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

Banco 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
11.
Mol Cell Proteomics ; 9(12): 2629-41, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20713453

RESUMEN

Cancer cells with MET overexpression are paradoxically more sensitive to MET inhibition than cells with baseline MET expression. The underlying molecular mechanisms are incompletely understood. Here, we have traced early responses of SNU5, a MET-overexpressing gastric cancer cell line, exposed to sublethal concentration of PHA-665752, a selective MET inhibitor, using iTRAQ-based quantitative proteomics. More than 1900 proteins were quantified, of which >800 proteins were quantified with at least five peptides. Proteins whose expression was perturbed by PHA-665752 included oxidoreductases, transfer/carrier proteins, and signaling proteins. Strikingly, 38% of proteins whose expression was confidently assessed to be perturbed by MET inhibition were mitochondrial proteins. Upon MET inhibition by a sublethal concentration of PHA-665752, mitochondrial membrane potential increased and mitochondrial permeability transition pore was inhibited concomitant with widespread changes in mitochondrial protein expression. We also showed the presence of highly activated MET in mitochondria, and striking suppression of MET activation by 50 nm PHA-665752. Taken together, our data indicate that mitochondria are a direct target of MET kinase inhibition, in addition to plasma membrane MET. Effects on activated MET in the mitochondria of cancer cells that are sensitive to MET inhibition might constitute a novel and critical noncanonical mechanism for the efficacy of MET-targeted therapeutics.


Asunto(s)
Mitocondrias/metabolismo , Proteómica , Proteínas Proto-Oncogénicas c-met/metabolismo , Neoplasias Gástricas/metabolismo , Western Blotting , Línea Celular Tumoral , Cromatografía Liquida , Perfilación de la Expresión Génica , Humanos , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Espectrometría de Masas en Tándem
12.
Mol Oncol ; 16(8): 1611-1624, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35194950

RESUMEN

Thyroid nodules occur in about 60% of the population. A major challenge in thyroid nodule diagnosis is to distinguish between follicular adenoma (FA) and carcinoma (FTC). Here, we present a comprehensive thyroid spectral library covering five types of thyroid tissues. This library includes 121 960 peptides and 9941 protein groups. This spectral library can be used to quantify up to 7863 proteins from thyroid tissues, and can also be used to develop parallel reaction monitoring (PRM) assays for targeted protein quantification. Next, to stratify follicular thyroid tumours, we compared the proteomes of 24 FA and 22 FTC samples, and identified 204 differentially expressed proteins (DEPs). Our data suggest altered ferroptosis pathways in malignant follicular carcinoma. In all, 31 selected proteins effectively distinguished follicular tumours. Of those DEPs, nine proteins were further verified by PRM in an independent cohort of 18 FA and 19 FTC. Together, we present a comprehensive spectral library for DIA and targeted proteomics analysis of thyroid tissue specimens, and identified nine proteins that could potentially distinguish FA and FTC.


Asunto(s)
Adenocarcinoma Folicular , Adenoma , Carcinoma , Neoplasias de la Tiroides , Adenocarcinoma Folicular/diagnóstico , Adenocarcinoma Folicular/metabolismo , Adenocarcinoma Folicular/patología , Adenoma/diagnóstico , Humanos , Proteómica , Neoplasias de la Tiroides/metabolismo
13.
Cell Rep Med ; 3(2): 100526, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35243423

RESUMEN

Peritoneal carcinomatosis (PC) present a ubiquitous clinical conundrum in all intra-abdominal malignancies. Via functional and transcriptomic experiments of ascites-treated PC cells, we identify STAT3 as a key signaling pathway. Integrative analysis of publicly available databases and correlation with clinical cohorts (n = 7,359) reveal putative clinically significant activating ligands of STAT3 signaling. We further validate a 3-biomarker prognostic panel in ascites independent of clinical covariates in a prospective study (n = 149). Via single-cell sequencing experiments, we uncover that PAI-1, a key component of the prognostic biomarker panel, is largely secreted by fibroblasts and mesothelial cells. Molecular stratification of ascites using PAI-1 levels and STAT3 activation in ascites-treated cells highlight a therapeutic opportunity based on a phenomenon of paracrine addiction. These results are recapitulated in patient-derived ascites-dependent xenografts. Here, we demonstrate therapeutic proof of concept of direct ligand inhibition of a prognostic target within an enclosed biological space.


Asunto(s)
Neoplasias Peritoneales , Animales , Ascitis , Modelos Animales de Enfermedad , Humanos , Ligandos , Ratones , Inhibidor 1 de Activador Plasminogénico/genética , Estudios Prospectivos
14.
Cell Discov ; 8(1): 85, 2022 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-36068205

RESUMEN

Determination of malignancy in thyroid nodules remains a major diagnostic challenge. Here we report the feasibility and clinical utility of developing an AI-defined protein-based biomarker panel for diagnostic classification of thyroid nodules: based initially on formalin-fixed paraffin-embedded (FFPE), and further refined for fine-needle aspiration (FNA) tissue specimens of minute amounts which pose technical challenges for other methods. We first developed a neural network model of 19 protein biomarkers based on the proteomes of 1724 FFPE thyroid tissue samples from a retrospective cohort. This classifier achieved over 91% accuracy in the discovery set for classifying malignant thyroid nodules. The classifier was externally validated by blinded analyses in a retrospective cohort of 288 nodules (89% accuracy; FFPE) and a prospective cohort of 294 FNA biopsies (85% accuracy) from twelve independent clinical centers. This study shows that integrating high-throughput proteomics and AI technology in multi-center retrospective and prospective clinical cohorts facilitates precise disease diagnosis which is otherwise difficult to achieve by other methods.

15.
J Proteome Res ; 10(10): 4535-46, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21842849

RESUMEN

Gastric fluid is a source of gastric cancer biomarkers. However, very little is known about the normal gastric fluid proteome and its biological variations. In this study, we performed a comprehensive analysis of the human gastric fluid proteome using samples obtained from individuals with benign gastric conditions. Gastric fluid proteins were prefractionated using ultracentrifuge filters (3 kDa cutoff) and analyzed by two-dimensional gel electrophoresis (2-DE) and multidimensional LC-MS/MS. Our 2-DE analysis of 170 gastric fluid samples revealed distinct protein profiles for acidic and neutral samples, highlighting pH effects on protein composition. By 2D LC-MS/MS analysis of pooled samples, we identified 284 and 347 proteins in acidic and neutral samples respectively (FDR ≤1%), of which 265 proteins (72.4%) overlapped. However, unlike neutral samples, most proteins in acidic samples were identified from peptides in the filtrate (i.e., <3 kDa). Consistent with this finding, immunoblot analysis of six potential gastric cancer biomarkers rarely detected full-length proteins in acidic samples. These findings have important implications for biomarker studies because a majority of gastric cancer patients have neutral gastric fluid compared to noncancer controls. Consequently, sample stratification, choice of proteomic approaches, and validation strategy can profoundly affect the interpretation of biomarker findings. These observations should help to refine gastric fluid biomarker studies.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Jugo Gástrico/metabolismo , Perfilación de la Expresión Génica , Proteoma/análisis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Cromatografía Líquida de Alta Presión/métodos , Electroforesis en Gel Bidimensional , Endoscopía/métodos , Femenino , Regulación de la Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Immunoblotting/métodos , Masculino , Espectrometría de Masas/métodos , Persona de Mediana Edad , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
16.
Mol Ther ; 18(7): 1346-56, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20424600

RESUMEN

Biosafety and efficacy considerations that impede clinical application of gene therapy could be addressed by nonviral ex vivo cell therapy, utilizing transgenic cells that have been comprehensively pre-evaluated for genotoxic potential and transgene expression. We evaluated the genotoxic potential of phiC31 bacteriophage integrase-mediated transgene integration in cord-lining epithelial cells (CLECs) readily cultured from the outer membrane of human umbilical cords, by sequencing and mapping integration sites, spectral karyotyping, high-resolution genome copy number, transcriptome, and transgene copy number analyses and in vivo tumorigenicity. Of 44 independent integration events, <5% were exonic and 85% of modified cells had integrated

Asunto(s)
Células Epiteliales/citología , Transgenes/genética , Cordón Umbilical/citología , Animales , Western Blotting , Células Cultivadas , Electroporación , Células Epiteliales/metabolismo , Factor VIII/genética , Factor VIII/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Ratones , Ratones SCID , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
17.
Adv Drug Deliv Rev ; 176: 113844, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34182017

RESUMEN

Biomarkers are assayed to assess biological and pathological status. Recent advances in high-throughput proteomic technology provide opportunities for developing next generation biomarkers for clinical practice aided by artificial intelligence (AI) based techniques. We summarize the advances and limitations of cancer biomarkers based on genomic and transcriptomic analysis, as well as classical antibody-based methodologies. Then we review recent progresses in mass spectrometry (MS)-based proteomics in terms of sample preparation, peptide fractionation by liquid chromatography (LC) and mass spectrometric data acquisition. We highlight applications of AI techniques in high-throughput clinical studies as compared with clinical decisions based on singular features. This review sets out our approach for discovering clinical biomarkers in studies using proteomic big data technology conjoined with computational and statistical methods.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias/patología , Proteómica/métodos , Inteligencia Artificial , Macrodatos , Cromatografía Liquida/métodos , Ensayos Analíticos de Alto Rendimiento , Humanos , Espectrometría de Masas/métodos , Neoplasias/genética
18.
Stem Cells ; 27(1): 29-39, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18845764

RESUMEN

Cancer stem cells have been shown to initiate and sustain tumor growth. In many instances, clinical material is limited, compounded by a lack of methods to preserve such cells at convenient time points. Although brain tumor-initiating cells grown in a spheroid manner have been shown to maintain their integrity through serial transplantation in immune-compromised animals, practically, it is not always possible to have access to animals of suitable ages to continuously maintain these cells. We therefore explored vitrification as a cryopreservation technique for brain tumor-initiating cells. Tumor neurospheres were derived from five patients with glioblastoma multiforme (GBM). Cryopreservation in 90% serum and 10% dimethyl sulfoxide yielded greatest viability and could be explored in future studies. Vitrification yielded cells that maintained self-renewal and multipotentiality properties. Karyotypic analyses confirmed the presence of GBM hallmarks. Upon implantation into NOD/SCID mice, our vitrified cells reformed glioma masses that could be serially transplanted. Transcriptome analysis showed that the vitrified and nonvitrified samples in either the stem-like or differentiated states clustered together, providing evidence that vitrification does not change the genotype of frozen cells. Upon induction of differentiation, the transcriptomes of vitrified cells associated with the original primary tumors, indicating that tumor stem-like cells are a genetically distinct population from the differentiated mass, underscoring the importance of working with the relevant tumor-initiating population. Our results demonstrate that vitrification of brain tumor-initiating cells preserves the biological phenotype and genetic profiles of the cells. This should facilitate the establishment of a repository of tumor-initiating cells for subsequent experimental designs.


Asunto(s)
Criopreservación , Glioblastoma/patología , Neuronas/patología , Esferoides Celulares/patología , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Agregación Celular , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glicoproteínas/metabolismo , Humanos , Cariotipificación , Ratones , Ratones SCID , Células Madre Multipotentes/patología , Células Madre Neoplásicas/patología , Péptidos/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Am Soc Mass Spectrom ; 31(11): 2296-2304, 2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33104352

RESUMEN

A novel approach for phenotype prediction is developed for data-independent acquisition (DIA) mass spectrometric (MS) data without the need for peptide precursor identification using existing DIA software tools. The first step converts the DIA-MS data file into a new file format called DIA tensor (DIAT), which can be used for the convenient visualization of all the ions from peptide precursors and fragments. DIAT files can be fed directly into a deep neural network to predict phenotypes such as appearances of cats, dogs, and microscopic images. As a proof of principle, we applied this approach to 102 hepatocellular carcinoma samples and achieved an accuracy of 96.8% in distinguishing malignant from benign samples. We further applied a refined model to classify thyroid nodules. Deep learning based on 492 training samples achieved an accuracy of 91.7% in an independent cohort of 216 test samples. This approach surpassed the deep-learning model based on peptide and protein matrices generated by OpenSWATH. In summary, we present a new strategy for DIA data analysis based on a novel data format called DIAT, which enables facile two-dimensional visualization of DIA proteomics data. DIAT files can be directly used for deep learning for biological and clinical phenotype classification. Future research will interpret the deep-learning models emerged from DIAT analysis.


Asunto(s)
Espectrometría de Masas/métodos , Proteoma/análisis , Proteómica/métodos , Carcinoma Hepatocelular/química , Carcinoma Hepatocelular/diagnóstico , Aprendizaje Profundo , Humanos , Neoplasias Hepáticas/química , Neoplasias Hepáticas/diagnóstico , Péptidos/análisis , Programas Informáticos , Glándula Tiroides/química
20.
Oral Oncol ; 111: 105035, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33091845

RESUMEN

OBJECTIVES: We have previously identified and validated a panel of molecular prognostic markers (ATP13A3, SSR3, and ANO1) for Head and Neck Squamous Cell Carcinoma (HNSCC). The aim of this study was to investigate the consequence of ATP13A3 dysregulation on signaling pathways, to aid in formulating a therapeutic strategy targeting ATP13A3-overexpressing HNSCC. MATERIALS AND METHODS: Gene Set Enrichment Analysis (GSEA) was performed on HNSCC microarray expression data (Internal local dataset [n = 92], TCGA [n = 232], EMBL [n = 81]) to identify pathways associated with high expression of ATP13A3. Validation was performed using immunohistochemistry (IHC) on tissue microarrays (TMAs) of head and neck cancers (n = 333), staining for ATP13A3 and phosphorylated Aurora kinase A (phospho-T288). Short interfering RNA was used to knockdown ATP13A3 expression in patient derived HNSCC cell lines. Protein expression of ATP13A3 and Aurora kinase A was then assessed by immunoblotting. RESULTS: GSEA identified Aurora kinase pathway to be associated with high expression of ATP13A3 (p = 0.026). The Aurora kinase pathway was also associated with a trend towards poor prognosis and tumor aggressiveness (p = 0.086, 0.094, respectively). Furthermore, the immunohistochemical staining results revealed a significant association between Aurora kinase activity and high ATP13A3 expression (p < 0.001). Knockdown of ATP13A3 in human head and neck cell lines showed decrease in Aurora kinase A levels. CONCLUSION: Tumors with high ATP13A3 are associated with high Aurora kinase activity. This suggests a potential therapeutic role of Aurora kinase inhibitors in a subset of poor prognosis HNSCC patients with overexpression of ATP13A3.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Aurora Quinasa A/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Adenosina Trifosfatasas/genética , Aurora Quinasa A/antagonistas & inhibidores , Línea Celular Tumoral , Femenino , Silenciador del Gen , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunohistoquímica , Masculino , Proteínas de Transporte de Membrana/genética , Terapia Molecular Dirigida/métodos , Pronóstico , ARN Interferente Pequeño , Transducción de Señal/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Análisis de Matrices Tisulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA