Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioeng Transl Med ; 7(2): e10286, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35600659

RESUMEN

Abundance of stromal cells and extracellular matrix (ECM) is observed in breast cancer, acting as a barrier for drug penetration and presenting a key issue for developing efficient therapeutics. In this study, we aimed to develop a three-dimensional (3D) multicellular tumor model comprising cancer and stromal cells that could effectively mimic the drug resistance properties of breast cancer. Three different types of spheroid models were designed by co-culturing breast cancer cells (MDA-MB-231) with three different types of stromal cells: human adipose-derived stromal cells (hASCs), human bone marrow stromal cells, or human dermal fibroblasts. Compared with other models, in the hASC co-culture model, tissue inhibitor of metalloproteinases-1 (TIMP-1) was highly expressed and the activity of matrix metalloproteinases was decreased, resulting in a higher ECM deposition on the spheroid surfaces. This spheroid model showed less drug penetration and treatment efficacy than the other models. TIMP-1 silencing in hASCs reduced ECM protein expression and increased drug penetration and vulnerability. A quantitative structure-activity relationship study using multiple linear regression drew linear relationships between the chemical properties of drugs and experimentally determined permeability values. Drugs that did not match the drug-likeness rules exhibited lower permeability in the 3D tumor model. Taken together, our findings indicate that this 3D multicellular tumor model may be used as a reliable platform for efficiently screening therapeutics agents for solid tumors.

2.
NPJ Regen Med ; 6(1): 48, 2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408157

RESUMEN

Peripheral artery disease is a progressive, devastating disease that leads to critical limb ischemia (CLI). Therapeutic angiogenesis using stem cell therapy has emerged as a promising approach for its treatment; however, adapting cell-based therapy has been limited by poor cell survival and low treatment efficiency. To overcome unmet clinical needs, we developed a fibroblast growth factor 2 (FGF2)-immobilized matrix that enabled control of cell adhesion to the surface and exerted a priming effect on the cell. Human adipose-derived stem cells (hASCs) grown in this matrix formed a functionally enhanced cells spheroid (FECS-Ad) that secreted various angiogenic factors including interleukin-8 (IL-8). We demonstrated that IL-8 was upregulated by the FGF2-mediated priming effect during FECS-Ad formation. Immobilized FGF2 substrate induced stronger IL-8 expression than soluble FGF2 ligands, presumably through the FGFR1/JNK/NF-κB signaling cascade. In IL-8-silenced FECS-Ad, vascular endothelial growth factor (VEGF) expression was decreased and angiogenic potential was reduced. Intramuscular injection of FECS-Ad promoted angiogenesis and muscle regeneration in mouse ischemic tissue, while IL-8 silencing in FECS-Ad inhibited these effects. Taken together, our data demonstrate that IL-8 contributes to therapeutic angiogenesis and suggest that FECS-Ad generated using the MBP-FGF2 matrix might provide a reliable platform for developing therapeutic agents to treat CLI.

3.
Biochem Biophys Res Commun ; 517(3): 452-457, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31376938

RESUMEN

Hepatocyte growth factor (HGF) is a versatile neurotrophic factor that mediates a variety of cellular activities. In this study, we investigated the effects of intramuscularly injected recombinant AAV vectors expressing HGF in two pathologic conditions: the sciatic nerve crush and the SOD1-G93A transgenic mouse models. AAV serotype 6 (rAAV6) was chosen based on its expression levels in, and capability of moving to, the spinal cord from the injected muscle area. In the nerve crush model, rAAV6-HGF was shown to reduce the degree of mechanical allodynia, increase the cross-sectional area of muscle fibers, promote regrowth of peripheral axons, and improve motor functions. In the SOD1-G93A TG mouse model, rAAV6-HGF increased the mass of the tibialis anterior and gastrocnemius, alleviated disease symptoms, and prolonged survival. Improvements in integrity and functions of muscle in these models seemed to have come from the ability of HGF produced from rAAV6-HGF to regulate the expression of various atrogenes through the control of the FOXO signaling pathway. Our findings suggested that intramuscular injection of rAAV6-HGF might be used to relieve various symptoms associated with muscle atrophy and/or nerve damages observed in a majority of neuromuscular diseases.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Factor de Crecimiento de Hepatocito/genética , Músculo Esquelético/metabolismo , Unión Neuromuscular/metabolismo , Superóxido Dismutasa-1/genética , Animales , Dependovirus/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Fuerza de la Mano/fisiología , Factor de Crecimiento de Hepatocito/metabolismo , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Hiperalgesia/prevención & control , Inyecciones Intramusculares , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Músculo Esquelético/inervación , Músculo Esquelético/patología , Mutación , Compresión Nerviosa/métodos , Unión Neuromuscular/patología , Prueba de Desempeño de Rotación con Aceleración Constante , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Nervio Ciático/patología , Superóxido Dismutasa-1/deficiencia
4.
Front Physiol ; 10: 914, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31404148

RESUMEN

Hepatocyte growth factor (HGF) is well known for its role in the migration of embryonic muscle progenitors and the activation of adult muscle stem cells, yet its functions during the adult muscle regeneration process remain to be elucidated. In this study, we showed that HGF/c-met signaling was activated during muscle regeneration, and that among various infiltrated cells, the macrophage is the major cell type affected by HGF. Pharmacological inhibition of the c-met receptor by PHA-665752 increased the expression levels of pro-inflammatory (M1) macrophage markers such as IL-1ß and iNOS while lowering those of pro-regenerative (M2) macrophage markers like IL-10 and TGF-ß, resulting in compromised muscle repair. In Raw 264.7 cells, HGF decreased the RNA level of LPS-induced TNF-α, IL-1ß, and iNOS while enhancing that of IL-10. HGF was also shown to increase the phosphorylation of AMPKα through CaMKKß, thereby overcoming the effects of the LPS-induced deactivation of AMPKα. Transfection with specific siRNA to AMPKα diminished the effects of HGF on the LPS-induced gene expressions of M1 and M2 markers. Exogenous delivery of HGF through intramuscular injection of the HGF-expressing plasmid vector promoted the transition to M2 macrophage and facilitated muscle regeneration. Taken together, our findings suggested that HGF/c-met might play an important role in the transition of the macrophage during muscle repair, indicating the potential use of HGF as a basis for developing therapeutics for muscle degenerative diseases.

5.
Mol Ther Nucleic Acids ; 12: 568-577, 2018 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-30195792

RESUMEN

Hepatocyte growth factor (HGF) has been well characterized for its roles in the migration of muscle progenitors during embryogenesis and the differentiation of muscle stem cells, but its function in adult neurogenic muscle atrophic conditions is poorly understood. Here we investigated whether HGF/c-met signaling has any effects on muscle-atrophic conditions. It was found that HGF expression was upregulated in skeletal muscle tissue following surgical denervation and in hSOD1-G93A transgenic mice showing severe muscle loss. Pharmacological inhibition of the c-met receptor decreased the expression level of pri-miR-206, enhanced that of HDAC4 and atrogenes, and resulted in increased muscle atrophy. In C2C12 cells, HGF inhibited phosphorylation of Smad3 and relieved TGF-ß-mediated suppression of miR-206 expression via JNK. When extra HGF was exogenously provided through intramuscular injection of plasmid DNA expressing HGF, the extent of muscle atrophy was reduced, and the levels of all affected biochemical markers were changed accordingly, including those of primary and mature miR-206, HDAC4, and various atrogenes. Taken together, our finding suggested that HGF might play an important role in regard to neurogenic muscle atrophy and that HGF might be used as a platform to develop therapeutic agents for neuromuscular disorders.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...