Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Gene Med ; 21(1): e3067, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30592790

RESUMEN

BACKGROUND: Diabetes mellitus is caused by a partial or complete lack of insulin production in the body. We have previously shown that a single injection of an adeno-associated virus serotype 8 (AAV8) vector carrying a modified and codon optimized human insulin gene induced hepatic production of insulin and corrected streptozotocin (STZ)-induced diabetes in mice for more than 1 year. Insulin production was constitutive, analogous to long-acting insulin therapy. METHODS: We have developed a single AAV8 vector with a Tet-Off regulatable system as a safety mechanism to turn off insulin secretion should hypoglycaemia develop in vector-treated diabetic mice. We first transfected HepG2 cells or freshly isolated rat hepatocytes in vitro with the Tet-Off system (pAAV-Tetoffbidir -Alb-luc) regulating a luciferase reporter gene. We subsequently incorporated a furin-cleavable codon-optimised human proinsulin cDNA into pAAV-Tetoffbidir backbone to form the doxycycline inducible pAAV-Tetoffbidir -Alb-hINSco. RESULTS: Using STZ-induced diabetic mice, we were able to switch off insulin secretion repeatedly with doxycycline administration, and showed full restoration of insulin secretion on withdrawing doxycycline. CONCLUSIONS: The present study provides proof of concept that, under circumstances when inappropriate basal insulin secretion is a safety concern, insulin secretion from AAV8 gene therapy can be turned off reversibly with doxycycline.


Asunto(s)
Dependovirus/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/terapia , Terapia Genética , Vectores Genéticos/genética , Insulina/genética , Hígado/metabolismo , Animales , Diabetes Mellitus Experimental/diagnóstico , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Genes Reporteros , Terapia Genética/métodos , Hepatocitos/metabolismo , Humanos , Ratones , Imagen Molecular , Transfección
2.
Inorg Chem ; 58(13): 8293-8299, 2019 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-31184865

RESUMEN

Triphenylphosphonium (TPP+) species comprising multiple charges, i.e., bis-TPP+, are predicted to be superior mitochondrial-targeting vectors and are expected to have mitochondrial accumulations 1000-fold greater than TPP+, the current "gold standard". However, bis-TPP+ vectors linked by short hydrocarbon chains ( n < 5) are unable to be taken up by the mitochondria, thus hindering their development as mitochondrial delivery vectors. Through the incorporation of methylated TPP+ moieties (T*PP+), we successfully enabled the accumulation of bis-TPP+ with a short linker chain in isolated mitochondria, as measured by high performance liquid chromatography. These experimental results are further supported by molecular dynamics and ab initio calculations, revealing the strong correlations between mitochondria uptake and molecular volume, surface area, and chemical hardness. Most notably, the molecular volume has been shown to be a strong predictor of accumulation for both mono- and bis-TPP+ salts. Our study underscores the potential of T*PP+ moieties as alternative mitochondrial vectors to overcome low permeation into the mitochondria.


Asunto(s)
Mitocondrias/metabolismo , Compuestos Onio/metabolismo , Compuestos Organofosforados/metabolismo , Transporte Biológico , Teoría Funcional de la Densidad , Células HeLa , Humanos , Modelos Químicos , Simulación de Dinámica Molecular , Estructura Molecular , Compuestos Onio/síntesis química , Compuestos Onio/química , Compuestos Organofosforados/síntesis química , Compuestos Organofosforados/química , Relación Estructura-Actividad Cuantitativa , Termodinámica
3.
Proteomics ; 18(16): e1700474, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29963755

RESUMEN

Epidemiological data indicate that human cancer risk is significantly reduced by the consumption of soy-based foods containing the "phytoestrogen" genistein, which can signal via host cell estrogen receptors. While additional chemoprotective effects of genistein induced by epigenetic factors have also been reported, the key molecules and mechanisms involved are poorly defined. We therefore investigated genistein effects on chromatin-bound proteins in the estrogen receptor-deficient cell line MDA-MB-231 which is insensitive to phytoestrogen signaling. After exposure to low-dose genistein for >1 month, MDA-MB-231 cells exhibited stable epigenetic alterations that are analyzed via partial MNase digestion and TMT-based quantitative proteomics. 3177 chromatin-bound proteins are identified with high confidence, including 882 molecules that displayed altered binding topology after cell conditioning with genistein. Prolonged phytochemical exposure conferred heritable changes in the binding topology of key epigenetic regulators including ATRX, SUV39H1/H2, and HP1BP3 that are preserved in untreated progeny, resulting in sustained downregulation of proliferation genes and reduced cell growth. These data indicate that soy derivative genistein exerts complex estrogen receptor-independent effects on the epigenome likely to influence tumorigenesis by restricting cell growth.


Asunto(s)
Anticarcinógenos/farmacología , Neoplasias de la Mama/patología , Proliferación Celular , Cromatina , Genisteína/farmacología , Fitoquímicos/farmacología , Proteoma/análisis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Ciclo Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Transducción de Señal , Glycine max/química , Células Tumorales Cultivadas
4.
J Cell Mol Med ; 22(7): 3698-3702, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29682884

RESUMEN

Genetically modified FVIII-expressing autologous bone marrow-derived mesenchymal stromal cells (BMSCs) could cure haemophilia A. However, culture-expanded BMSCs engraft poorly in extramedullary sites. Here, we compared the intramedullary cavity, skeletal muscle, subcutaneous tissue and systemic circulation as tissue microenvironments that could support durable engraftment of FVIII-secreting BMSC in vivo. A zinc finger nuclease integrated human FVIII transgene into PPP1R12C (intron 1) of culture-expanded primary canine BMSCs. FVIII-secretory capacity of implanted BMSCs in each dog was expressed as an individualized therapy index (number of viable BMSCs implanted × FVIII activity secreted/million BMSCs/24 hours). Plasma samples before and after implantation were assayed for transgenic FVIII protein using an anti-human FVIII antibody having negligible cross-reactivity with canine FVIII. Plasma transgenic FVIII persisted for at least 48 weeks after implantation in the intramedullary cavity. Transgenic FVIII protein levels were low after intramuscular implantation and undetectable after both intravenous infusion and subcutaneous implantation. All plasma samples were negative for anti-human FVIII antibodies. Plasma concentrations and durability of transgenic FVIII secretion showed no correlation with the therapy index. Thus, the implantation site microenvironment is crucial. The intramedullary microenvironment, but not extramedullary tissues, supported durable engraftment of genetically modified autologous FVIII-secreting BMSCs.


Asunto(s)
Factor VIII/genética , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Animales , Animales Modificados Genéticamente , Células de la Médula Ósea , Perros , Factor VIII/metabolismo , Masculino , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Proteínas Recombinantes/sangre , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Nucleasas con Dedos de Zinc/genética , Nucleasas con Dedos de Zinc/metabolismo
5.
Bioconjug Chem ; 28(2): 590-599, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28049291

RESUMEN

In this study, unique methyl-functionalized derivatives (T*PP+) of the drug carrier triphenylphosphonium (TPP+) that exhibit significant enhancement of the accumulation of both the cation and its conjugated cargo in cell mitochondria are designed. We show that the presence of methyl group(s) at key positions within the phenyl ring results in an increase in the hydrophobicity and solvent accessible surface area of T*PP+. In particular, when the para position of the phenyl ring in T*PP+ is functionalized with a methyl group, the cation is most exposed to the surrounding environment, leading to a large decrease in water entropy and an increase in the level of van der Waals interaction with and partition into a nonpolar solvent. Therefore, stronger binding between the hydrophobic T*PP+ and mitochondrial membrane occurs. This is exemplified in a (hexachloro-fluorescein)-TPP+ conjugate system, where an ∼12 times increase in the rate of mitochondrial uptake and a 2 times increase in photodynamic therapy (PDT) efficacy against HeLa and FU97 cancer cells are achieved when TPP+ is replaced with T*PP+. Importantly, nearly all the FU97 cells treated with the (hexachloro-fluorescein)-T*PP+ conjugate are killed as compared to only half the population of cells in the case of the (hexachloro-fluorescein)-TPP+ conjugate at a similar PDT light dosage. This study thus forms a platform for the healthcare community to explore alternative TPP+ derivatives that can act as optimal drug transporters for enhanced mitochondrially targeted therapies.


Asunto(s)
Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacología , Mitocondrias/metabolismo , Compuestos Organofosforados/metabolismo , Compuestos Organofosforados/farmacología , Fármacos Fotosensibilizantes/metabolismo , Fármacos Fotosensibilizantes/farmacología , Línea Celular , Portadores de Fármacos/química , Fluoresceínas/química , Fluoresceínas/metabolismo , Fluoresceínas/farmacología , Halogenación , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Metilación , Microscopía Confocal , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Compuestos Organofosforados/química , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Termodinámica
6.
Mol Ther ; 24(3): 607-19, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26689265

RESUMEN

Costly coagulation factor VIII (FVIII) replacement therapy is a barrier to optimal clinical management of hemophilia A. Therapy using FVIII-secreting autologous primary cells is potentially efficacious and more affordable. Zinc finger nucleases (ZFN) mediate transgene integration into the AAVS1 locus but comprehensive evaluation of off-target genome effects is currently lacking. In light of serious adverse effects in clinical trials which employed genome-integrating viral vectors, this study evaluated potential genotoxicity of ZFN-mediated transgenesis using different techniques. We employed deep sequencing of predicted off-target sites, copy number analysis, whole-genome sequencing, and RNA-seq in primary human umbilical cord-lining epithelial cells (CLECs) with AAVS1 ZFN-mediated FVIII transgene integration. We combined molecular features to enhance the accuracy and activity of ZFN-mediated transgenesis. Our data showed a low frequency of ZFN-associated indels, no detectable off-target transgene integrations or chromosomal rearrangements. ZFN-modified CLECs had very few dysregulated transcripts and no evidence of activated oncogenic pathways. We also showed AAVS1 ZFN activity and durable FVIII transgene secretion in primary human dermal fibroblasts, bone marrow- and adipose tissue-derived stromal cells. Our study suggests that, with close attention to the molecular design of genome-modifying constructs, AAVS1 ZFN-mediated FVIII integration in several primary human cell types may be safe and efficacious.


Asunto(s)
Endonucleasas/metabolismo , Factor VIII/genética , Estudio de Asociación del Genoma Completo , Mutagénesis Insercional , Dedos de Zinc , Sitios de Unión , Factor VIII/metabolismo , Expresión Génica , Marcación de Gen , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células K562 , Unión Proteica , Transgenes
7.
J Proteome Res ; 15(12): 4258-4264, 2016 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-27790916

RESUMEN

Hypoxia is thought to promote tumor radio-resistance via effects on gene expression in cancer cells that modulate their metabolism, proliferation, and DNA repair pathways to enhance survival. Here we demonstrate for the first time that under hypoxic condition A431 epithelial carcinoma cells exhibit increased viability when exposed to low-dose γ-irradiation, indicating that radiotherapy can promote tumor cell survival when oxygen supply is limited. When assessed using iTRAQ quantitative proteomics and Western blotting, irradiated tumor cells were observed to significantly up-regulate the expression of calcium-binding proteins CALM1, CALU, and RCN1, suggesting important roles for these mediators in promoting tumor cell survival during hypoxia. Accordingly, shRNA-knockdown of CALM1, CALU, and RCN1 expression reduced hypoxic tumor cell resistance to low-dose radiation and increased apoptosis. These data indicate that γ-irradiation of hypoxic tumor cells induces up-regulation of calcium-binding proteins that promote cancer cell survival and may limit the efficacy of radiotherapy in the clinic.


Asunto(s)
Proteínas de Unión al Calcio/efectos de la radiación , Rayos gamma/efectos adversos , Hipoxia , Neoplasias/radioterapia , Carga Tumoral/efectos de la radiación , Western Blotting , Proteínas de Unión al Calcio/genética , Calmodulina/genética , Calmodulina/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Células Epiteliales/patología , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Proteómica/métodos , Regulación hacia Arriba/efectos de la radiación
8.
Front Oncol ; 14: 1272432, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38939336

RESUMEN

Introduction: Field cancerization is suggested to arise from imbalanced differentiation in individual basal progenitor cells leading to clonal expansion of mutant cells that eventually replace the epithelium, although without evidence. Methods: We performed deep sequencing analyses to characterize the genomic and transcriptomic landscapes of field change in two patients with synchronous aerodigestive tract tumors. Results: Our data support the emergence of numerous genetic alterations in cancer-associated genes but refutes the hypothesis that founder mutation(s) underpin this phenomenon. Mutational signature analysis identified defective homologous recombination as a common underlying mutational process unique to synchronous tumors. Discussion: Our analyses suggest a common etiologic factor defined by mutational signatures and/or transcriptomic convergence, which could provide a therapeutic opportunity.

9.
J Proteome Res ; 11(6): 3405-13, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22533479

RESUMEN

Gastric cancer remains highly fatal due to a dearth of diagnostic biomarkers for early stage disease and molecular targets for therapy. Plasma membrane proteins, including cluster of differentiation (CD) proteins and receptor tyrosine kinases (RTKs), are a rich reservoir of biomarkers. Recognizing that interrogating plasma membrane proteins individually overlooks extensive interactions among them, we have systematically investigated the membrane proteomes and transcriptomes of six gastric cancer cell lines. Our data revealed aberrantly high expression of proteins whose functions accurately reflect the clinical phenotype of gastric cancer, and prioritized critical RTKs and CD proteins in gastric cancer. Expression of selected surface proteins was confirmed by flow cytometry and immunostaining of clinical gastric cancer tissues. Close to 90% of the gastric cancer tissues in a cohort showed up-regulation of at least one of four proteins, that is, MET, EPHA2, FGFR2, and CD104/ITGB4. All intestinal type gastric cancer tumors in this cohort overexpressed at least one of a panel of three proteins, MET, FGFR2, and EPHA2. This study reports the first quantitative global landscape of the surface proteome of gastric cancer cells and provides a shortlist of gastric cancer biomarkers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias Gástricas/metabolismo , Línea Celular Tumoral , Humanos , Proteoma/metabolismo , Espectrometría de Masas en Tándem , Análisis de Matrices Tisulares
10.
Cell Mol Life Sci ; 68(11): 1983-2002, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20953656

RESUMEN

We integrated LC-MS/MS-based and protein antibody array-based proteomics with genomics approaches to investigate the phosphoproteome and transcriptome of gastric cancer cell lines and endoscopic gastric biopsies from normal subjects and patients with benign gastritis or gastric cancer. More than 3,000 non-redundant phosphorylation sites in over 1,200 proteins were identified in gastric cancer cells. We correlated phosphoproteome data with transcriptome data sets and reported the expression of 41 protein kinases, 5 phosphatases and 65 phosphorylated mitochondrial proteins in gastric cancer cells. Transcriptional expression levels of 190 phosphorylated proteins were >2-fold higher in gastric cancer cells compared to normal stomach tissue. Pathway analysis demonstrated over-presentation of DNA damage response pathway and underscored critical roles of phosphorylated p53 in gastric cancer. This is the first study to comprehensively report the gastric cancer phosphoproteome. Integrative analysis of the phosphoproteome and transcriptome provided an expansive view of molecular signaling pathways in gastric cancer.


Asunto(s)
Perfilación de la Expresión Génica , Proteoma/genética , Neoplasias Gástricas/fisiopatología , Línea Celular Tumoral , Cromatografía de Gases y Espectrometría de Masas , Humanos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA