but its role in breast cancer is unclear. We set out to elucidate the expression profile and function of HDGF during breast carcinogenesis. Immunoblot and immunohistochemical studies revealed elevated HDGF expression in human breast cancer cell lines and tissues. Nuclear HDGF labelling index was positively correlated with tumour grade, stage and proliferation index, but negatively correlated with survival rate in breast cancer patients. HDGF over-expression was associated with lymph node metastasis and represented an independent prognostic factor for tumour recurrence. Gene transfer studies were performed to elucidate the influence of cellular HDGF level on the malignant behaviour and epithelial-mesenchymal transition (EMT) of breast cancer cells. Adenovirus-mediated HDGF over-expression stimulated the invasiveness and colony formation of MCF-7 cells. Moreover, HDGF over-expression promoted breast cancer cell EMT by E-cadherin down-regulation and vimentin up-regulation. Conversely, HDGF knockdown by RNA interference in MDA-MB-231 cells attenuated the malignant behaviour and elicited EMT reversal by enhancing E-cadherin expression while depleting vimentin expression. Because HDGF is a secreted protein, we evaluated the cellular function of recombinant HDGF and found that exogenously supplied HDGF enhanced the invasiveness of breast cancer cells by down-regulating E-cadherin and up-regulating vimentin at transcriptional and translational levels. In contrast, blockade of HDGF secretion with an HDGF antibody inhibited the malignant behaviours and EMT. Finally, exogenous HDGF partially reversed benzyl isothiocyanate (BITC)-induced EMT suppression. HDGF over-expression may exert a prognostic role for tumour metastasis and recurrence in breast cancer by modulating EMT. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias de la Mama/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/secundario , Adulto , Anticuerpos Bloqueadores/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Técnicas de Transferencia de Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/inmunología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Antígeno Ki-67/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática , Mastectomía , Persona de Mediana Edad , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Recurrencia Local de Neoplasia , Pronóstico , Proteínas Recombinantes
17.
Biochem Biophys Res Commun ; 425(2): 169-76, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22819846

RESUMEN

Hepatoma-derived growth factor (HDGF) stimulates the migration, invasion and metastasis in several types of cancer cells. However, the mechanism underlying HDGF-stimulated migration remains unclear. In this study, we investigated the influence of HDGF on cytoskeleton remodeling and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway in non-transformed NIH/3T3 cells. Exogenous HDGF promoted the migration and the formation of dorsal ruffles and podosome rosettes. Besides, HDGF supply increased the PI3K expression and Akt phosphorylation in dose- and time-dependent manners. Application of LY294002, a PI3K inhibitor, attenuated the HDGF-induced migration, dorsal ruffles and podosome rosettes formation. Consistently, the HDGF-overexpressing NIH/3T3 transfectants exhibited significantly increased motility and elevated PI3K/Akt activities, which were repressed by LY294002 or adenovirus-mediated overexpression of endogenous PI3K antagonist, PTEN. In summary, HDGF elicits the activation of PI3K/Akt signaling cascade, thereby promoting cytoskeleton remodeling to stimulate cellular migration.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Fosfatidilinositol 3-Quinasa/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Formación de Roseta , Animales , Movimiento Celular , Citoesqueleto/genética , Citoesqueleto/metabolismo , Activación Enzimática , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Células 3T3 NIH , Transfección
18.
J Neurooncol ; 107(1): 101-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22037800

RESUMEN

Hepatoma-derived growth factor (HDGF) is a neurotrophic factor found in mouse spinal cord and hippocampal neurons. In various malignant tumors, the role of HDGF in tumor progression and its use as a diagnostic biomarker or therapeutic target have been extensively explored. However, the prognostic function and mitogenic role of HDGF in gliomagenesis are yet to be verified. In this study, we found a significant incidence of HDGF prevalence between the different pathological types and stages of glioma in 105 patients. We also found a prognostic significance in 41 glioblastoma multiforme (GBM) patients, with prevalence of nuclear HDGF predicting short survival of patients with GBM after surgery. To delineate the mitogenic role of HDGF in gliomagenesis, an adenoviral-expressed HDGF small interfering RNA (Ad-HDGF siRNA) was used to knock down expression of nuclear HDGF. After knocking down nuclear HDGF expression in human GBM cells, cell growth and cell invasion and induction on apoptosis by caspase-3 activation were significantly inhibited. We conclude that HDGF is a mitogenic growth factor in glioma progression and can be a useful prognostic marker for GBM and therapeutic target for clinical management of glioma in the future.


Asunto(s)
Glioma/metabolismo , Glioma/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Adenoviridae/genética , Apoptosis , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Caspasa 3/metabolismo , Adhesión Celular , Ciclo Celular , Movimiento Celular , Proliferación Celular , Colágeno/metabolismo , Progresión de la Enfermedad , Combinación de Medicamentos , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Glioma/mortalidad , Humanos , Técnicas para Inmunoenzimas , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Laminina/metabolismo , Masculino , Persona de Mediana Edad , Proteoglicanos/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Tasa de Supervivencia , Células Tumorales Cultivadas
19.
Biomedicines ; 10(2)2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35203529

RESUMEN

Due to its aggressiveness and high mortality rate, oral cancer still represents a tough challenge for current cancer therapeutics. Similar to other carcinomas, cancerous invasion and metastasis are the most important prognostic factors and the main obstacles to therapy for human oral squamous cell carcinoma (OSCC). Fortunately, with the rise of the nanotechnical era and innovative nanomaterial fabrication, nanomaterials are widely used in biomedicine, cancer therapeutics, and chemoprevention. Recently, phytochemical substances have attracted increasing interest as adjuvants to conventional cancer therapy. The ginger phenolic compound zingerone, a multitarget pharmacological and bioactive phytochemical, possesses potent anti-inflammatory, antioxidant, and anticancer activities. In our previous study, we generated phytochemically derived zingerone nanoparticles (NPs), and documented their superior antitumorigenic effect on human hepatoma cells. In the present study, we further investigated the effects of zingerone NPs on inhibiting the invasiveness and metastasis of human OSCC cell lines. Zingerone NPs elicited significant cytotoxicity in three OSCC cell lines compared to zingerone. Moreover, the lower dose of zingerone NPs (25 µM) markedly inhibited colony formation and colony survival by at least five-fold compared to zingerone treatment. Additionally, zingerone NPs significantly attenuated cell motility and invasiveness. In terms of the signaling mechanism, we determined that the zingerone NP-mediated downregulation of Akt signaling played an important role in the inhibition of cell viability and cell motility. Zingerone NPs inhibited matrix metalloproteinase (MMP) activity, which was highly correlated with the attenuation of cell migration and cell invasion. By further detecting the roles of zingerone NPs in epithelial-mesenchymal transition (EMT), we observed that zingerone NPs substantially altered the levels of EMT-related markers by decreasing the levels of the mesenchymal markers, N-cadherin and vimentin, rather than the epithelial proteins, ZO-1 and E-cadherin, compared with zingerone. In conclusion, as novel and efficient phytochemically derived nanoparticles, zingerone NPs may serve as a potent adjuvant to protect against cell invasion and metastasis, which will provide a beneficial strategy for future applications in chemoprevention and conventional therapeutics in OSCC treatment.

20.
Genes (Basel) ; 13(4)2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35456435

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most common RCC subtype with a high mortality. It has been reported that delta-like 1 homologue (DLK1) participates in the tumor microenvironmental remodeling of ccRCC, but the relationship between delta-like 2 homologue (DLK2, a DLK1 homologue) and ccRCC is still unclear. Thus, this study aims to investigate the role of DLK2 in the biological function and disease prognosis of ccRCC using bioinformatics analysis. The TNMplot database showed that DLK2 was upregulated in ccRCC tissues. From the UALCAN analysis, the overexpression of DLK2 was associated with advanced stage and high grade in ccRCC. Moreover, the Kaplan-Meier plotter (KM Plotter) database showed that DLK2 upregulation was associated with poor survival outcome in ccRCC. By the LinkedOmics analysis, DLK2 signaling may participated in the modulation of ccRCC extracellular matrix (ECM), cell metabolism, ribosome biogenesis, TGF-ß signaling and Notch pathway. Besides, Tumor Immune Estimation Resource (TIMER) analysis showed that the macrophage and CD8+ T cell infiltrations were associated with good prognosis in ccRCC patients. Finally, DLK2 overexpression was associated with the reduced macrophage recruitments and the M1-M2 polarization of macrophage in ccRCC tissues. Together, DLK2 may acts as a novel biomarker, even therapeutic target in ccRCC. However, this study lacks experimental validation, and further studies are required to support this viewpoint.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/metabolismo , Biología Computacional , Femenino , Humanos , Neoplasias Renales/metabolismo , Masculino , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA