Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 83(24): 4161-4178, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-38098449

RESUMEN

Current treatment approaches for renal cell carcinoma (RCC) face challenges in achieving durable tumor responses due to tumor heterogeneity and drug resistance. Combination therapies that leverage tumor molecular profiles could offer an avenue for enhancing treatment efficacy and addressing the limitations of current therapies. To identify effective strategies for treating RCC, we selected ten drugs guided by tumor biology to test in six RCC patient-derived xenograft (PDX) models. The multitargeted tyrosine kinase inhibitor (TKI) cabozantinib and mTORC1/2 inhibitor sapanisertib emerged as the most effective drugs, particularly when combined. The combination demonstrated favorable tolerability and inhibited tumor growth or induced tumor regression in all models, including two from patients who experienced treatment failure with FDA-approved TKI and immunotherapy combinations. In cabozantinib-treated samples, imaging analysis revealed a significant reduction in vascular density, and single-nucleus RNA sequencing (snRNA-seq) analysis indicated a decreased proportion of endothelial cells in the tumors. SnRNA-seq data further identified a tumor subpopulation enriched with cell-cycle activity that exhibited heightened sensitivity to the cabozantinib and sapanisertib combination. Conversely, activation of the epithelial-mesenchymal transition pathway, detected at the protein level, was associated with drug resistance in residual tumors following combination treatment. The combination effectively restrained ERK phosphorylation and reduced expression of ERK downstream transcription factors and their target genes implicated in cell-cycle control and apoptosis. This study highlights the potential of the cabozantinib plus sapanisertib combination as a promising treatment approach for patients with RCC, particularly those whose tumors progressed on immune checkpoint inhibitors and other TKIs. SIGNIFICANCE: The molecular-guided therapeutic strategy of combining cabozantinib and sapanisertib restrains ERK activity to effectively suppress growth of renal cell carcinomas, including those unresponsive to immune checkpoint inhibitors.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Sistema de Señalización de MAP Quinasas , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Diana Mecanicista del Complejo 1 de la Rapamicina , Células Endoteliales/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Anilidas/farmacología , Anilidas/uso terapéutico , ARN Nuclear Pequeño/uso terapéutico
2.
Clin Cancer Res ; 29(24): 5104-5115, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-37819944

RESUMEN

PURPOSE: To investigate the efficacy, safety, and biomarkers of systemic chemotherapy with oxaliplatin, leucovorin, and 5-fluorouracil (FOLFOX) in combination with lenvatinib and toripalimab as the first-line treatment for advanced hepatocellular carcinoma (HCC) with extrahepatic metastasis. PATIENTS AND METHODS: In this biomolecular exploratory, phase II trial, eligible patients underwent the triple combination therapy of lenvatinib, toripalimab, plus FOLFOX chemotherapy. Primary endpoint was progression-free survival (PFS) rate at 6 months by RECIST v1.1. Single-nucleus RNA sequencing (snRNA-seq) of tumor biopsy samples was performed for exploratory biomarker analyses. RESULTS: Between November 19, 2019, and July 4, 2021, 30 patients were enrolled. The primary endpoint was a 6-month PFS rate of 66.7%, with a median PFS of 9.73 months [95% confidence interval (CI), 2.89-16.58]. The median overall survival (OS) was 14.63 months (95% CI, 11.77-17.50), with an objective response rate of 43.3%. Twenty-four (80.0%) patients exhibited high-risk features, among whom the median OS and PFS were 13.7 months (95% CI, 9.24-18.16) and 8.3 months (95% CI, 3.02-13.58), respectively. The most common adverse events were neutropenia, and increased aspartate aminotransferase and alanine aminotransferase levels. Exploratory analyses of snRNA-seq profiles suggested that patients with higher abundance of tumor-infiltrating immune cells were more likely to benefit from this combination. In addition, two subtypes of hepatocytes (AKR1C2+ and CFHR4+ malignant hepatocytes) were associated with reduced clinical benefits. CONCLUSIONS: FOLFOX chemotherapy in combination with lenvatinib and toripalimab showed promising antitumor activity with manageable toxicities in advanced HCC with extrahepatic metastasis. AKR1C2+ and CFHR4+ hepatocyte subtypes may be predictive biomarkers of resistance to the combination therapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Fluorouracilo , Leucovorina , Biomarcadores , ARN Nuclear Pequeño/uso terapéutico
3.
J Neuroinflammation ; 20(1): 152, 2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37365617

RESUMEN

BACKGROUND: Ambient RNAs contamination in single-nuclei RNA sequencing (snRNA-seq) is a challenging problem, but the consequences of ambient RNAs contamination of damaged and/or diseased tissues are poorly understood. Cognitive impairments and white/gray matter injuries are characteristic of deeper cerebral hypoperfusion mouse models induced by bilateral carotid artery stenosis (BCAS), but the molecular mechanisms still need to be further explored. More importantly, the BCAS mice can also offer an excellent model to examine the signatures of ambient RNAs contamination in damaged tissues when performing snRNA-seq. METHODS: After the sham and BCAS mice were established, cortex-specific single-nuclei libraries were constructed. Single-nuclei transcriptomes were described informatically by the R package Seurat, and ambient RNA markers of were identified in each library. Then, after removing ambient RNAs in each sample using the in silico approaches, the combination of CellBender and subcluster cleaning, single-nuclei transcriptomes were reconstructed. Next, the comparison of ambient RNA contamination was performed using irGSEA analysis before and after the in silico approaches. Finally, further bioinformatic analyses were performed. RESULTS: The ambient RNAs are more predominant in the BCAS group than the sham group. The contamination mainly originated from damaged neuronal nuclei, but could be reduced largely using the in silico approaches. The integrative analysis of cortex-specific snRNA-seq data and the published bulk transcriptome revealed that microglia and other immune cells were the primary effectors. In the sequential microglia/immune subgroups analysis, the subgroup of Apoe+ MG/Mac (microglia/macrophages) was identified. Interestingly, this subgroup mainly participated in the pathways of lipid metabolism, associated with the phagocytosis of cell debris. CONCLUSIONS: Taken together, our current study unravels the features of ambient RNAs in snRNA-seq datasets under diseased conditions, and the in silico approaches can effectively eliminate the incorrected cell annotation and following misleading analysis. In the future, snRNA-seq data analysis should be carefully revisited, and ambient RNAs removal needs to be taken into consideration, especially for those diseased tissues. To our best knowledge, our study also offers the first cortex-specific snRNA-seq data of deeper cerebral hypoperfusion, which provides with novel therapeutic targets.


Asunto(s)
Estenosis Carotídea , Microglía , Animales , Ratones , Microglía/metabolismo , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/farmacología , ARN Nuclear Pequeño/uso terapéutico , Macrófagos , Estenosis Carotídea/complicaciones , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Ratones Endogámicos C57BL
4.
Hum Mol Genet ; 27(14): 2466-2476, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29701768

RESUMEN

Familial dysautonomia (FD) is a rare genetic disease with no treatment, caused by an intronic point mutation (c.2204+6T>C) that negatively affects the definition of exon 20 in the elongator complex protein 1 gene (ELP1 also known as IKBKAP). This substitution modifies the 5' splice site and, in combination with regulatory splicing factors, induces different levels of exon 20 skipping, in various tissues. Here, we evaluated the therapeutic potential of a novel class of U1 snRNA molecules, exon-specific U1s (ExSpeU1s), in correcting ELP1 exon 20 recognition. Lentivirus-mediated expression of ELP1-ExSpeU1 in FD fibroblasts improved ELP1 splicing and protein levels. We next focused on a transgenic mouse model that recapitulates the same tissue-specific mis-splicing seen in FD patients. Intraperitoneal delivery of ELP1-ExSpeU1s-adeno-associated virus particles successfully increased the production of full-length human ELP1 transcript and protein. This splice-switching class of molecules is the first to specifically correct the ELP1 exon 20 splicing defect. Our data provide proof of principle of ExSpeU1s-adeno-associated virus particles as a novel therapeutic strategy for FD.


Asunto(s)
Proteínas Portadoras/genética , Disautonomía Familiar/terapia , Terapia Genética , ARN Nuclear Pequeño/genética , Empalme Alternativo/genética , Animales , Proteínas Portadoras/uso terapéutico , Dependovirus/genética , Modelos Animales de Enfermedad , Disautonomía Familiar/genética , Disautonomía Familiar/fisiopatología , Exones/genética , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Intrones/genética , Ratones , Ratones Transgénicos , Empalme del ARN/genética , ARN Nuclear Pequeño/uso terapéutico , Factores de Elongación Transcripcional
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 15-20, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27639833

RESUMEN

The c.2101A>G synonymous change (p.G674G) in the gene for ATR, a key player in the DNA-damage response, has been the first identified genetic cause of Seckel Syndrome (SS), an orphan disease characterized by growth and mental retardation. This mutation mainly causes exon 9 skipping, through an ill-defined mechanism. Through ATR minigene expression studies, we demonstrated that the detrimental effect of this mutation (6±1% of correct transcripts only) depends on the poor exon 9 definition (47±4% in the ATRwt context), because the change was ineffective when the weak 5' or the 3' splice sites (ss) were strengthened (scores from 0.54 to 1) by mutagenesis. Interestingly, the exonic c.2101A nucleotide is conserved across species, and the SS-causing mutation is predicted to concurrently strengthen a Splicing Silencer (ESS) and weaken a Splicing Enhancer (ESE). Consistently, the artificial c.2101A>C change, predicted to weaken the ESE only, moderately impaired exon inclusion (28±7% of correct transcripts). The observation that an antisense oligonucleotide (AONATR) targeting the c.2101A position recovers exon inclusion in the mutated context supports a major role of the underlying ESS. A U1snRNA variant (U1ATR) designed to perfectly base-pair the weak 5'ss, rescued exon inclusion (63±3%) in the ATRSS-allele. Most importantly, upon lentivirus-mediated delivery, the U1ATR partially rescued ATR mRNA splicing (from ~19% to ~54%) and protein (from negligible to ~6%) in embryonic fibroblasts derived from humanized ATRSS mice. Altogether these data elucidate the molecular mechanisms of the ATR c.2101A>G mutation and identify two potential complementary RNA-based therapies for Seckel syndrome.


Asunto(s)
Enanismo/terapia , Exones , Vectores Genéticos/uso terapéutico , Microcefalia/terapia , Oligonucleótidos Antisentido/uso terapéutico , Empalme del ARN , ARN Nuclear Pequeño/uso terapéutico , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Secuencia de Bases , Línea Celular , Enanismo/genética , Facies , Terapia Genética , Vectores Genéticos/genética , Células HEK293 , Humanos , Intrones , Lentivirus/genética , Ratones , Microcefalia/genética , Oligonucleótidos Antisentido/genética , Mutación Puntual , ARN Nuclear Pequeño/genética
6.
Blood ; 122(23): 3825-31, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-24085767

RESUMEN

Antisense molecules are emerging as a powerful tool to correct splicing defects. Recently, we identified a homozygous deep-intronic mutation (F5 c.1296+268A>G) activating a cryptic donor splice site in a patient with severe coagulation factor V (FV) deficiency and life-threatening bleeding episodes. Here, we assessed the ability of 2 mutation-specific antisense molecules (a morpholino oligonucleotide [MO] and an engineered U7 small nuclear RNA [snRNA]) to correct this splicing defect. COS-1 and HepG2 cells transfected with a F5 minigene construct containing the patient's mutation expressed aberrant messenger RNA (mRNA) in excess of normal mRNA. Treatment with mutation-specific antisense MO (1-5 µM) or a construct expressing antisense U7snRNA (0.25-2 µg) dose-dependently increased the relative amount of correctly spliced mRNA by 1 to 2 orders of magnitude, whereas control MO and U7snRNA were ineffective. Patient-derived megakaryocytes obtained by differentiation of circulating progenitor cells did not express FV, but became positive for FV at immunofluorescence staining after administration of antisense MO or U7snRNA. However, treatment adversely affected cell viability, mainly because of the transfection reagents used to deliver the antisense molecules. Our data provide in vitro and ex vivo proof of principle for the efficacy of RNA therapy in severe FV deficiency, but additional cytotoxicity studies are warranted.


Asunto(s)
Deficiencia del Factor V/genética , Deficiencia del Factor V/terapia , Factor V/genética , Terapia Genética , Mutación , ARN sin Sentido/uso terapéutico , Animales , Células COS , Chlorocebus aethiops , Deficiencia del Factor V/sangre , Células Hep G2 , Homocigoto , Humanos , Intrones , Megacariocitos/metabolismo , Megacariocitos/patología , Empalme del ARN/genética , ARN sin Sentido/genética , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/uso terapéutico , Transfección
7.
EMBO Mol Med ; 5(7): 1128-45, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23716398

RESUMEN

Exon skipping mediated by antisense oligoribonucleotides (AON) is a promising therapeutic approach for genetic disorders, but has not yet been evaluated for cardiac diseases. We investigated the feasibility and efficacy of viral-mediated AON transfer in a Mybpc3-targeted knock-in (KI) mouse model of hypertrophic cardiomyopathy (HCM). KI mice carry a homozygous G>A transition in exon 6, which results in three different aberrant mRNAs. We identified an alternative variant (Var-4) deleted of exons 5-6 in wild-type and KI mice. To enhance its expression and suppress aberrant mRNAs we designed AON-5 and AON-6 that mask splicing enhancer motifs in exons 5 and 6. AONs were inserted into modified U7 small nuclear RNA and packaged in adeno-associated virus (AAV-U7-AON-5+6). Transduction of cardiac myocytes or systemic administration of AAV-U7-AON-5+6 increased Var-4 mRNA/protein levels and reduced aberrant mRNAs. Injection of newborn KI mice abolished cardiac dysfunction and prevented left ventricular hypertrophy. Although the therapeutic effect was transient and therefore requires optimization to be maintained over an extended period, this proof-of-concept study paves the way towards a causal therapy of HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/terapia , Proteínas Portadoras/genética , Exones , Oligorribonucleótidos Antisentido/uso terapéutico , ARN Nuclear Pequeño/uso terapéutico , Adenoviridae/genética , Empalme Alternativo , Animales , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/fisiopatología , Técnicas de Sustitución del Gen , Terapia Genética , Células HEK293 , Corazón/fisiopatología , Humanos , Hipertrofia Ventricular Izquierda/prevención & control , Ratones , Mutación , Miocardio/metabolismo , Miocardio/patología , Oligorribonucleótidos Antisentido/administración & dosificación , Oligorribonucleótidos Antisentido/genética , Isoformas de Proteínas/genética , ARN Mensajero/genética , ARN Nuclear Pequeño/administración & dosificación , ARN Nuclear Pequeño/genética , Transducción Genética
8.
Hum Mol Genet ; 21(11): 2559-71, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22388933

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe neuromuscular disorder caused by mutations in the dystrophin gene that result in the absence of functional protein. Antisense-mediated exon skipping is one of the most promising approaches for the treatment of DMD and recent clinical trials have demonstrated encouraging results. However, antisense oligonucleotide-mediated exon skipping for DMD still faces major hurdles such as extremely low efficacy in the cardiac muscle, poor cellular uptake and relatively rapid clearance from circulation, which means that repeated administrations are required to achieve some therapeutic efficacy. To overcome these limitations, we previously proposed the use of small nuclear RNAs (snRNAs), especially U7snRNA to shuttle the antisense sequences after vectorization into adeno-associated virus (AAV) vectors. In this study, we report for the first time the efficiency of the AAV-mediated exon skipping approach in the utrophin/dystrophin double-knockout (dKO) mouse which is a very severe and progressive mouse model of DMD. Following a single intravenous injection of scAAV9-U7ex23 in dKO mice, near-normal levels of dystrophin expression were restored in all muscles examined, including the heart. This resulted in a considerable improvement of their muscle function and dystrophic pathology as well as a remarkable extension of the dKO mice lifespan. These findings suggest great potential for AAV-U7 in systemic treatment of the DMD phenotype.


Asunto(s)
Dependovirus/genética , Distrofina/genética , Exones , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , ARN Nuclear Pequeño/genética , Utrofina/genética , Animales , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Distrofina/metabolismo , Terapia Genética , Ratones , Distrofia Muscular de Duchenne/patología , Fenotipo , ARN Nuclear Pequeño/uso terapéutico , Utrofina/metabolismo
9.
Methods Mol Biol ; 867: 259-71, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22454067

RESUMEN

Antisense-mediated splicing modulation of premessenger RNA represents a novel therapeutic strategy for several types of pathologies such as genetic disorders, cancers, and infectious diseases. Antisense oligonucleotides designed to bind to specific mRNA molecules have been actively developed for more than 20 years as a form of molecular medicine to modulate splicing patterns or inhibit protein translation. More recently, small nuclear RNA such as U7 or U1 small nuclear RNA have been used to carry antisense sequences, offering the advantage of long-term effect when delivered to cells using viral vectors. We have previously demonstrated the therapeutic potential of U7snRNA targeting dystrophin mRNA as a treatment for Duchenne muscular dystrophy. In particular, we showed that bifunctional U7 snRNAs harboring silencer motifs induce complete skipping of exon 51, and thus restore dystrophin expression in DMD patients cells to near wild-type levels. These new constructs are very promising for the optimization of therapeutic exon skipping for DMD, but also offer powerful and versatile tools to modulate pre-mRNA splicing in a wide range of applications. Here, we outline the design of these U7snRNA constructs to achieve efficient exon-skipping and describe methods to evaluate the efficacy of such U7snRNA constructs in vitro using the dystrophin gene as an example.


Asunto(s)
Distrofina/genética , Exones , Vectores Genéticos/genética , Distrofia Muscular de Duchenne/genética , ARN Nuclear Pequeño/genética , Animales , Línea Celular , Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Humanos , Lentivirus/genética , Ratones , Distrofia Muscular de Duchenne/terapia , Mioblastos/metabolismo , ARN Nuclear Pequeño/uso terapéutico , Transducción Genética/métodos
10.
Expert Opin Biol Ther ; 9(7): 849-66, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19527108

RESUMEN

Muscular dystrophies refer to a group of inherited disorders characterized by progressive muscle weakness, wasting and degeneration. So far, there is no effective treatment but new gene-based therapies are currently being developed with particular noted advances in using conventional gene replacement strategies, RNA-based approaches, or cell-based gene therapy with a main focus on Duchenne muscular dystrophy (DMD). DMD is the most common and severe form of muscular dystrophy and current treatments are far from adequate. However, genetic and cell-based therapies, in particular exon skipping induced by antisense strategies, and corrective gene therapy via functionally engineered dystrophin genes hold great promise, with several clinical trials ongoing. Proof-of-concept of exon skipping has been obtained in animal models, and most recently in clinical trials; this approach represents a promising therapy for a subset of patients. In addition, gene-delivery-based strategies exist both for antisense-induced reading frame restoration, and for highly efficient delivery of functional dystrophin mini- and micro-genes to muscle fibres in vivo and muscle stem cells ex-vivo. In particular, AAV-based vectors show efficient systemic gene delivery to skeletal muscle directly in vivo, and lentivirus-based vectors show promise of combining ex vivo gene modification strategies with cell-mediated therapies.


Asunto(s)
Terapia Genética/métodos , Terapia Genética/tendencias , Distrofias Musculares/terapia , Animales , Humanos , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología , Miostatina/antagonistas & inhibidores , ARN/genética , ARN Nuclear Pequeño/uso terapéutico
11.
Hum Mol Genet ; 18(3): 546-55, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19010792

RESUMEN

In spinal muscular atrophy (SMA), the leading genetic cause of early childhood death, the survival motor neuron 1 gene (SMN1) is deleted or inactivated. The nearly identical SMN2 gene has a silent mutation that impairs the utilization of exon 7 and the production of functional protein. It has been hypothesized that therapies boosting SMN2 exon 7 inclusion might prevent or cure SMA. Exon 7 inclusion can be stimulated in cell culture by oligonucleotides or intracellularly expressed RNAs, but evidence for an in vivo improvement of SMA symptoms is lacking. Here, we unambiguously confirm the above hypothesis by showing that a bifunctional U7 snRNA that stimulates exon 7 inclusion, when introduced by germline transgenesis, can efficiently complement the most severe mouse SMA model. These results are significant for the development of a somatic SMA therapy, but may also provide new means to study pathophysiological aspects of this devastating disease.


Asunto(s)
Terapia Genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , ARN Nuclear Pequeño/uso terapéutico , Animales , Secuencia de Bases , Exones , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Atrofia Muscular Espinal/metabolismo , Empalme del ARN , ARN Nuclear Pequeño/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
13.
Blood ; 101(1): 104-11, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12393543

RESUMEN

Mutations at nucleotides 654, 705, or 745 in intron 2 of the human beta-globin gene activate aberrant 3' and 5' splice sites within the intron and prevent correct splicing of beta-globin pre-mRNA, resulting in inhibition of beta-globin synthesis and in consequence beta-thalassemia. Transfection of HeLa cells expressing the 3 thalassemic mutants with modified U7 snRNA (U7.623), containing a sequence antisense to a region between the aberrant splice sites, reduced the incorrect splicing of pre-mRNA and led to increased levels of the correctly spliced beta-globin mRNA and protein. A lentiviral vector carrying the U7.623 gene was effective in restoration of correct splicing in the model cell lines for at least 6 months. Importantly, the therapeutic value of this system was demonstrated in hematopoietic stem cells and erythroid progenitor cells from a patient with IVS2-745/IVS2-1 thalassemia. Twelve days after transduction of the patient cells with the U7.623 lentiviral vector, the levels of correctly spliced beta-globin mRNA and hemoglobin A were approximately 25-fold over background. These results should be regarded as a proof of principle for lentiviral vector-based gene therapy for beta-thalassemia.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Hemoglobina A/metabolismo , ARN sin Sentido/farmacología , ARN Nuclear Pequeño/farmacología , Talasemia/terapia , Terapia Genética , Vectores Genéticos , Globinas/genética , Células HeLa , Hemoglobina A/genética , Humanos , Lentivirus/genética , Mutación , Empalme del ARN/efectos de los fármacos , ARN sin Sentido/administración & dosificación , ARN sin Sentido/uso terapéutico , ARN Nuclear Pequeño/administración & dosificación , ARN Nuclear Pequeño/uso terapéutico , Talasemia/sangre , Talasemia/genética , Transfección
14.
J Natl Cancer Inst ; 91(18): 1548-56, 1999 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10491431

RESUMEN

BACKGROUND: Expression of scatter factor (SF), also known as hepatocyte growth factor (HGF), and its receptor, c-met, is often associated with malignant progression of human tumors, including gliomas. Overexpression of SF/HGF in experimental gliomas enhances tumorigenicity and tumor-associated angiogenesis (i.e., growth of new blood vessels). However, the role of endogenous SF/HGF or c-met expression in the malignant progression of gliomas has not been examined directly. In this study, we tested the hypothesis that human glioblastomas can be SF/HGF-c-met dependent and that a reduction in endogenous SF/HGF or c-met expression can lead to inhibition of tumor growth and tumorigenicity. METHODS: Expression of the SF/HGF and c-met genes was inhibited by transfecting glioblastoma cells with chimeric transgenes consisting of U1 small nuclear RNA, a hammerhead ribozyme, and antisense sequences. The effects of reduced SF/HGF and c-met expression on 1) SF/HGF-dependent induction of immediate early genes (c-fos and c-jun), indicative of signal transduction; 2) anchorage-independent colony formation (clonogenicity), an in vitro correlate of solid tumor malignancy; and 3) intracranial tumor formation in immunodeficient mice were quantified. Statistical tests were two-sided. RESULTS: Introduction of the transgenes into glioblastoma cells reduced expression of the SF/HGF and c-met genes to as little as 2% of control cell levels. Reduction in c-met expression specifically inhibited SF/HGF-dependent signal transduction (P<.01). Inhibition of SF/HGF or c-met expression in glioblastoma cells possessing an SF/HGF-c-met autocrine loop reduced tumor cell clonogenicity (P =.005 for SF/HGF and P=.009 for c-met) and substantially inhibited tumorigenicity (P<.0001) and tumor growth in vivo (P<.0001). CONCLUSIONS: To our knowledge, this is the first successful inhibition of SF/HGF and c-met expression in a tumor model directly demonstrating a role for endogenous SF/HGF and c-met in human glioblastoma. Our results suggest that targeting the SF/HGF-c-met signaling pathway may be an important approach in controlling tumor progression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , ARN Catalítico/genética , ARN Neoplásico/metabolismo , ARN Nuclear Pequeño/genética , Animales , Northern Blotting , Adhesión Celular , División Celular , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Terapia Genética , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/terapia , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Hibridación in Situ , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fenotipo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , ARN sin Sentido/genética , ARN sin Sentido/uso terapéutico , ARN Catalítico/metabolismo , ARN Neoplásico/genética , ARN Nuclear Pequeño/uso terapéutico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...