Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 29(14): 2651-2667, 2023 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-36780194

RESUMO

PURPOSE: Anaplastic lymphoma kinase (ALK) aberrations have been identified in pediatric-type infant gliomas, but their occurrence across age groups, functional effects, and treatment response has not been broadly established. EXPERIMENTAL DESIGN: We performed a comprehensive analysis of ALK expression and genomic aberrations in both newly generated and retrospective data from 371 glioblastomas (156 adult, 205 infant/pediatric, and 10 congenital) with in vitro and in vivo validation of aberrations. RESULTS: ALK aberrations at the protein or genomic level were detected in 12% of gliomas (45/371) in a wide age range (0-80 years). Recurrent as well as novel ALK fusions (LRRFIP1-ALK, DCTN1-ALK, PRKD3-ALK) were present in 50% (5/10) of congenital/infant, 1.4% (3/205) of pediatric, and 1.9% (3/156) of adult GBMs. ALK fusions were present as the only candidate driver in congenital/infant GBMs and were sometimes focally amplified. In contrast, adult ALK fusions co-occurred with other oncogenic drivers. No activating ALK mutations were identified in any age group. Novel and recurrent ALK rearrangements promoted STAT3 and ERK1/2 pathways and transformation in vitro and in vivo. ALK-fused GBM cellular and mouse models were responsive to ALK inhibitors, including in patient cells derived from a congenital GBM. Relevant to the treatment of infant gliomas, we showed that ALK protein appears minimally expressed in the forebrain at perinatal stages, and no gross effects on perinatal brain development were seen in pregnant mice treated with the ALK inhibitor ceritinib. CONCLUSIONS: These findings support use of brain-penetrant ALK inhibitors in clinical trials across infant, pediatric, and adult GBMs. See related commentary by Mack and Bertrand, p. 2567.


Assuntos
Glioblastoma , Glioma , Camundongos , Animais , Quinase do Linfoma Anaplásico/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Estudos Retrospectivos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Glioma/tratamento farmacológico
2.
Nat Immunol ; 23(8): 1148-1156, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35879449

RESUMO

Long recognized as an evolutionarily ancient cell type involved in tissue homeostasis and immune defense against pathogens, macrophages are being re-discovered as regulators of several diseases, including cancer. Tumor-associated macrophages (TAMs) represent the most abundant innate immune population in the tumor microenvironment (TME). Macrophages are professional phagocytic cells of the hematopoietic system specializing in the detection, phagocytosis and destruction of bacteria and other harmful micro-organisms, apoptotic cells and metabolic byproducts. In contrast to these healthy macrophage functions, TAMs support cancer cell growth and metastasis and mediate immunosuppressive effects on the adaptive immune cells of the TME. Cancer is one of the most potent insults on macrophage physiology, inducing changes that are intimately linked with disease progression. In this Review, we outline hallmarks of TAMs and discuss the emerging mechanisms that contribute to their pathophysiological adaptations and the vulnerabilities that provide attractive targets for therapeutic exploitation in cancer.


Assuntos
Neoplasias , Microambiente Tumoral , Progressão da Doença , Humanos , Macrófagos , Fagocitose
4.
Adv Biosyst ; 4(12): e1900307, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33274611

RESUMO

There is a need for novel analytical techniques to study the composition of single extracellular vesicles (EV). Such techniques are required to improve the understanding of heterogeneous EV populations, to allow identification of unique subpopulations, and to enable earlier and more sensitive disease detection. Because of the small size of EV and their low protein content, ultrahigh sensitivity technologies are required. Here, an immuno-droplet digital polymerase chain reaction (iddPCR) amplification method is described that allows multiplexed single EV protein profiling. Antibody-DNA conjugates are used to label EV, followed by stochastic microfluidic incorporation of single EV into droplets. In situ PCR with fluorescent reporter probes converts and amplifies the barcode signal for subsequent read-out by droplet imaging. In these proof-of-principle studies, it is shown that multiplex protein analysis is possible in single EV, opening the door for future analyses.


Assuntos
Vesículas Extracelulares , Técnicas Analíticas Microfluídicas/instrumentação , Reação em Cadeia da Polimerase , Animais , Linhagem Celular , Desenho de Equipamento , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Humanos , Camundongos , Reação em Cadeia da Polimerase/instrumentação , Reação em Cadeia da Polimerase/métodos , Análise de Célula Única/instrumentação
5.
Sci Rep ; 10(1): 15905, 2020 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-32963251

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
iScience ; 23(8): 101420, 2020 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-32795915

RESUMO

Glioblastoma (GBM) may arise from astrocytes through a multistep process involving a progressive accumulation of mutations. We explored whether GBM-derived extracellular vesicles (EVs) may facilitate neoplastic transformation and malignant growth of astrocytes. We utilized conditioned media (CM) of cultured glioma cells, its sequential filtration, diverse cell-based assays, RNA sequencing, and metabolic assays to compare the effects of EV-containing and EV-depleted CM. GBM EVs facilitated the neoplastic growth of pre-transformed astrocytes but not normal human or mouse astrocytes. They induced proliferation, self-renewal, and colony formation of pre-transformed astrocytes and enhanced astrocytoma growth in a mouse allograft model. GBM EVs appear to reprogram astrocyte metabolism by inducing a shift in gene expression that may be partly associated with EV-mediated transfer of full-length mRNAs encoding ribosomal proteins, oxidative phosphorylation, and glycolytic factors. Our study suggests an EV/extracellular RNA (exRNA)-mediated mechanism that contributes to astrocyte transformation via metabolic reprograming and implicates horizontal mRNA transfer.

7.
Sci Rep ; 9(1): 17252, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31754127

RESUMO

PD-1 is a target of cancer immunotherapy but responses are limited to a fraction of patients. Identifying patients with T cells subjected to PD-1-mediated inhibition will allow selection of suitable candidates for PD-1-blocking therapy and will improve the therapeutic success. We sought to develop an approach to detect PD-1-mediated inhibitory signaling. The cytoplasmic tail of PD-1 contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) encompassing Y223 and an immunoreceptor tyrosine-based switch motif (ITSM) encompassing Y248, which is indispensable for interaction of SHP-2 and delivery of PD-1 inhibitory function. We generated an antibody specific for phosphorylated PD-1-Y248 and examined PD-1pY248+ (pPD-1) expression in human T cells. pPD-1 was upregulated by TCR/CD3 + CD28 stimulation and simultaneous PD-1 ligation. pPD-1+CD8+ T cells were identified in human peripheral blood and had impaired effector function. pPD-1+ T cells were also detected in tumor-draining lymph nodes of tumor bearing mice and in biopsies of patients with glioblastoma multiform. Detection of pPD-1+ T cells might serve as a biomarker for identification of T cells subjected to PD-1-mediated immunosuppression.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Motivo de Inibição do Imunorreceptor Baseado em Tirosina/fisiologia , Receptor de Morte Celular Programada 1/metabolismo , Animais , Antígenos CD/metabolismo , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/metabolismo , Biomarcadores/sangue , Antígenos CD28/metabolismo , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Motivo de Inibição do Imunorreceptor Baseado em Tirosina/genética , Células Matadoras Naturais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Cultura Primária de Células , Receptor de Morte Celular Programada 1/genética , Receptores Imunológicos/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/metabolismo , Tirosina/metabolismo
8.
EBioMedicine ; 48: 23-35, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31628025

RESUMO

BACKGROUND: Malignant gliomas are rapidly progressive brain tumors with high mortality. Fluorescence guided surgery (FGS) with 5-aminolevulinic acid (5-ALA) provides fluorescent delineation of malignant tissue, which helps achieve maximum safe resection. 5-ALA-based fluorescence is due to preferential accumulation of the fluorophore protoporphyrin-IX (PpIX) in malignant glioma tissue. Additionally, gliomas cells release extracellular vesicles (EVs) which carry biomarkers of disease. Herein, we performed animal and human studies to investigate whether 5-ALA dosed glioma cells, in vitro and in vivo, release PpIX positive EVs in circulation which can be captured and analyzed. METHODS: We used imaging flow cytometry (IFC) to characterize PpIX-positive EVs released from 5-ALA-dosed glioma cells, glioma-bearing xenograft models, as well as patients with malignant glioma undergoing FGS. FINDINGS: We first show that glioma cells dosed with 5-ALA release 247-fold higher PpIX positive EVs compared to mock dosed glioma cells. Second, we demonstrate that the plasma of glioma-bearing mice (n = 2) dosed with 5-ALA contain significantly higher levels of circulating PpIX-positive EVs than their pre-dosing background (p = 0.004). Lastly, we also show that the plasma of patients with avidly fluorescent tumors (n = 4) undergoing FGS contain circulating PpIX-positive EVs at levels significantly higher than their pre-dosing background (p = 0.00009) and this rise in signal correlates with enhancing tumor volumes (r 2  = 0.888). INTERPRETATION: Our findings highlight the potential of plasma-derived PpIX-positive EV-based diagnostics for malignant gliomas, offering a novel liquid biopsy platform for confirming and monitoring tumor status.


Assuntos
Vesículas Extracelulares/metabolismo , Corantes Fluorescentes/administração & dosagem , Glioma/metabolismo , Ácidos Levulínicos/administração & dosagem , Fármacos Fotossensibilizantes/metabolismo , Protoporfirinas/metabolismo , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Glioma/diagnóstico , Glioma/cirurgia , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Imagem Óptica/métodos , Cirurgia Assistida por Computador , Ácido Aminolevulínico
9.
Cell ; 177(2): 231-242, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30951667

RESUMO

The Extracellular RNA Communication Consortium (ERCC) was launched to accelerate progress in the new field of extracellular RNA (exRNA) biology and to establish whether exRNAs and their carriers, including extracellular vesicles (EVs), can mediate intercellular communication and be utilized for clinical applications. Phase 1 of the ERCC focused on exRNA/EV biogenesis and function, discovery of exRNA biomarkers, development of exRNA/EV-based therapeutics, and construction of a robust set of reference exRNA profiles for a variety of biofluids. Here, we present progress by ERCC investigators in these areas, and we discuss collaborative projects directed at development of robust methods for EV/exRNA isolation and analysis and tools for sharing and computational analysis of exRNA profiling data.


Assuntos
Ácidos Nucleicos Livres/genética , Ácidos Nucleicos Livres/metabolismo , Vesículas Extracelulares/genética , Biomarcadores , Humanos , Bases de Conhecimento , MicroRNAs/genética , RNA/genética
10.
Oncogene ; 37(9): 1121-1141, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29242608

RESUMO

Glioblastoma multiforme (GBM) is a highly malignant primary brain cancer with a dreadful overall survival and for which treatment options are limited. Recent breakthroughs in novel immune-related treatment strategies for cancer have spurred interests in usurping the power of the patient's immune system to recognize and eliminate GBM. Here, we discuss the unique properties of GBM's tumor microenvironment, the effects of GBM standard on care therapy on tumor-associated immune cells, and review several approaches aimed at therapeutically targeting the immune system for GBM treatment. We believe that a comprehensive understanding of the intricate micro-environmental landscape of GBM will abound into the development of novel immunotherapy strategies for GBM patients.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Imunoterapia , Animais , Neoplasias Encefálicas/imunologia , Glioma/imunologia , Humanos
11.
J Cell Biochem ; 118(9): 2516-2527, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28230277

RESUMO

Glioblastoma Multiforme (GBM) is a highly malignant primary brain cancer that is associated with abysmal prognosis. The median survival of GBM patients is ∼15 months and there have not been any significant advance in therapies in over a decade, leaving treatment options limited. There is clearly an unmet need for GBM treatment. Immunotherapies are treatments based on usurping the power of the host's immune system to recognize and eliminate cancer cells. They have recently proven to be a successful strategy for combating a variety of cancers. Of the various types of immunotherapies, checkpoint blockade approaches have thus far produced significant clinical responses in several cancers including melanoma, non small-cell lung cancer, renal cancer, and prostate cancer. This review focuses on the biological rationale for using checkpoint blockade immunotherapeutic approaches in primary brain cancer and an up-to-date summary of current and ongoing checkpoint inhibitors-based clinical trials for malignant glioma. In addition, we expand on new concepts for further improving checkpoint blockade treatments, with a particular focus on the advantages of using genetically engineered mouse models for studies of immunotherapies in GBM. J. Cell. Biochem. 118: 2516-2527, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Imunoterapia/métodos , Neoplasias Experimentais , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Glioblastoma/genética , Glioblastoma/imunologia , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Camundongos , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia
12.
Acta Biomater ; 20: 32-38, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25861948

RESUMO

Current methods for treatment of high-risk neuroblastoma patients include surgical intervention, in addition to systemic chemotherapy. However, only limited therapeutic tools are available to pediatric surgeons involved in neuroblastoma care, so the development of intraoperative treatment modalities is highly desirable. This study presents a silk film library generated for focal therapy of neuroblastoma; these films were loaded with either the chemotherapeutic agent doxorubicin or the targeted drug crizotinib. Drug release kinetics from the silk films were fine-tuned by changing the amount and physical crosslinking of silk; doxorubicin loaded films were further refined by applying a gold nanocoating. Doxorubicin-loaded, physically crosslinked silk films showed the best in vitro activity and superior in vivo activity in orthotopic neuroblastoma studies when compared to the doxorubicin-equivalent dose administered intravenously. Silk films were also suitable for delivery of the targeted drug crizotinib, as crizotinib-loaded silk films showed an extended release profile and an improved response both in vitro and in vivo when compared to freely diffusible crizotinib. These findings, when combined with prior in vivo data on silk, support a viable future for silk-based anticancer drug delivery systems.


Assuntos
Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos , Neuroblastoma/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Seda/química , Animais , Linhagem Celular Tumoral , Crizotinibe , Doxorrubicina/uso terapêutico , Feminino , Humanos , Camundongos Nus , Pirazóis/uso terapêutico , Piridinas/uso terapêutico
13.
J Clin Invest ; 124(12): 5159-74, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25401476

RESUMO

Angioplasty and stenting is the primary treatment for flow-limiting atherosclerosis; however, this strategy is limited by pathological vascular remodeling. Using a systems approach, we identified a role for the network hub gene glutathione peroxidase-1 (GPX1) in pathological remodeling following human blood vessel stenting. Constitutive deletion of Gpx1 in atherosclerotic mice recapitulated this phenotype of increased vascular smooth muscle cell (VSMC) proliferation and plaque formation. In an independent patient cohort, gene variant pair analysis identified an interaction of GPX1 with the orphan protooncogene receptor tyrosine kinase ROS1. A meta-analysis of the only genome-wide association studies of human neointima-induced in-stent stenosis confirmed the association of the ROS1 variant with pathological remodeling. Decreased GPX1 expression in atherosclerotic mice led to reductive stress via a time-dependent increase in glutathione, corresponding to phosphorylation of the ROS1 kinase activation site Y2274. Loss of GPX1 function was associated with both oxidative and reductive stress, the latter driving ROS1 activity via s-glutathiolation of critical residues of the ROS1 tyrosine phosphatase SHP-2. ROS1 inhibition with crizotinib and deglutathiolation of SHP-2 abolished GPX1-mediated increases in VSMC proliferation while leaving endothelialization intact. Our results indicate that GPX1-dependent alterations in oxido-reductive stress promote ROS1 activation and mediate vascular remodeling.


Assuntos
Aterosclerose/enzimologia , Proteínas Musculares/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Remodelação Vascular , Substituição de Aminoácidos , Animais , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Crizotinibe , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Glutationa Peroxidase/biossíntese , Glutationa Peroxidase/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Músculo Liso Vascular/patologia , Mutação de Sentido Incorreto , Miócitos de Músculo Liso/patologia , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Pirazóis/farmacologia , Piridinas/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Glutationa Peroxidase GPX1
14.
Stem Cells ; 32(2): 338-48, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24115218

RESUMO

Glioblastoma multiforme (GBM) is the most lethal form of primary brain tumors, characterized by highly invasive and aggressive tumors that are resistant to all current therapeutic options. GBMs are highly heterogeneous in nature and contain a small but highly tumorigenic and self-renewing population of stem or initiating cells (glioblastoma stem cells or GSCs). GSCs have been shown to contribute to tumor propagation and resistance to current therapeutic modalities. Recent studies of human GBMs have elucidated the genetic alterations common in these tumors, but much remains unknown about specific signaling pathways that regulate GSCs. Here we identify a distinct fraction of cells in a genetically engineered mouse model of EGFR-driven GBM that respond to anti-EGFR therapy by inducing high levels of c-MET expression. The MET-positive cells displayed clonogenic potential and long-term self-renewal ability in vitro and are capable of differentiating into multiple lineages. The MET-positive GBM cells are resistant to radiation and highly tumorigenic in vivo. Activation of MET signaling led to an increase in expression of the stemness transcriptional regulators Oct4, Nanog, and Klf4. Pharmacological inhibition of MET activity in GSCs prevented the activation of Oct4, Nanog, and Klf4 and potently abrogated stemness. Finally, the MET expressing cells were preferentially localized in perivascular regions of mouse tumors consistent with their function as GSCs. Together, our findings indicate that EGFR inhibition in GBM induces MET activation in GSCs, which is a functional requisite for GSCs activity and thus represents a promising therapeutic target.


Assuntos
Neoplasias Encefálicas/genética , Receptores ErbB/genética , Glioblastoma/genética , Proteínas Proto-Oncogênicas c-met/genética , Animais , Antineoplásicos , Neoplasias Encefálicas/patologia , Linhagem da Célula , Receptores ErbB/antagonistas & inibidores , Glioblastoma/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Transdução de Sinais/genética
15.
Nat Med ; 18(12): 1835-40, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23142818

RESUMO

Glioblastomas shed large quantities of small, membrane-bound microvesicles into the circulation. Although these hold promise as potential biomarkers of therapeutic response, their identification and quantification remain challenging. Here, we describe a highly sensitive and rapid analytical technique for profiling circulating microvesicles directly from blood samples of patients with glioblastoma. Microvesicles, introduced onto a dedicated microfluidic chip, are labeled with target-specific magnetic nanoparticles and detected by a miniaturized nuclear magnetic resonance system. Compared with current methods, this integrated system has a much higher detection sensitivity and can differentiate glioblastoma multiforme (GBM) microvesicles from nontumor host cell-derived microvesicles. We also show that circulating GBM microvesicles can be used to analyze primary tumor mutations and as a predictive metric of treatment-induced changes. This platform could provide both an early indicator of drug efficacy and a potential molecular stratifier for human clinical trials.


Assuntos
Biomarcadores Tumorais/química , Glioblastoma/tratamento farmacológico , Vesículas Transportadoras/química , Linhagem Celular Tumoral , Glioblastoma/sangue , Glioblastoma/fisiopatologia , Humanos , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita , Técnicas Analíticas Microfluídicas
16.
Cancer Res ; 72(15): 3764-74, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22659450

RESUMO

Patients with lung cancer often present with metastatic disease and therefore have a very poor prognosis. The recent discovery of several novel ROS receptor tyrosine kinase molecular alterations in non-small cell lung cancer (NSCLC) presents a therapeutic opportunity for the development of new targeted treatment strategies. Here, we report that the NSCLC-derived fusion CD74-ROS, which accounts for 30% of all ROS fusion kinases in NSCLC, is an active and oncogenic tyrosine kinase. We found that CD74-ROS-expressing cells were highly invasive in vitro and metastatic in vivo. Pharmacologic inhibition of CD74-ROS kinase activity reversed its transforming capacity by attenuating downstream signaling networks. Using quantitative phosphoproteomics, we uncovered a mechanism by which CD74-ROS activates a novel pathway driving cell invasion. Expression of CD74-ROS resulted in the phosphorylation of the extended synaptotagmin-like protein E-Syt1. Elimination of E-Syt1 expression drastically reduced invasiveness both in vitro and in vivo without modifying the oncogenic activity of CD74-ROS. Furthermore, expression of CD74-ROS in noninvasive NSCLC cell lines readily conferred invasive properties that paralleled the acquisition of E-Syt1 phosphorylation. Taken together, our findings indicate that E-Syt1 is a mediator of cancer cell invasion and molecularly define ROS fusion kinases as therapeutic targets in the treatment of NSCLC.


Assuntos
Antígenos de Diferenciação de Linfócitos B/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Antígenos de Histocompatibilidade Classe II/genética , Proteínas Sensoras de Cálcio Intracelular/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteínas de Membrana/metabolismo , Proteínas de Fusão Oncogênica/fisiologia , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Animais , Antígenos de Diferenciação de Linfócitos B/metabolismo , Antígenos de Diferenciação de Linfócitos B/fisiologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/metabolismo , Antígenos de Histocompatibilidade Classe II/fisiologia , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos SCID , Invasividade Neoplásica/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Fosforilação/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Ratos , Transdução de Sinais/genética
17.
ACS Nano ; 3(9): 2495-504, 2009 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-19673534

RESUMO

Small interfering RNAs (siRNAs) mediate cleavage of specific, complementary mRNA sequences and thus regulate gene expression. Not surprisingly, their use for treatment of diseases that are rooted in aberrant gene expression, such as cancer, has become a paradigm that has gained wide interest. Here, we report the development of dendrimer-conjugated magnetofluorescent nanoworms that we call "dendriworms" as a modular platform for siRNA delivery in vivo. This platform maximizes endosomal escape to robustly produce protein target knockdown in vivo, and is tolerated well in mouse brain. We demonstrate that siRNA-carrying dendriworms can be readily internalized by cells and enable endosomal escape across a wide range of loading doses, whereas dendrimers or nanoworms alone are inefficient. Further, we show that dendriworms carrying siRNA against the epidermal growth factor receptor (EGFR) reduce protein levels of EGFR in human glioblastoma cells by 70-80%, 2.5-fold more efficiently than commercial cationic lipids. Dendriworms were well-tolerated after 7-days of convection-enhanced delivery to the mouse brain and in an EGFR-driven transgenic model of glioblastoma, anti- EGFR dendriworms led to specific and significant suppression of EGFR expression. Collectively, these data establish dendriworms as a multimodal platform that enables fluorescent tracking of siRNA delivery in vivo, cellular entry, endosomal escape, and knockdown of target proteins.


Assuntos
Dendrímeros/química , Dendrímeros/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Nanoestruturas/química , RNA Interferente Pequeno/metabolismo , Animais , Sequência de Bases , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Citosol/metabolismo , Dendrímeros/síntese química , Dendrímeros/toxicidade , Portadores de Fármacos/síntese química , Portadores de Fármacos/toxicidade , Endocitose , Receptores ErbB/deficiência , Receptores ErbB/genética , Receptores ErbB/metabolismo , Corantes Fluorescentes/química , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Inativação Gênica , Glioblastoma/genética , Glioblastoma/patologia , Células HeLa , Humanos , Magnetismo , Camundongos , Camundongos Transgênicos , RNA Interferente Pequeno/genética , Fatores de Tempo
18.
Mech Dev ; 123(12): 869-80, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17070019

RESUMO

PTP (protein-tyrosine phosphatase)-PEST is a ubiquitously expressed cellular regulator of integrin signalling. It has been shown to bind several molecules such as Shc, paxillin and Grb2, that are involved downstream of FAK (focal adhesion kinase) pathway. Through its specific association to p130cas and further dephosphorylation, PTP-PEST plays a critical role in cell-matrix interactions, which are essential during embryogenesis. We report here that ablation of the gene leads to early embryonic lethality, correlating well with the high expression of the protein during embryonic development. We observed an increased level of tyrosine phosphorylation of p130cas protein in E9.5 PTP-PEST(-/-) embryos, a first evidence of biochemical defect leading to abnormal growth and development. Analysis of null mutant embryos revealed that they reach gastrulation, initiate yolk sac formation, but fail to progress through normal subsequent developmental events. E9.5-10.5 PTP-PEST(-/-) embryos had morphological abnormalities such as defective embryo turning, improper somitogenesis and vasculogenesis, impaired liver development, accompanied by degeneration in both neuroepithelium and somatic epithelia. Moreover, in embryos surviving until E10.5, the caudal region was truncated, with severe mesenchyme deficiency and no successful liver formation. Defects in embryonic mesenchyme as well as subsequent failure of proper vascularization, liver development and somatogenesis, seemed likely to induce lethality at this stage of development, and these results confirm that PTP-PEST plays an essential function in early embryogenesis.


Assuntos
Vasos Sanguíneos/embriologia , Embrião de Mamíferos/irrigação sanguínea , Genes Letais , Fígado/embriologia , Sistema Nervoso/embriologia , Proteínas Tirosina Fosfatases/fisiologia , Albuminas/genética , Albuminas/metabolismo , Animais , Aorta/citologia , Vasos Sanguíneos/enzimologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/enzimologia , Desenvolvimento Embrionário/genética , Células Endoteliais/citologia , Fígado/anormalidades , Fígado/enzimologia , Mesoderma/citologia , Mesoderma/enzimologia , Camundongos , Camundongos Mutantes , Sistema Nervoso/enzimologia , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 12 , Proteínas Tirosina Fosfatases/análise , Proteínas Tirosina Fosfatases/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Tirosina/metabolismo , Saco Vitelino/irrigação sanguínea , Saco Vitelino/citologia
19.
Development ; 132(1): 165-76, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15576410

RESUMO

Mouse embryos genetically null for all alphav integrins develop intracerebral hemorrhage owing to defective interactions between blood vessels and brain parenchymal cells. Here, we have used conditional knockout technology to address whether the cerebral hemorrhage is due to primary defects in vascular or neural cell types. We show that ablating alphav expression in the vascular endothelium has no detectable effect on cerebral blood vessel development, whereas deletion of alphav expression in central nervous system glial cells leads to embryonic and neonatal cerebral hemorrhage. Conditional deletion of alphav integrin in both central nervous system glia and neurons also leads to cerebral hemorrhage, but additionally to severe neurological defects. Approximately 30% of these mutants develop seizures and die by 4 weeks of age. The remaining mutants survive for several months, but develop axonal deterioration in the spinal cord and cerebellum, leading to ataxia and loss of hindlimb coordination. Collectively, these data provide evidence that alphav integrins on embryonic central nervous system neural cells, particularly glia, are necessary for proper cerebral blood vessel development, and also reveal a novel function for alphav integrins expressed on axons in the postnatal central nervous system.


Assuntos
Axônios/metabolismo , Sistema Nervoso Central/fisiologia , Hemorragia Cerebral/metabolismo , Integrina alfaV/fisiologia , Animais , Astrócitos/citologia , Comportamento Animal , Encéfalo/embriologia , Cerebelo/metabolismo , Cerebelo/patologia , Circulação Cerebrovascular , Deleção de Genes , Genótipo , Humanos , Imuno-Histoquímica , Integrina alfaV/genética , Integrinas/metabolismo , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Modelos Genéticos , Mutação , Neurônios/metabolismo , Fenótipo , Fatores de Tempo
20.
Genes Chromosomes Cancer ; 37(1): 58-71, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12661006

RESUMO

The transmembrane proto-oncogene receptor tyrosine kinase (RTK) ROS is an orphan receptor that is aberrantly expressed in neoplasms of the central nervous system. Here, we report the fusion of its carboxy-terminal kinase domain to the amino-terminal portion of a protein called FIG (Fused in Glioblastoma) in a human glioblastoma multiforme (GBM). By characterizing both FIG and ROS genes in normal and in U118MG GBM cells, we determined that an intra-chromosomal homozygous deletion of 240 kilobases on 6q21 is responsible for the formation of the FIG-ROS locus. The FIG-ROS transcript is encoded by 7 FIG exons and 9 ROS-derived exons. We also demonstrate that the FIG-ROS locus encodes for an in-frame fusion protein with a constitutively active kinase activity, suggesting that FIG-ROS may act as an oncogene. This is the first example of a fusion RTK protein that results from an intra-chromosomal deletion, and it represents the first fusion RTK protein isolated from a human astrocytoma.


Assuntos
Proteínas de Transporte/genética , Deleção Cromossômica , Cromossomos Humanos Par 6/genética , Glioblastoma/enzimologia , Glioblastoma/genética , Proteínas de Membrana/genética , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Sequência de Bases/genética , Células COS , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Domínio Catalítico/genética , Chlorocebus aethiops , Mapeamento Cromossômico , Proteínas da Matriz do Complexo de Golgi , Humanos , Glicoproteínas de Membrana/genética , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Dados de Sequência Molecular , Peso Molecular , Proteínas de Fusão Oncogênica/química , Biossíntese de Proteínas , Proteínas Tirosina Quinases , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/química , Fases de Leitura/genética , Receptores Proteína Tirosina Quinases/química , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA