Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Cell ; 163(1): 39-53, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26406370

RESUMO

Significant advances have been made in developing novel therapeutics for cancer treatment, and targeted therapies have revolutionized the treatment of some cancers. Despite the promise, only about five percent of new cancer drugs are approved, and most fail due to lack of efficacy. The indication is that current preclinical methods are limited in predicting successful outcomes. Such failure exacts enormous cost, both financial and in the quality of human life. This Primer explores the current status, promise, and challenges of preclinical evaluation in advanced mouse cancer models and briefly addresses emerging models for early-stage preclinical development.


Assuntos
Modelos Animais de Doenças , Camundongos , Neoplasias/tratamento farmacológico , Animais , Engenharia Genética , Xenoenxertos , Transplante de Neoplasias
2.
Cell ; 159(1): 80-93, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25259922

RESUMO

The poor clinical outcome in pancreatic ductal adenocarcinoma (PDA) is attributed to intrinsic chemoresistance and a growth-permissive tumor microenvironment. Conversion of quiescent to activated pancreatic stellate cells (PSCs) drives the severe stromal reaction that characterizes PDA. Here, we reveal that the vitamin D receptor (VDR) is expressed in stroma from human pancreatic tumors and that treatment with the VDR ligand calcipotriol markedly reduced markers of inflammation and fibrosis in pancreatitis and human tumor stroma. We show that VDR acts as a master transcriptional regulator of PSCs to reprise the quiescent state, resulting in induced stromal remodeling, increased intratumoral gemcitabine, reduced tumor volume, and a 57% increase in survival compared to chemotherapy alone. This work describes a molecular strategy through which transcriptional reprogramming of tumor stroma enables chemotherapeutic response and suggests vitamin D priming as an adjunct in PDA therapy. PAPERFLICK:


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Calcitriol/análogos & derivados , Carcinoma Ductal Pancreático/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Receptores de Calcitriol/metabolismo , Adenocarcinoma/patologia , Animais , Calcitriol/farmacologia , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neoplasias Pancreáticas/patologia , Pancreatite/tratamento farmacológico , Pancreatite/prevenção & controle , Transdução de Sinais , Células Estromais/patologia
3.
Cell ; 135(3): 408-10, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18984150

RESUMO

The tumor microenvironment is known to play an important role in tumorigenesis. In this issue, Yang et al. (2008) demonstrate that mast cells heterozygous for the Nf1 gene promote the growth of neurofibromas in a mouse model of neurofibromatosis and that genetic and pharmacological inhibition of these cells is sufficient to block tumor growth.


Assuntos
Mastócitos/metabolismo , Neurofibroma/metabolismo , Animais , Humanos , Camundongos , Neurofibroma/tratamento farmacológico , Neurofibroma/genética , Neurofibroma/patologia , Neurofibromina 1/genética , Neurofibromina 1/metabolismo
4.
Biol Reprod ; 102(5): 1055-1064, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-31930396

RESUMO

Ovarian cancer (OvCa) remains the most common cause of death from gynecological malignancies. Genetically engineered mouse models have been used to study initiation, origin, progression, and/or mechanisms of OvCa. Based on the clinical features of OvCa, we examined a quadruple combination of pathway perturbations including PTEN, TRP53, RB1, and/or CDH1. To characterize the cancer-promoting events in the ovarian surface epithelium (OSE), Amhr2cre/+ mice were used to ablate floxed alleles of Pten, Trp53, and Cdh1, which were crossed with TgK19GT121 mice to inactivate RB1 in KRT19-expressing cells. Inactivation of PTEN, TRP53, and RB1 with or without CDH1 led to the development of type I low-grade OvCa with enlarged serous papillary carcinomas and some high-grade serous carcinomas (HGSCs) in older mice. Initiation of epithelial hyperplasia and micropapillary carcinoma started earlier at 1 month in the triple mutations of Trp53, Pten, and Rb1 mice as compared to 2 months in quadruple mutations of Trp53, Pten, Rb1, and Cdh1 mice, whereas both genotypes eventually developed enlarged proliferating tumors that invaded into the ovary at 3-4 months. Mice with triple and quadruple mutations developed HGSC and/or metastatic tumors, which disseminated into the peritoneal cavity at 4-6 months. In summary, inactivation of PTEN, TRP53, and RB1 initiates OvCa from the OSE. Additional ablation of CDH1 further increased persistence of tumor dissemination and ascites fluid accumulation enhancing peritoneal metastasis.


Assuntos
Caderinas/metabolismo , Neoplasias Ovarianas/patologia , Ovário/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteínas de Ligação a Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Caderinas/genética , Transformação Celular Neoplásica , Epitélio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Mutação , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/secundário , PTEN Fosfo-Hidrolase/genética , Proteínas de Ligação a Retinoblastoma/genética , Proteína Supressora de Tumor p53/genética
5.
Proc Natl Acad Sci U S A ; 110(44): 17933-8, 2013 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-24114272

RESUMO

Glioblastoma (GBM), the most common brain malignancy, remains fatal with no effective treatment. Analyses of common aberrations in GBM suggest major regulatory pathways associated with disease etiology. However, 90% of GBMs are diagnosed at an advanced stage (primary GBMs), providing no access to early disease stages for assessing disease progression events. As such, both understanding of disease mechanisms and the development of biomarkers and therapeutics for effective disease management are limited. Here, we describe an adult-inducible astrocyte-specific system in genetically engineered mice that queries causation in disease evolution of regulatory networks perturbed in human GBM. Events yielding disease, both engineered and spontaneous, indicate ordered grade-specific perturbations that yield high-grade astrocytomas (anaplastic astrocytomas and GBMs). Impaired retinoblastoma protein RB tumor suppression yields grade II histopathology. Additional activation of v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) network drives progression to grade III disease, and further inactivation of phosphatase and tensin homolog (PTEN) yields GBM. Spontaneous missense mutation of tumor suppressor Trp53 arises subsequent to KRAS activation, but before grade III progression. The stochastic appearance of mutations identical to those observed in humans, particularly the same spectrum of p53 amino acid changes, supports the validity of engineered lesions and the ensuing interpretations of etiology. Absence of isocitrate dehydrogenase 1 (IDH1) mutation, asymptomatic low grade disease, and rapid emergence of GBM combined with a mesenchymal transcriptome signature reflect characteristics of primary GBM and provide insight into causal relationships.


Assuntos
Astrocitoma/etiologia , Evolução Biológica , Modelos Animais de Doenças , Engenharia Genética/métodos , Glioblastoma/etiologia , Animais , Sequência de Bases , Progressão da Doença , Perfilação da Expressão Gênica , Redes Reguladoras de Genes/genética , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Dados de Sequência Molecular , Mutação de Sentido Incorreto/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Análise de Sequência de DNA , Proteína Supressora de Tumor p53/genética
6.
J Biol Chem ; 289(36): 25227-40, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-24990947

RESUMO

The serine/threonine protein kinase Akt promotes cell survival, growth, and proliferation through phosphorylation of different downstream substrates. A key effector of Akt is the mammalian target of rapamycin (mTOR). Akt is known to stimulate mTORC1 activity through phosphorylation of tuberous sclerosis complex 2 (TSC2) and PRAS40, both negative regulators of mTOR activity. We previously reported that IκB kinase α (IKKα), a component of the kinase complex that leads to NF-κB activation, plays an important role in promoting mTORC1 activity downstream of activated Akt. Here, we demonstrate IKKα-dependent regulation of mTORC1 using multiple PTEN null cancer cell lines and an animal model with deletion of IKKα. Importantly, IKKα is shown to phosphorylate mTOR at serine 1415 in a manner dependent on Akt to promote mTORC1 activity. These results demonstrate that IKKα is an effector of Akt in promoting mTORC1 activity.


Assuntos
Quinase I-kappa B/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Ativação Enzimática , Feminino , Células HEK293 , Células HeLa , Humanos , Quinase I-kappa B/genética , Immunoblotting , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Knockout , Complexos Multiproteicos/genética , Mutação , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/genética , Interferência de RNA , Proteína Regulatória Associada a mTOR , Serina/genética , Serina/metabolismo , Serina-Treonina Quinases TOR/genética , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
7.
Dev Biol ; 371(1): 77-85, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22939930

RESUMO

Aurora A is a mitotic kinase essential for cell proliferation. In mice, ablation of Aurora A results in mitotic arrest and pre-implantation lethality, preventing studies at later stages of development. Here we report the effects of Aurora A ablation on embryo patterning at early post-implantation stages. Inactivation of Aurora A in the epiblast or visceral endoderm layers of the conceptus leads to apoptosis and inhibition of embryo growth, causing lethality and resorption at approximately E9.5. The effects on embryo patterning, however, depend on the tissue affected by the mutation. Embryos with an epiblast ablation of Aurora A properly establish the anteroposterior axis but fail to progress through gastrulation. In contrast, mutation of Aurora A in the visceral endoderm, leads to posteriorization of the conceptus or failure to elongate the anteroposterior axis. Injection of ES cells into Aurora A epiblast knockout blastocysts reconstitutes embryonic development to E9.5, indicating that the extra-embryonic tissues in these mutant embryos can sustain development to organogenesis stages. Our results reveal new ways to induce apoptosis and to ablate cells in a tissue-specific manner in vivo. Moreover, they show that epiblast-ablated embryos can be used to test the potency of stem cells.


Assuntos
Padronização Corporal/genética , Embrião de Mamíferos/embriologia , Endoderma/embriologia , Camadas Germinativas/embriologia , Proteínas Serina-Treonina Quinases/deficiência , Animais , Apoptose/genética , Aurora Quinase A , Aurora Quinases , Primers do DNA/genética , Células-Tronco Embrionárias/metabolismo , Imunofluorescência , Técnicas de Inativação de Genes , Hibridização In Situ , Camundongos , Proteínas Serina-Treonina Quinases/genética , beta-Galactosidase
8.
Cancers (Basel) ; 15(18)2023 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-37760565

RESUMO

MTBP is implicated in cell cycle progression, DNA replication, and cancer metastasis. However, the function of MTBP remains enigmatic and is dependent on cellular contexts and its cellular localization. To understand the in vivo physiological role of MTBP, it is important to generate Mtbp knockout mice. However, complete deletion of the Mtbp gene in mice results in early embryonic lethality, while its heterozygous deletion shows modest biological phenotypes, including enhanced cancer metastasis. To overcome this and better characterize the in vivo physiological function of MTBP, we, for the first time, generated mice that carry an Mtbp hypomorphic allele (MtbpH) in which Mtbp protein is expressed at approximately 30% of that in the wild-type allele. We treated wild-type, Mtbp+/-, and MtbpH/- mice with a liver carcinogen, diethylnitrosamine (DEN), and found that the MtbpH/- mice showed worse overall survival when compared to the wild-type mice. Consistent with previous reports using human liver cancer cells, mouse embryonic fibroblasts (MEFs) from the MtbpH/- mice showed an increase in the nuclear localization of p-Erk1/2 and migratory potential. Thus, MtbpH/- mice and cells from MtbpH/- mice are valuable to understand the in vivo physiological role of Mtbp and validate the diverse functions of MTBP that have been observed in human cells.

9.
Cancer Cell ; 1(2): 157-68, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12086874

RESUMO

We have inactivated pRb, p107, and p130 in astrocytes by transgenic expression of T(121) (a truncated SV40 T antigen) under the GFAP promoter. Founder mice died perinatally with extensive expansion of neural precursor and anaplastic astrocyte populations. In astrocytes, aberrant proliferation and extensive apoptosis were induced. Using a conditional allele of T(121), early lethality was circumvented, and adult mice developed high-grade astrocytoma, in which regions of decreased apoptosis expressed activated Akt. Indeed, astrocytoma development was accelerated in a PTEN(+/-), but not p53(+/-), background. These studies establish a highly penetrant preclinical model for astrocytoma based on events observed in the human disease and further provide insight into the role of PTEN mutation in astrocytoma progression.


Assuntos
Astrócitos/metabolismo , Astrocitoma/metabolismo , Astrocitoma/patologia , Predisposição Genética para Doença , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Serina-Treonina Quinases , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Envelhecimento , Animais , Apoptose , Astrócitos/patologia , Astrocitoma/genética , Encéfalo/anormalidades , Encéfalo/patologia , Divisão Celular , Ativação Enzimática , Heterozigoto , Camundongos , Camundongos Transgênicos , Mutação/genética , Especificidade de Órgãos , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética
10.
Front Oncol ; 12: 904479, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35814428

RESUMO

Glioblastoma (GBM) remains lethal with no effective treatments. Despite the comprehensive identification of commonly perturbed molecular pathways, little is known about the disease's etiology, particularly in early stages. Several studies indicate that GBM is initiated in neural progenitor and/or stem cells. Here, we report that differentiated astrocytes are susceptible to GBM development when initiated by perturbation of the RB pathway, which induces a progenitor phenotype. In vitro and in vivo inactivation of Rb tumor suppression (TS) induces cortical astrocytes to proliferate rapidly, express progenitor markers, repress differentiation markers, and form self-renewing neurospheres that are susceptible to multi-lineage differentiation. This phenotype is sufficient to cause grade II astrocytomas which stochastically progress to GBM. Together with previous findings, these results demonstrate that cell susceptibility to GBM depends on the initiating driver.

11.
Mol Cancer Res ; 19(8): 1422-1436, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33888600

RESUMO

Only a subset of patients responds to immune checkpoint blockade (ICB) in melanoma. A preclinical model recapitulating the clinical activity of ICB would provide a valuable platform for mechanistic studies. We used melanoma tumors arising from an Hgftg;Cdk4R24C/R24C genetically engineered mouse (GEM) model to evaluate the efficacy of an anti-mouse PD-L1 antibody similar to the anti-human PD-L1 antibodies durvalumab and atezolizumab. Consistent with clinical observations for ICB in melanoma, anti-PD-L1 treatment elicited complete and durable response in a subset of melanoma-bearing mice. We also observed tumor growth delay or regression followed by recurrence. For early treatment assessment, we analyzed gene expression profiles, T-cell infiltration, and T-cell receptor (TCR) signatures in regressing tumors compared with tumors exhibiting no response to anti-PD-L1 treatment. We found that CD8+ T-cell tumor infiltration corresponded to response to treatment, and that anti-PD-L1 gene signature response indicated an increase in antigen processing and presentation, cytokine-cytokine receptor interaction, and natural killer cell-mediated cytotoxicity. TCR sequence data suggest that an anti-PD-L1-mediated melanoma regression response requires not only an expansion of the TCR repertoire that is unique to individual mice, but also tumor access to the appropriate TCRs. Thus, this melanoma model recapitulated the variable response to ICB observed in patients and exhibited biomarkers that differentiate between early response and resistance to treatment, providing a valuable platform for prediction of successful immunotherapy. IMPLICATIONS: Our melanoma model recapitulates the variable response to anti-PD-L1 observed in patients and exhibits biomarkers that characterize early antibody response, including expansion of the TCR repertoire.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Biomarcadores/metabolismo , Melanoma/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/metabolismo , Modelos Animais de Doenças , Melanoma/tratamento farmacológico , Camundongos
12.
Angiogenesis ; 13(1): 59-69, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20221685

RESUMO

Von Hippel-Lindau (VHL) inactivation develops a tumor syndrome characterized by highly vascularized tumors as a result of hypoxia inducible factors (HIF) stabilization. The most common manifestation is the development of hemangioblastomas typically located in the central nervous system and other organs including the liver. PTEN (Phosphatase and tension homologue deleted on chromosome 10) inactivation also upregulates HIF-1alpha and may take part in promoting vascular lesions in tumors. The coordinate effect of loss of these tumor suppressors on HIF levels, and the subsequent effect on vascular lesion formation would elucidate the potential for mechanisms to modify HIF dosage supplementally and impact tumor phenotype. We therefore employed models of somatic conditional inactivation of Vhl, Pten, or both tumor suppressor genes in individual cells of the liver by Cre-loxP recombination to study the cooperativity of these two tumor suppressors in preventing tumor formation. Nine months after tumor suppressor inactivation, Vhl conditional deletion (Vhl (loxP/loxP)) mice showed no abnormalities, Pten conditional deletion (Pten (loxP/loxP)) mice developed liver steatosis and focal nodular expansion of hepatocytes containing lipid droplet and fat. Vhl and Pten conditional deletion (Vhl (loxP/loxP);Pten (loxP/loxP)) mice, however, developed multiple cavernous liver lesions reminiscent of hemangioblastoma. Liver hemangioblastomas in VHL disease may, therefore, require secondary mutation in addition to VHL loss of heterozygosity which is permissive for vascular lesion development or augments levels of HIF-1alpha.


Assuntos
Fígado/irrigação sanguínea , Fígado/patologia , PTEN Fosfo-Hidrolase/metabolismo , Doenças Vasculares/enzimologia , Doenças Vasculares/patologia , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Animais , Proliferação de Células , Dependovirus , Deleção de Genes , Técnicas de Transferência de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Injeções Intravenosas , Metabolismo dos Lipídeos , Fígado/enzimologia , Camundongos , PTEN Fosfo-Hidrolase/deficiência , Fosforilação , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Recombinação Genética , Regulação para Cima/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Proc Natl Acad Sci U S A ; 104(52): 20820-5, 2007 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18093951

RESUMO

Brain tumors are typically resistant to conventional chemotherapeutics, most of which initiate apoptosis upstream of mitochondrial cytochrome c release. In this study, we demonstrate that directly activating apoptosis downstream of the mitochondria, with cytosolic cytochrome c, kills brain tumor cells but not normal brain tissue. Specifically, cytosolic cytochrome c is sufficient to induce apoptosis in glioblastoma and medulloblastoma cell lines. In contrast, primary neurons from the cerebellum and cortex are remarkably resistant to cytosolic cytochrome c. Importantly, tumor tissue from mouse models of both high-grade astrocytoma and medulloblastoma display hypersensitivity to cytochrome c when compared with surrounding brain tissue. This differential sensitivity to cytochrome c is attributed to high Apaf-1 levels in the tumor tissue compared with low Apaf-1 levels in the adjacent brain tissue. These differences in Apaf-1 abundance correlate with differences in the levels of E2F1, a previously identified activator of Apaf-1 transcription. ChIP assays reveal that E2F1 binds the Apaf-1 promoter specifically in tumor tissue, suggesting that E2F1 contributes to the expression of Apaf-1 in brain tumors. Together, these results demonstrate an unexpected sensitivity of brain tumors to postmitochondrial induction of apoptosis. Moreover, they raise the possibility that this phenomenon could be exploited therapeutically to selectively kill brain cancer cells while sparing the surrounding brain parenchyma.


Assuntos
Fator Apoptótico 1 Ativador de Proteases/metabolismo , Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Citocromos c/metabolismo , Regulação Neoplásica da Expressão Gênica , Apoptose , Astrocitoma/metabolismo , Caspases/metabolismo , Citocromos c/química , Fator de Transcrição E2F1/química , Humanos , Meduloblastoma/metabolismo , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Transcrição Gênica
14.
Nat Med ; 26(5): 781-791, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32284588

RESUMO

Although immunotherapy has revolutionized cancer treatment, only a subset of patients demonstrate durable clinical benefit. Definitive predictive biomarkers and targets to overcome resistance remain unidentified, underscoring the urgency to develop reliable immunocompetent models for mechanistic assessment. Here we characterize a panel of syngeneic mouse models, representing a variety of molecular and phenotypic subtypes of human melanomas and exhibiting their diverse range of responses to immune checkpoint blockade (ICB). Comparative analysis of genomic, transcriptomic and tumor-infiltrating immune cell profiles demonstrated alignment with clinical observations and validated the correlation of T cell dysfunction and exclusion programs with resistance. Notably, genome-wide expression analysis uncovered a melanocytic plasticity signature predictive of patient outcome in response to ICB, suggesting that the multipotency and differentiation status of melanoma can determine ICB benefit. Our comparative preclinical platform recapitulates melanoma clinical behavior and can be employed to identify mechanisms and treatment strategies to improve patient care.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais , Imunoterapia , Melanoma/patologia , Melanoma/terapia , Animais , Antineoplásicos Imunológicos/uso terapêutico , Antígeno CTLA-4/imunologia , Células Cultivadas , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Heterogeneidade Genética , Humanos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Ipilimumab/uso terapêutico , Melanoma/diagnóstico , Melanoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Prognóstico , Receptor de Morte Celular Programada 1/imunologia , RNA-Seq , Resultado do Tratamento , Sequenciamento Completo do Genoma
15.
Mol Carcinog ; 48(12): 1139-48, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19676100

RESUMO

Malignant rhabdoid tumors (MRTs) are poorly differentiated pediatric cancers that arise in various anatomical locations and have a very poor outcome. The large majority of these malignancies are caused by loss of function of the SNF5/INI1 component of the SWI/SNF chromatin remodeling complex. However, the mechanism of tumor development associated with SNF5 loss remains unclear. Multiple studies have demonstrated a role for SNF5 in the regulation of cyclin D1, p16(INK4A), and pRb(f) activities suggesting it functions through the SWI/SNF complex to affect transcription of genes involved in cell cycle control. Previous studies in genetically engineered mouse models (GEMM) have shown that loss of SNF5 on a p53-null background significantly accelerates tumor development. Here, we use established GEMM to further define the relationship between the SNF5 and p53 tumor suppressor pathways. Combined haploinsufficiency of p53 and Snf5 leads to decreased latency for MRTs arising in alternate anatomical locations but not for the standard facial MRTs. We also observed acceleration in the appearance of T-cell lymphomas in the p53(+/-);Snf5(+/-) mice. Our studies suggest that loss of SNF5 activity does not bestow a selective advantage on the p53 spectrum of tumors in the p53(+/-);Snf5(+/-) mice. However, reduced p53 expression specifically accelerated the growth of a subset of MRTs in these mice.


Assuntos
Neoplasias Ósseas/patologia , Proteínas Cromossômicas não Histona/fisiologia , Linfoma de Células T/patologia , Osteossarcoma/patologia , Tumor Rabdoide/patologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Neoplasias Ósseas/genética , Proteínas de Ciclo Celular/metabolismo , Feminino , Linfoma de Células T/genética , Masculino , Camundongos , Camundongos Knockout , Osteossarcoma/genética , Tumor Rabdoide/genética , Proteína SMARCB1 , Taxa de Sobrevida
16.
Mol Ther ; 16(2): 280-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18059373

RESUMO

Self-complementary adeno-associated virus (scAAV) vectors can significantly minimize the vector load required to achieve sustained transgene expression. In this study, transcriptional regulatory elements were systematically screened to produce constitutive and liver-specific scAAV factor IX (FIX) expression cassettes. In addition, optimization of GC content, cis- regulatory elements, and codon usage in the human FIX (hFIX) transgene increased expression 4-20-fold. A vector was developed that was capable of expressing high FIX levels in comparison with the single-stranded (ss) AAV vector used in a recent clinical trial. The ssAAV and scAAV vectors display different transgene expression and genome stability patterns in the liver, as determined by immunohistochemical staining, in situ messenger RNA (mRNA) hybridization and vector genome quantitation. The ssAAV2 vector promoted strong FIX expression in only a subset of hepatocytes. The scAAV2-hFIX vector showed widespread ( approximately 80% of hepatocytes), moderate FIX expression levels similar to normal livers with correction of coagulation function in FIX-deficient mice. The ability of low dose scAAV-FIX vectors to achieve near-physiological expression may circumvent inflammatory responses in the liver. In addition to providing an improved scAAV vector for potential application in future hemophilia B clinical trials and liver-directed gene delivery, these studies underscore the need for rigorous analysis and optimization of vector genome cassettes.


Assuntos
Dependovirus/genética , Fator IX/genética , Vetores Genéticos/genética , Hemofilia B/terapia , Animais , Composição de Bases/genética , Southern Blotting , Linhagem Celular Tumoral , Códon/genética , Fator IX/metabolismo , Fator IX/fisiologia , Expressão Gênica/genética , Terapia Genética/métodos , Humanos , Imuno-Histoquímica , Hibridização In Situ , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transcrição Gênica
17.
Mol Ther ; 16(7): 1252-1260, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28178482

RESUMO

We report a DNA shuffling-based approach for developing cell type-specific vectors through directed evolution. Capsid genomes of adeno-associated virus (AAV) serotypes 1-9 were randomly fragmented and reassembled using PCR to generate a chimeric capsid library. A single infectious clone (chimeric-1829) containing genome fragments from AAV1, 2, 8, and 9 was isolated from an integrin minus hamster melanoma cell line previously shown to have low permissiveness to AAV. Molecular modeling studies suggest that AAV2 contributes to surface loops at the icosahedral threefold axis of symmetry, while AAV1 and 9 contribute to two- and fivefold symmetry interactions, respectively. The C-terminal domain (AAV9) was identified as a critical structural determinant of melanoma tropism through rational mutagenesis. Chimeric-1829 utilizes heparan sulfate as a primary receptor and transduces melanoma cells more efficiently than all serotypes. Further, chimeric-1829 demonstrates altered tropism in rodent skeletal muscle, liver, and brain including nonhuman primates. We determined a unique immunological profile based on neutralizing antibody (NAb) titer and crossreactivity studies strongly supporting isolation of a synthetic laboratory-derived capsid variant. Application of this technology to alternative cell/tissue types using AAV or other viral capsid sequences is likely to yield a new class of biological nanoparticles as vectors for human gene transfer.

18.
Mol Ther ; 16(7): 1252-60, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18500254

RESUMO

We report a DNA shuffling-based approach for developing cell type-specific vectors through directed evolution. Capsid genomes of adeno-associated virus (AAV) serotypes 1-9 were randomly fragmented and reassembled using PCR to generate a chimeric capsid library. A single infectious clone (chimeric-1829) containing genome fragments from AAV1, 2, 8, and 9 was isolated from an integrin minus hamster melanoma cell line previously shown to have low permissiveness to AAV. Molecular modeling studies suggest that AAV2 contributes to surface loops at the icosahedral threefold axis of symmetry, while AAV1 and 9 contribute to two- and fivefold symmetry interactions, respectively. The C-terminal domain (AAV9) was identified as a critical structural determinant of melanoma tropism through rational mutagenesis. Chimeric-1829 utilizes heparan sulfate as a primary receptor and transduces melanoma cells more efficiently than all serotypes. Further, chimeric-1829 demonstrates altered tropism in rodent skeletal muscle, liver, and brain including nonhuman primates. We determined a unique immunological profile based on neutralizing antibody (NAb) titer and crossreactivity studies strongly supporting isolation of a synthetic laboratory-derived capsid variant. Application of this technology to alternative cell/tissue types using AAV or other viral capsid sequences is likely to yield a new class of biological nanoparticles as vectors for human gene transfer.


Assuntos
Embaralhamento de DNA , Dependovirus/genética , Vetores Genéticos/isolamento & purificação , Genoma Viral/genética , Nanopartículas , Animais , Anticorpos/imunologia , Encéfalo/metabolismo , Capsídeo/imunologia , Cricetinae , Dependovirus/ultraestrutura , Evolução Molecular Direcionada , Biblioteca Gênica , Vetores Genéticos/genética , Humanos , Fígado/metabolismo , Melanoma , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/metabolismo , Primatas , Transdução Genética , Internalização do Vírus
19.
Cancer Res ; 67(7): 3002-9, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17409406

RESUMO

Malignant rhabdoid tumors (MRT) are rare aggressive cancers that occur in young children. Seventy-five percent of sporadic MRTs harbor inactivating SNF5 mutations, and mice heterozygous for an Snf5-null allele develop MRTs with partial penetrance. The diagnosis of choroid plexus carcinomas (CPC) in addition to MRTs in families with a single mutant SNF5 allele prompted us to assess the role of SNF5 loss in CPC in genetically engineered mice. With high frequency, TgT(121) mice develop CPCs that are initiated by inactivation of retinoblastoma protein (pRb) and related proteins p107 and p130. However, CPC penetrance and latency were not significantly affected by Snf5 heterozygosity, consistent with recent evidence that CPCs in SNF5 families were, in many cases, misdiagnosed MRTs. Surprisingly, although the CPC phenotype was unaffected, TgT(121);Snf5(+/-) mice developed MRTs with increased penetrance and decreased latency compared with TgT(121);Snf5(+/+) littermates. MRTs expressed the T(121) protein with a concomitant increase in mitotic activity. The predominant appearance of TgT(121);Snf5(+/-) MRTs in the spinal cord led to the discovery that these tumors likely arose from a subset of spinal cord neural progenitor cells expressing T(121) rather than from transdifferentiation of CPC. Significantly, the target cell type(s) for MRT is unknown. Hence, this study not only shows that pRb(f) and SNF5 inactivation cooperate to induce MRTs but also provides new insight into the MRT target population.


Assuntos
Neoplasias do Plexo Corióideo/genética , Proteínas Cromossômicas não Histona/genética , Proteína do Retinoblastoma/genética , Tumor Rabdoide/genética , Animais , DNA de Neoplasias/genética , DNA de Neoplasias/isolamento & purificação , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína SMARCB1
20.
Mol Cell Biol ; 25(17): 7796-802, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16107724

RESUMO

Aneuploidy is a common feature of human tumors, often correlating with poor prognosis. The mitotic spindle checkpoint is thought to play a major role in aneuploidy suppression. To investigate the role of the spindle checkpoint in tumor suppression in vivo, we developed transgenic mice in which thymocytes express a dominant interfering fragment of Bub1, a kinase regulator of the spindle checkpoint. We report that, despite high-level expression of dominant-negative Bub1 (Bub1DN), a protein known to inhibit spindle checkpoint activity in cultured cells, thymocytes show no evidence of spindle checkpoint impairment. Transgenic animals also failed to show an increased predisposition to spontaneous tumors. Moreover, the Bub1DN transgene failed to alter the timing or characteristics of thymic lymphoma development in p53 heterozygous or homozygous null backgrounds, indicating that the lack of tumorigenesis is not due to suppression by p53-dependent checkpoints. These results indicate that overexpression of a Bub1 N-terminal fragment is insufficient to impair the spindle checkpoint in vivo or to drive tumorigenesis in the highly susceptible murine thymocyte system, either alone or in combination with G(1) checkpoint disruption.


Assuntos
Mutação/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Animais , Ciclo Celular , Transformação Celular Neoplásica/genética , Células Cultivadas , Regulação da Expressão Gênica , Genes Dominantes/genética , Camundongos , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases , Taxa de Sobrevida , Neoplasias da Glândula Tireoide/metabolismo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA