Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cell Stress Chaperones ; 29(3): 359-380, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38570009

RESUMO

Protein misfolding and mislocalization are common themes in neurodegenerative disorders, including motor neuron disease, and amyotrophic lateral sclerosis (ALS). Maintaining proteostasis is a crosscutting therapeutic target, including the upregulation of heat shock proteins (HSP) to increase chaperoning capacity. Motor neurons have a high threshold for upregulating stress-inducible HSPA1A, but constitutively express high levels of HSPA8. This study compared the expression of these HSPs in cultured motor neurons expressing three variants linked to familial ALS: TAR DNA binding protein 43 kDa (TDP-43)G348C, fused in sarcoma (FUS)R521G, or superoxide dismutase I (SOD1)G93A. All variants were poor inducers of Hspa1a, and reduced levels of Hspa8 mRNA and protein, indicating multiple compromises in chaperoning capacity. To promote HSP expression, cultures were treated with the putative HSP coinducer, arimoclomol, and class I histone deacetylase inhibitors, to promote active chromatin for transcription, and with the combination. Treatments had variable, often different effects on the expression of Hspa1a and Hspa8, depending on the ALS variant expressed, mRNA distribution (somata and dendrites), and biomarker of toxicity measured (histone acetylation, maintaining nuclear TDP-43 and the neuronal Brm/Brg-associated factor chromatin remodeling complex component Brg1, mitochondrial transport, FUS aggregation). Overall, histone deacetylase inhibition alone was effective on more measures than arimoclomol. As in the FUS model, arimoclomol failed to induce HSPA1A or preserve Hspa8 mRNA in the TDP-43 model, despite preserving nuclear TDP-43 and Brg1, indicating neuroprotective properties other than HSP induction. The data speak to the complexity of drug mechanisms against multiple biomarkers of ALS pathogenesis, as well as to the importance of HSPA8 for neuronal proteostasis in both somata and dendrites.

2.
Front Oncol ; 10: 555945, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072581

RESUMO

Glioblastoma multiforme is an aggressive malignancy, resistant to standard treatment modalities and associated with poor prognosis. We analyzed the role of the IGF system in intracerebral glioma growth using human and rat glioma cells. The glioma cells C6 and U87MG were transduced with a genetically engineered retrovirus expressing type 1 insulin-like growth factor (IGF-IR) antisense RNA, either before or after intra-cerebral implantation of the cells into Sprague Dawley rats or nude mice, respectively and tumor growth and animal survival were monitored. Rat glioma cells transduced prior to orthotopic, intra-cerebral implantation had a significantly increased apoptotic rate in vivo and a significantly reduced tumor volume as seen 24 days post implantation (p < 0.0015). This resulted in increased survival, as greater than 70% of the rats were still alive 182 days after tumor implantation (p < 0.01), as compared to 80% mortality by day 24 in the control group. Histomorphology and histochemical studies performed on brain tissue that was obtained from rats that survived for 182 days revealed numerous single cells that were widely disseminated throughout the brain. These cells expressed the ß-galactosidase marker protein, but were Ki67negative, suggesting that they acquired a dormant phenotype. Direct targeting of the C6 cells with retroviral particles in vivo was effective and reduced tumor volumes by 22% relative to controls. A significant effect on tumor growth was also seen with human glioma U87MG cells that were virally transduced and implanted intra-cerebrally in nude mice. We observed in these mice a significant reduction in tumor volumes and 70% of the animals were still alive 6 months after tumor implantation, as compared to 100% mortality in the control group by day 63. Our results show that IGF-IR targeting can inhibit the intracerebral growth of glioma cells. They also suggest that IGF-IR expression levels may determine a delicate balance between glioma cell growth, death and the acquisition of a dormant state in the brain.

3.
Clin Neurol Neurosurg ; 173: 20-30, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30055402

RESUMO

OBJECTIVE: Glial tumor growth may accelerate during gestation, but epidemiological studies consistently demonstrated that parousity reduces life long risk of glial tumors. Pregnancy may also accelerate growth of medulloblastoma and meningioma, but parousity does not confer protection against these tumors. We were the first to show that medroxyprogesterone acetate (MPA) reduces rat C6 glioma growth in vitro. Now we aimed to determine the effects of MPA on human brain cancers (particularly glioblastoma) in vitro and C6 glioma in vivo. PATIENTS AND METHODS: We evaluated the effects of MPA on: i) monolayer growth of human U87 and U251 glioblastoma, ii) 3D-spheroid growth and invasion of C6 rat glioma and human U251 glioma, iii) interactions with PI3-Kinase inhibitors and coxsackie-adenovirus receptor (CAR) in modifying 3D-spheroid invasion of glioma. RESULTS: MPA at low doses (3.25-13 µM) insignificantly stimulated and at high doses (above 52 µM) strongly suppressed the growth of human U87 and U251 cells in vitro. MPA also binds to glucocorticoid receptors similar to dexamethasone (Dex) and unexpectedly, PI3-Kinase inhibitors at low doses suppressed anti-invasive efficacies of MPA and Dex. MPA exerted higher invasion-inhibitory effects on CAR-expressing human glioma cells. Lastly, MPA suppressed growth of C6 glioma implanted into rat brain. CONCLUSION: Progesterone analogues deserve to be studied in future experimental models of high grade glial brain tumors.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Glioma/tratamento farmacológico , Medroxiprogesterona/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Neoplasias Cerebelares/tratamento farmacológico , Dexametasona/farmacologia , Modelos Animais de Doenças , Glioblastoma/patologia , Glioma/patologia , Humanos , Meningioma/tratamento farmacológico , Ratos , Células Tumorais Cultivadas/efeitos dos fármacos
4.
BMC Res Notes ; 7: 183, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24674422

RESUMO

BACKGROUND: Alterations in cell migration are a hallmark of cancer cell invasion and metastasis. In vitro assays commonly used to study cell migration, including the scratch wound healing assay, Boyden chamber assay, and newly developed advanced systems with microfluidics, each have several disadvantages. FINDINGS: Here we describe an easy and cost-effective in vitro assay for cell migration employing cloning rings to create gaps in the cell monolayer ("ring cell migration assay"). The assay was used to quantitate innate differences in cell motility and the effect of various extracellular matrix proteins on migration of five cancer cell lines: U87 and U251N glioma cells, MDA-MB-231 and MCF-7 breast cancer cells, and HeLa cervical cancer cells. Interestingly, collagen was a general promoter of cell migration for all five cancer cell lines, without affecting cell proliferation. CONCLUSIONS: Taken together, the ring cell migration assay is an easy, convenient and cost-effective assay to study cell migration in vitro.


Assuntos
Ensaios de Migração Celular , Movimento Celular/efeitos dos fármacos , Proteínas da Matriz Extracelular/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas da Matriz Extracelular/química , Humanos
5.
PLoS One ; 8(8): e73296, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24015300

RESUMO

The Coxsackievirus and Adenovirus Receptor (CAR) is a cell adhesion molecule originally characterized as a virus receptor but subsequently shown to be involved in physiological processes such as neuronal and heart development, epithelial tight junction integrity, and tumour suppression. Proteolysis of cell adhesion molecules and a wide variety of other cell surface proteins serves as a mechanism for protein turnover and, in some cases, cell signaling. Metalloproteases such as A Disintegrin and Metalloprotease (ADAM) family members cleave cell surface receptors to release their substrates' ectodomains, while the presenilin/ɣ-secretase complex mediates regulated intramembrane proteolysis (RIP), releasing intracellular domain fragments from the plasma membrane. In the case of some substrates such as Notch and amyloid precursor protein (APP), the released intracellular domains enter the nucleus to modulate gene expression. We report that CAR ectodomain is constitutively shed from glioma cells and developing neurons, and is also shed when cells are treated with the phorbol ester phorbol 12-myristate 13-acetate (PMA) and the calcium ionophore ionomycin. We identified ADAM10 as a sheddase of CAR using assays involving shRNA knockdown and rescue, overexpression of wild-type ADAM10 and inhibition of ADAM10 activity by addition of its prodomain. In vitro peptide cleavage, mass spectrometry and mutagenesis revealed the amino acids M224 to L227 of CAR as the site of ADAM10-mediated ectodomain cleavage. CAR also undergoes RIP by the presenilin/γ-secretase complex, and the intracellular domain of CAR enters the nucleus. Ectodomain shedding is a prerequisite for RIP of CAR. Thus, CAR belongs to the increasing list of cell surface molecules that undergo ectodomain shedding and that are substrates for ɣ-secretase-mediated RIP.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Membrana Celular/metabolismo , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Presenilinas/metabolismo , Proteólise , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/genética , Animais , Carcinógenos/farmacologia , Linhagem Celular Tumoral , Membrana Celular/genética , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/genética , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Mutagênese Sítio-Dirigida , Neurônios/citologia , Neurônios/metabolismo , Presenilinas/genética , Estrutura Terciária de Proteína , Acetato de Tetradecanoilforbol/farmacologia
6.
J Gene Med ; 14(12): 746-60, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23071006

RESUMO

BACKGROUND: Gutless adenovirus (helper-dependent adenoviral vector; HDAd) and lentiviral vectors (LV) are attractive vectors for the gene therapy of muscle diseases. Because the organization of their DNA (episomal versus integrated) differs, we investigated whether the strength and specificity of ΔUSEx3, a novel muscle-specific promoter previously tested with plasmid, were maintained in the context of these vectors. METHODS: Two HDAds expressing ß-galactosidase regulated by ΔUSEx3 or CAG [cytomegalovirus (CMV) enhancer/ß-actin promoter], and three LV expressing green fluorescent protein regulated by ΔUSEx3, CMV or a modified skeletal α-actin promoter (SPcΔ5-12), were constructed. Gene expression was compared in cell culture and after intravenous (HDAd only) and intramuscular injection of mice. RESULTS: Irrespective of the vector used, ΔUSEx3 remained poorly active in nonmuscle cells and tissues. In myotubes, ΔUSEx3 was as strong as CMV and SPcΔ5-12, although it was ten-fold weaker than CAG, a proven powerful promoter in muscle. In cell culture, ΔUSEx3 activity in the context of LV was more stable than CMV, indicating it is less prone to silencing. In the context of HDAd, the behavior of ΔUSEx3 in skeletal muscle mirrored that of cell culture (10% of the CAG activity and half the number of transduced fibers). Surprisingly, in muscles treated with LV, ΔUSEx3 activity was five-fold lower than SPcΔ5-12. CONCLUSIONS: The data obtained in the present study confirm that ΔUSEx3 is a strong and robust muscle-specific promoter in the context of HDAd (cell culture and in vivo) and LV (cell culture). However, it was less efficient in vivo in the context of LV.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Lentivirus/genética , Músculo Esquelético/metabolismo , Regiões Promotoras Genéticas , Troponina I/genética , Animais , Linhagem Celular , Citomegalovirus/genética , Elementos Facilitadores Genéticos , Expressão Gênica , Ordem dos Genes , Humanos , Camundongos , Especificidade de Órgãos/genética , Troponina I/metabolismo
7.
Endocrinology ; 152(12): 4581-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21952243

RESUMO

The protein tyrosine phosphatase (PTPase) Src-homology 2-domain-containing phosphatase (SHP)-1 was recently reported to be a novel regulator of insulin's metabolic action. In order to examine the role of this PTPase in skeletal muscle, we used adenovirus (AdV)-mediated gene transfer to express an interfering mutant of SHP-1 [dominant negative (DN)SHP-1; mutation C453S] in L6 myocytes. Expression of DNSHP-1 increased insulin-induced Akt serine-threonine kinase phosphorylation and augmented glucose uptake and glycogen synthesis. Pharmacological inhibition of glucose transporter type 4 (GLUT4) activity using indinavir and GLUT4 translocation assays revealed an important role for this transporter in the increased insulin-induced glucose uptake in DNSHP-1-expressing myocytes. Both GLUT4 mRNA and protein expression were also found to be increased by DNSHP-1 expression. Furthermore, AdV-mediated delivery of DNSHP-1 in skeletal muscle of transgenic mice overexpressing Coxsackie and AdV receptor also enhanced GLUT4 protein expression. Together, these findings confirm that SHP-1 regulates muscle insulin action in a cell-autonomous manner and further suggest that the PTPase negatively modulates insulin action through down-regulation of both insulin signaling to Akt and GLUT4 translocation, as well as GLUT4 expression.


Assuntos
Glucose/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/antagonistas & inibidores , Receptor de Insulina/metabolismo , Animais , Transportador de Glucose Tipo 4/metabolismo , Glicogênio/biossíntese , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
8.
Behav Brain Funct ; 6: 35, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20594357

RESUMO

BACKGROUND: The Morris water maze task is a hippocampus-dependent learning and memory test that typically takes between 3 days to 2 weeks of training. This task is used to assess spatial learning and induces the expression of genes known to be crucial to learning and memory in the hippocampus. A major caveat in the protocol is the prolonged duration of training, and difficulty of assessing the time during training in which animals have learned the task. We introduce here a condensed version of the task that like traditional water maze tasks, creates lasting hippocampus-dependent spatial cognitive maps and elicits gene expression following learning. METHODS: This paradigm was designed for rats to quickly acquire a hippocampus-dependent spatial cognitive map and retain this memory for at least 24 hours. To accomplish this, we interspersed visible and hidden training trials, delivering them in a massed fashion so training takes a maximum of 15 minutes. Learning was assessed based on latencies to the platform during each training trial, as well as time spent in the goal quadrant during probe testing 30 minutes and 24 hours after training. Normal rats were compared to two impaired cohorts (rats with fimbria-fornix lesions and rats administered NMDA receptor antagonist (CPP)). To quantitate hippocampal expression of known learning genes, real-time polymerase chain reaction (RT-PCR) was performed on hippocampal cDNA. RESULTS: We show that massed training using alternating visible and hidden training trials generates robust short-term working and long-term reference memories in rats. Like the traditional Morris water maze paradigm, this task requires proper hippocampal function, as rats with fimbria-fornix lesions and rats administered CPP fail to learn the spatial component of the task. Furthermore, training in this paradigm elicits hippocampal expression of genes upregulated following learning in a variety of spatial tasks: homer1a, cfos and zif268. CONCLUSIONS: We introduce here a condensed version of the Morris water maze, which is like a traditional water maze paradigm, in that it is hippocampus-dependent, and elicits hippocampal expression of learning genes. However, this task is administered in 15 minutes and induces spatial memory for at least 24 hours.


Assuntos
Genes Precoces , Hipocampo/fisiologia , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Testes Neuropsicológicos , Percepção Espacial/fisiologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Precoces/efeitos dos fármacos , Genes fos , Hipocampo/efeitos dos fármacos , Proteínas de Arcabouço Homer , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Piperazinas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Long-Evans , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Percepção Espacial/efeitos dos fármacos , Fatores de Tempo
9.
Cancer Biol Ther ; 9(11): 843-52, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20372085

RESUMO

Dysregulation of various signaling pathways that govern cerebellar development with respect to cell proliferation, growth arrest, apoptosis and differentiation has been postulated to contribute to medulloblastoma tumourigenesis. This review will highlight the unique nature of cerebellar development in terms of its derivation from two germinal matrices and significant postnatal expansion of the granule cell precursor (GCP) compartment resulting in granule cell development and migration to form the mature cerebellar cortex. The molecular signals that are critical for timely cell cycle exit and differentiation may become dysregulated leading to unrestrained cell proliferation and enhanced cell survival; indeed, changes in these molecular markers have been observed in medulloblastoma biopsy specimens. Furthermore, transgenic models that faithfully replicate these changes develop medulloblastoma that, by in large, recapitulates the clinico-histopathological features of these tumours. Cellular and developmental biological approaches have contributed greatly to the current debate on the relevance of the cancer stem cell hypothesis in understanding medulloblastoma initiation and propagation. Penultimately, research findings are being translated into experimental therapeutics that target the aberrant signal transduction machinery in medulloblastoma cells and that will hopefully lead to an improved risk-benefit profile.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Cerebelares/tratamento farmacológico , Meduloblastoma/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Histona Desacetilases/uso terapêutico , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Modelos Biológicos
10.
J Gene Med ; 12(3): 266-75, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20082422

RESUMO

BACKGROUND: Efficient adenovirus (AdV)-mediated gene transfer is possible only in immature muscle or regenerating muscle, suggesting that a developmentally regulated event plays a major role in limiting AdV uptake in mature skeletal muscle. Previously, we showed that the expression of the primary coxsackie and adenovirus receptor (CAR) is severely down-regulated during muscle maturation and that, in muscle-specific CAR transgenic mice, there is significant enhancement of AdV-mediated gene transfer to mature skeletal muscle. METHODS: To evaluate whether increasing CAR expression can also augment gene transfer to dystrophic muscle that has many regenerating fibers, we crossed CAR transgenics with dystrophin-deficient mice (mdx/CAR). We also tested a two-step protocol in which CAR levels were increased in the target muscle, prior to administration of AdV, through the use of recombinant adeno-associated virus (AAV2) expressing CAR. Lastly, we assessed the effect of histone deacetylase inhibitors on CAR and AdV transduction efficiency in myoblasts and mdx muscle. RESULTS: Although somewhat higher rates of transduction can be achieved in adult mdx mice than in normal mice as a result of ongoing muscle regeneration in these animals, CAR expression in the mdx background (mdx/CAR transgenics) still markedly improved the susceptibility of mature muscle to AdV-mediated gene transfer of dystrophin. Prior administration of AAV2-CAR to normal muscle led to significantly increased transduction by subsequent injection of AdV. The histone deacetylase inhibitor valproate increased CAR transcript and protein levels in myoblasts and mdx muscle, and also increased AdV-mediated gene transfer. CONCLUSIONS: We have developed a method of increasing CAR levels in both normal and regenerating muscle.


Assuntos
Fibras Musculares Esqueléticas/metabolismo , Distrofias Musculares/genética , Receptores Virais/genética , Transdução Genética/métodos , Adenoviridae , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Distrofina/genética , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Regeneração , Transcrição Gênica/efeitos dos fármacos , Ácido Valproico/farmacologia
11.
Int J Oncol ; 35(5): 1045-51, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19787258

RESUMO

Medulloblastoma is the most frequent type of childhood brain tumour. The insulin-like growth factor I receptor (IGF-IR) plays a significant neuroprotective role in medulloblastoma survival through regulation of the downstream effectors of the phosphoinositide-3-kinase-protein kinase-B (PI3K-PKB/c-Akt) pathway. One such target is Forkhead box O1 (FOXO1; FKHR), which is part of the FOXO family of Forkhead transcription factors. Phosphorylation by Akt results in cytoplasmic sequestration of FOXO1 thus inhibiting the expression of genes controlling cell death, cell proliferation, differentiation, cellular metabolism and oxidative stress. Here we show that serum starvation of medulloblastoma cells is accompanied by nuclear translocation of FOXO1. IGF-I stimulation of serum-starved cells resulted in rapid phosphorylation of Akt and FOXO1, and was associated with a significant increase in cell viability. In contrast, expression of a constitutively active form of FOXO1 that cannot be phosphorylated led to a significant reduction in medulloblastoma cell viability, even in the presence of growth factors provided by fetal bovine serum (FBS). These data suggest that the transcription factor FOXO1 may be a critical effector of medulloblastoma growth suppression.


Assuntos
Neoplasias Cerebelares/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Meduloblastoma/metabolismo , Transdução de Sinais/fisiologia , Western Blotting , Linhagem Celular , Sobrevivência Celular/fisiologia , Ativação Enzimática/fisiologia , Imunofluorescência , Proteína Forkhead Box O1 , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Transporte Proteico/fisiologia , Receptor IGF Tipo 1/metabolismo
12.
Cancer Res ; 69(13): 5433-40, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19549905

RESUMO

Gliomas are the most common adult primary brain tumors, and the most malignant form, glioblastoma multiforme, is invariably fatal. The phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway is altered in most glioblastoma multiforme. PTEN, an important negative regulator of the PI3K-Akt pathway, is also commonly mutated in glioma, leading to constitutive activation of Akt. One ultimate consequence is phosphorylation and inactivation of FOXO forkhead transcription factors that regulate genes involved in apoptosis, cell cycle arrest, nutrient availability, DNA repair, stress, and angiogenesis. We tested the ability of a mutant FOXO1 factor that is not subject to Akt phosphorylation to overcome dysregulated PI3K-Akt signaling in two PTEN-null glioma cell lines, U87 and U251. Adenovirus-mediated gene transfer of the mutant FOXO1 successfully restored cell cycle arrest and induced cell death in vitro and prolonged survival in vivo in xenograft models of human glioma (33% survival at 1 year of animals bearing U251 tumors). However, U87 were much more resistant than U251 to mutant FOXO1-induced death, showing evidence of increased nuclear export and Akt-independent phosphorylation of FOXO1 at S249. A cyclin-dependent kinase 2 inhibitor decreased phosphorylation of S249 and rendered U87 cells significantly more susceptible to mutant FOXO1-induced death. Our results indicate that targeting FOXO1, which is at the convergence point of several growth factor receptor tyrosine kinase pathways, can effectively induce glioma cell death and inhibit tumor growth. They also highlight the importance of Akt-independent phosphorylation events in the nuclear export of FOXO1.


Assuntos
Neoplasias Encefálicas/patologia , Fatores de Transcrição Forkhead/efeitos dos fármacos , Glioma/patologia , Adenoviridae/genética , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Ciclina D1/fisiologia , Ciclina D2 , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Ciclinas/fisiologia , Reparo do DNA , Modelos Animais de Doenças , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Neovascularização Patológica/prevenção & controle , Fosforilação
13.
J Biol Chem ; 283(50): 34720-7, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18945675

RESUMO

Duchenne muscular dystrophy is caused by a genetic defect in the dystrophin gene. The absence of dystrophin results in muscle fiber necrosis and regeneration, leading to progressive muscle fiber loss. Utrophin is a close analogue of dystrophin. A substantial, ectopic expression of utrophin in the extrasynaptic sarcolemma of dystrophin-deficient muscle fibers can prevent deleterious effects of dystrophin deficiency. An alternative approach for the extrasynaptic up-regulation of utrophin involves the augmentation of utrophin transcription via the endogenous utrophin A promoter using custom-designed transcriptional activator proteins with zinc finger (ZFP) motifs. We tested a panel of custom-designed ZFP for their ability to activate the utrophin A promoter. Expression of one such ZFP efficiently increased, in a time-dependent manner, utrophin transcript and protein levels both in vitro and in vivo. In dystrophic mouse (mdx) muscles, administration of adenoviral vectors expressing this ZFP led to significant enhancement of muscle function with decreased necrosis, restoration of the dystrophin-associated proteins, and improved resistance to eccentric contractions. These studies provide evidence that specifically designed ZFPs can act as strong transcriptional activators of the utrophin A promoter. These may thus serve as attractive therapeutic agents for dystrophin deficiency states such as Duchenne muscular dystrophy.


Assuntos
Regulação da Expressão Gênica , Músculos/patologia , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Regulação para Cima , Utrofina/genética , Animais , Humanos , Camundongos , Camundongos Transgênicos , Contração Muscular , Músculos/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Necrose , Fatores de Tempo
14.
Cytometry A ; 73(10): 940-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18773455

RESUMO

Side population (SP) analyses and CD133 expression have identified cells with stem-like potential in normal and cancerous tissue. Whether stem-like cells exist in cancer cell lines is hotly debated. We have interrogated the DAOY medulloblastoma cell line with respect to stem-like potential. Vital staining for Hoechst 33342 efflux capacity and CD133 immunophenotyping were performed on DAOY cells to assess the presence of the SP and the CD133 stem cell markers, respectively. SP/non-SP and CD133(+)/CD133(-) DAOY cells were sorted into separate fractions for limiting dilution analysis (tumor sphere assay) and asymmetric division assessment. SP/non-SP cells were also sorted separately for viability (XTT assay), cell size, cell cycle status, and proliferative capacity (carboxyfluorescein succinimidyl ester (CFSE)) evaluation. A minor proportion of cells displayed either the SP or the CD133(+) phenotypes. CD133 expression mapped to both the SP and non-SP compartments, with CD133(+) cells being enriched almost fourfold within the non-SP gate. The SP, non-SP, CD133(+), and CD133(-) fractions were all capable of reconstituting the original parental DAOY population. Slight clonogenic enrichment was observed in only the SP fraction; however, both CD133(+) and CD133(-) cells displayed equivalent stem cell-like frequencies. SP cells were resistant to Hoechst 33342-mediated toxicity relative to the parental population and differed from the non-SP cells with respect to increased cell size, decreased S-phase, and slightly decreased proliferative capacity. The multiparametric strategy described in this study revealed that the SP and CD133(+) subset may be two independent compartments. Our results highlight the need for new reliable specific cancer stem cell marker(s) as Hoechst 33342 efflux and CD133 expression might not be suitable for selectively isolating cancer stem-like cells from cell lines, as shown for the DAOY cells. As such, care must be used in interpreting therapeutic studies targeting the stem cell compartment of cancer cell lines.


Assuntos
Antígenos CD/análise , Biomarcadores Tumorais/análise , Citometria de Fluxo , Glicoproteínas/análise , Meduloblastoma/patologia , Células-Tronco Neoplásicas/patologia , Peptídeos/análise , Antígeno AC133 , Antígenos CD/biossíntese , Benzimidazóis/análise , Biomarcadores Tumorais/biossíntese , Linhagem Celular Tumoral , Separação Celular/métodos , Citometria de Fluxo/métodos , Fluoresceínas/metabolismo , Glicoproteínas/biossíntese , Humanos , Meduloblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Succinimidas/metabolismo
15.
Hum Gene Ther ; 19(2): 133-42, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18067405

RESUMO

Adenoviral vectors that use the coxsackievirus and adenovirus receptor do not transduce mature muscle efficiently. Group B adenoviruses use CD46 as their cell attachment receptor. To evaluate the utility of vectors based on group B adenoviruses for gene transfer to human skeletal muscle, we assessed the expression of CD46 in biopsied normal skeletal muscle samples and in muscles from patients with Duchenne muscular dystrophy. Transcript levels of CD46 were extremely low in mature muscle and CD46 immunoreactivity was detected only on blood vessels in the muscle sections. Although myoblasts cultured from biopsied samples had robust cell surface CD46 expression by flow cytometry, CD46 transcript levels were barely detectable after differentiation of the myoblasts into myotubes. The myotubes were also much less susceptible to infection with an adenoviral vector carrying the fiber of serotype 35 adenovirus (AdF35). These results suggest that for skeletal muscle, vectors derived from group B adenoviruses may not be a suitable alternative to the commonly used Ad5 vectors.


Assuntos
Adenoviridae/metabolismo , Diferenciação Celular , Regulação para Baixo/genética , Proteína Cofatora de Membrana/genética , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Transdução Genética/métodos , Biópsia , Permeabilidade da Membrana Celular , Células Cultivadas , Citometria de Fluxo , Humanos , Fibras Musculares Esqueléticas , Distrofia Muscular de Duchenne , Mioblastos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta-Galactosidase
16.
Mol Ther ; 15(10): 1767-74, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17667948

RESUMO

Helper-dependent adenovirus vector (AdV)-mediated full-length dystrophin expression leads to significant mitigation of the dystrophic phenotype of the mdx mouse. However, dystrophin, as a neoantigen, elicits antibody formation. As an alternative approach, we evaluated gene transfer of full-length murine utrophin, a functional homologue of dystrophin that is normally present only at the neuromuscular junction. A single injection in the tibialis anterior (TA) muscle of the helper-dependent adenovirus vector encoding utrophin provided very good transduction, with 58% of fibers demonstrating sarcolemmal utrophin expression in the neonates, and 35% utrophin-positive (Utr(+)) fibers in adults. The presence of utrophin prevented extensive necrosis in the neonates, halted further necrosis in the adults, and led to restoration of sarcolemmal expression of dystrophin-associated proteins up to 1 year after injection. Marked physiological improvement was observed in both neonates and adults. Neither increased humoral responses nor cellular immune responses were evident. However, there was a time-related decline of the initial high utrophin expression. Although viral DNA persisted in animals that were injected in the neonatal stage, viral DNA levels decreased in muscles of adult mice. These results demonstrate that although utrophin gene transfer leads to amelioration of the dystrophic phenotype, the effects are not sustained upon loss of utrophin expression.


Assuntos
Adenoviridae/genética , Distrofina/genética , Utrofina/genética , Animais , Animais Recém-Nascidos , Formação de Anticorpos , DNA Viral/metabolismo , Imunidade Celular , Camundongos , Camundongos Mutantes , Músculo Esquelético/metabolismo , Reação em Cadeia da Polimerase , Transdução Genética , Utrofina/administração & dosagem , Utrofina/imunologia
17.
Cancer Res ; 67(14): 6700-7, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17638880

RESUMO

In solid tumors, cancer cells subjected to ischemic conditions trigger distinct signaling pathways contributing to angiogenic stimulation and tumor development. Characteristic features of tumor ischemia include hypoxia and glucose deprivation, leading to the activation of hypoxia-inducible factor-1-dependent signaling pathways and to complex signaling events known as the unfolded protein response. Here, we show that the activation of the endoplasmic reticulum stress sensor IRE1 is a common determinant linking hypoxia- and hypoglycemia-dependent responses to the up-regulation of vascular endothelial growth factor-A (VEGF-A). Tumor cells expressing a dominant-negative IRE1 transgene as well as Ire1alpha-null mouse embryonic fibroblasts were unable to trigger VEGF-A up-regulation upon either oxygen or glucose deprivation. These data correlated with a reduction of tumor angiogenesis and growth in vivo. Our results therefore suggest an essential role for IRE1-dependent signaling pathways in response to ischemia and identify this protein as a potential therapeutic target to control both the angiogenic switch and tumor development.


Assuntos
Endorribonucleases/fisiologia , Regulação Neoplásica da Expressão Gênica , Isquemia , Proteínas de Membrana/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Fibroblastos/metabolismo , Genes Dominantes , Humanos , Camundongos , Neovascularização Patológica , Oxigênio/metabolismo , Transdução de Sinais
18.
Cancer Res ; 67(10): 4541-4, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17510377

RESUMO

Although relatively rare, malignant glioma (MG) is frequently used for testing novel cancer treatments. However, human MG trials have often been initiated on the basis of preclinical models that involve numerous discontinuities with the human disease. Below, we discuss various limitations of the mainstay model used in MG preclinical research, the murine orthotopic xenograft. After discussing alternative model systems like transgenic mouse models and canine xenografts, we argue that companion animals with spontaneous brain cancers offer a scientifically and ethically attractive system for preclinical testing of novel MG interventions. Ethical advantages and practical challenges of companion animal brain cancer trials are briefly discussed.


Assuntos
Animais Domésticos , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/veterinária , Ensaios Clínicos como Assunto/veterinária , Modelos Animais de Doenças , Glioma/terapia , Glioma/veterinária , Bem-Estar do Animal/ética , Animais , Ensaios Clínicos como Assunto/ética , Ensaios Clínicos como Assunto/métodos , Cães , Humanos , Camundongos , Camundongos Transgênicos , Ensaios Antitumorais Modelo de Xenoenxerto
19.
FEBS Lett ; 581(14): 2702-8, 2007 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-17531226

RESUMO

The Coxsackie and adenovirus receptor (CAR) is a cell adhesion molecule that is highly expressed in the developing brain. CAR is enriched in growth cone particles (GCP) after subcellular fractionation. In GCP, we identified actin as an interaction partner of the cytoplasmic domain of CAR. In vivo, actin and CAR co-immunoprecipitate and co-localize. In vitro, the binding is direct, with a K(d) of approximately 2.6 microM, and leads to actin bundling. We previously demonstrated that CAR interacts with microtubules. These data suggest a role for CAR in processes requiring dynamic reorganization of the cytoskeleton such as neurite outgrowth and cell migration.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Receptores Virais/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Cones de Crescimento/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Imunoprecipitação , Cinética , Camundongos , Microscopia Eletrônica , Microscopia de Fluorescência , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/metabolismo , Ligação Proteica , Receptores Virais/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura
20.
J Virol Methods ; 144(1-2): 32-40, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17467815

RESUMO

Recombinant adeno-associated virus (rAAV) has emerged in recent years as a promising gene therapy vector that may be used in the treatment of diverse human diseases. The major obstacle to broadening the usage of rAAV vectors remains the limited capacity of available production systems to provide sufficient rAAV quantities for preclinical and clinical trials. The impracticality of expanding commonly used adherent cell lines represents a limitation to large-scale production. This paper describes successful productions of rAAV type 2 using suspension-growing human embryonic kidney (HEK293) cells in serum-free medium. The developed process, based on triple transfection employing polyethylenimine (PEI) as DNA transporter, allowed for a serum-free production of AAV, yielding viral vector titer up to 4.5x10(11) infectious viral particles (IVP) in a 3.5-L bioreactor. A maximum ratio of VG:IVP in the order of 200:1 was obtained, indicating the efficient encapsidation of viral vectors in HEK293 cells. The effect of varying the ratio of three plasmids and the influence of cell density at transfection were studied. The conditioned medium did not limit or inhibit the rAAV production; therefore, the elimination of the medium exchange step before or after transfection greatly simplified the scale-up of rAAV production. The cell-specific viral titers obtained in bioreactor suspension cultures were similar or higher than those obtained with control adherent cell cultures which further supported the scalability of the process. From multiple aspects including process simplicity, scalability, and low operating costs, this transfection method appears to be the most promising technology for large-scale production of rAAV.


Assuntos
Dependovirus/crescimento & desenvolvimento , Transfecção , Cultura de Vírus/métodos , Reatores Biológicos , Técnicas de Cultura de Células , Linhagem Celular , Dependovirus/genética , Vetores Genéticos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA