Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Stem Cells ; 34(5): 1321-31, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26756672

RESUMO

Inhibitor of DNA binding proteins (Id1-Id4) function to inhibit differentiation and promote proliferation of many different cell types. Among the Id family members, Id2 has been most extensively studied in the central nervous system (CNS). Id2 contributes to cultured neural precursor cell (NPC) proliferation as well as to the proliferation of CNS tumors such as glioblastoma that are likely to arise from NPC-like cells. We identified three phosphorylation sites near the N-terminus of Id2 in NPCs. To interrogate the importance of Id2 phosphorylation, Id2(-/-) NPCs were modified to express wild type (WT) Id2 or an Id2 mutant protein that could not be phosphorylated at the identified sites. We observed that NPCs expressing this mutant lacking phosphorylation near the N-terminus had higher steady-state levels of Id2 when compared to NPCs expressing WT Id2. This elevated level was the result of a longer half-life and reduced proteasome-mediated degradation. Moreover, NPCs expressing constitutively de-phosphorylated Id2 proliferated more rapidly than NPCs expressing WT Id2, a finding consistent with the well-characterized function of Id2 in driving proliferation. Observing that phosphorylation of Id2 modulates the degradation of this important cell-cycle regulator, we sought to identify a phosphatase that would stabilize Id2 enhancing its activity in NPCs and extended our analysis to include human glioblastoma-derived stem cells (GSCs). We found that expression of the phosphatase PP2A altered Id2 levels. Our findings suggest that inhibition of PP2A may be a novel strategy to regulate the proliferation of normal NPCs and malignant GSCs by decreasing Id2 levels. Stem Cells 2016;34:1321-1331.


Assuntos
Proteína 2 Inibidora de Diferenciação/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Proteólise , Sequência de Aminoácidos , Animais , Ciclo Celular , Proliferação de Células , Glioblastoma/patologia , Proteína 2 Inibidora de Diferenciação/química , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Fosfatase 2/metabolismo
2.
J Pediatr Hematol Oncol ; 38(4): 249-60, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26989915

RESUMO

Great progress has been made in many areas of pediatric oncology. However, tumors of the central nervous system (CNS) remain a significant challenge. A recent explosion of data has led to an opportunity to understand better the molecular basis of these diseases and is already providing a foundation for the pursuit of rationally chosen therapeutics targeting relevant molecular pathways. The molecular biology of pediatric brain tumors is shifting from a singular focus on basic scientific discovery to a platform upon which insights are being translated into therapies.


Assuntos
Neoplasias do Sistema Nervoso Central , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Neoplasias do Sistema Nervoso Central/patologia , Neoplasias do Sistema Nervoso Central/terapia , Criança , Glioma , Humanos , Meduloblastoma , Biologia Molecular/métodos , Biologia Molecular/tendências
3.
Stem Cells ; 31(5): 1010-21, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23390122

RESUMO

Id2 is a helix-loop-helix transcription factor essential for normal development, and its expression is dysregulated in many human neurological conditions. Although it is speculated that elevated Id2 levels contribute to the pathogenesis of these disorders, it is unknown whether dysregulated Id2 expression is sufficient to perturb normal brain development or function. Here, we show that mice with elevated Id2 expression during embryonic stages develop microcephaly, and that females in particular are prone to generalized tonic-clonic seizures. Analyses of Id2 transgenic brains indicate that Id2 activity is highly cell context specific: elevated Id2 expression in naive neural stem cells (NSCs) in early neuroepithelium induces apoptosis and loss of NSCs and intermediate progenitors. Activation of Id2 in maturing neuroepithelium results in less severe phenotypes and is accompanied by elevation of G1 cyclin expression and p53 target gene expression. In contrast, activation of Id2 in committed intermediate progenitors has no significant phenotype. Functional analysis with Id2-overexpressing and Id2-null NSCs shows that Id2 negatively regulates NSC self-renewal in vivo, in contrast to previous cell culture experiments. Deletion of p53 function from Id2-transgenic brains rescues apoptosis and results in increased incidence of brain tumors. Furthermore, Id2 overexpression normalizes the increased self-renewal of p53-null NSCs, suggesting that Id2 activates and modulates the p53 pathway in NSCs. Together, these data suggest that elevated Id2 expression in embryonic brains can cause deregulated NSC self-renewal, differentiation, and survival that manifest in multiple neurological outcomes in mature brains, including microcephaly, seizures, and brain tumors.


Assuntos
Encéfalo/anormalidades , Encéfalo/citologia , Proteína 2 Inibidora de Diferenciação/biossíntese , Células-Tronco Neurais/metabolismo , Animais , Encéfalo/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/citologia
4.
Stem Cells ; 29(7): 1090-101, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21608079

RESUMO

Neural progenitor cells (NPCs) have the capacity to proliferate and give rise to all major central nervous system cell types and represent a possible cell of origin in gliomagenesis. Deletion of the tumor suppressor gene Tp53 (p53) results in increased proliferation and self-renewal of NPCs and is a common genetic mutation found in glioma. We have identified inhibitor of DNA binding 2 (Id2) as a novel target gene directly repressed by p53 to maintain normal NPC proliferation. p53((-/-)) NPCs express elevated levels of Id2 and suppression of Id2 expression is sufficient to inhibit the increased proliferation and self-renewal which results from p53 loss. Elevated expression of Id2 in wild-type NPCs phenocopies the behavior of p53((-/-)) NPCs by enhancing NPC proliferation and self-renewal. Interestingly, p53 directly binds to a conserved site within the Id2 promoter to mediate these effects. Finally, we have identified elevated Id2 expression in glioma cell lines with mutated p53 and demonstrated that constitutive expression of Id2 plays a key role in the proliferation of glioma stem-like cells. These findings indicate that Id2 functions as a proproliferative gene that antagonizes p53-mediated cell cycle regulation in NPCs and may contribute to the malignant proliferation of glioma-derived tumor stem cells.


Assuntos
Genes p53 , Proteína 2 Inibidora de Diferenciação/genética , Células-Tronco Neurais/citologia , Células-Tronco/citologia , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Proteína 2 Inibidora de Diferenciação/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Regiões Promotoras Genéticas
5.
J Biol Chem ; 285(31): 24164-73, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20460371

RESUMO

Inhibitor of DNA binding 4 (ID4) is a helix-loop-helix protein that heterodimerizes with basic helix-loop-helix transcription factors inhibiting their function. ID4 expression is important for adipogenic differentiation of the 3T3-L1 cell line, and inhibition of ID4 is associated with a concomitant decrease in CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma mRNA and protein expression. Mice with a homozygous deletion of Id4 (Id4(-/-)) have reduced body fat and gain much less weight compared with wild-type littermates when placed on diets with high fat content. Mouse embryonic fibroblasts (MEFs) isolated from Id4(-/-) mice have reduced adipogenic potential when compared with wild-type MEFs. In agreement with changes in morphological differentiation, the levels of CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma were also reduced in MEFs from Id4(-/-) mice. Our results demonstrate the importance of ID4 in adipocyte differentiation and the implications of this regulation for adipose tissue formation.


Assuntos
Adipócitos/citologia , Tecido Adiposo/metabolismo , Proteínas Inibidoras de Diferenciação/fisiologia , Células 3T3-L1 , Animais , Peso Corporal , Diferenciação Celular , Dimerização , Fibroblastos/metabolismo , Deleção de Genes , Genótipo , Homozigoto , Proteínas Inibidoras de Diferenciação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos
6.
Radiographics ; 31(6): 1717-40, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21997991

RESUMO

Glioblastoma is a highly malignant brain tumor that relentlessly defies therapy. Efforts over the past decade have begun to tease out the biochemical details that lead to its aggressive behavior and poor prognosis. There is hope that this new understanding will lead to improved treatment strategies for patients with glioblastoma, in the form of targeted, molecularly based therapies that are individualized to specific changes in individual tumors. However, these new therapies have the potential to fundamentally alter the biologic behavior of glioblastoma and, as a result, its imaging appearance. Knowledge about common genetic alterations and the resultant cellular and tissue changes (ie, induced angiogenesis and abnormal cell survival, proliferation, and invasion) in glioblastomas is important as a basis for understanding imaging findings before treatment. It is equally critical that radiologists understand which genetic pathway is targeted by each specific therapeutic agent or class of agents in order to accurately interpret changes in the imaging appearances of treated tumors.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Imageamento por Ressonância Magnética , Animais , Neoplasias Encefálicas/classificação , Neoplasias Encefálicas/patologia , Proliferação de Células , Sobrevivência Celular/genética , Cloridrato de Erlotinib , Gefitinibe , Regulação Neoplásica da Expressão Gênica , Glioblastoma/classificação , Glioblastoma/patologia , Humanos , Invasividade Neoplásica/genética , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia
7.
Sci Rep ; 11(1): 10883, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34035431

RESUMO

Proteasome inhibitors bortezomib and carfilzomib are approved for the treatment of multiple myeloma and mantle cell lymphoma and have demonstrated clinical efficacy for the treatment of acute lymphoblastic leukemia (ALL). The t(4;11)(q21;q23) chromosomal translocation that leads to the expression of MLL-AF4 fusion protein and confers a poor prognosis, is the major cause of infant ALL. This translocation sensitizes tumor cells to proteasome inhibitors, but toxicities of bortezomib and carfilzomib may limit their use in pediatric patients. Many of these toxicities are caused by on-target inhibition of proteasomes in non-lymphoid tissues (e.g., heart muscle, gut, testicles). We found that MLL-AF4 cells express high levels of lymphoid tissue-specific immunoproteasomes and are sensitive to pharmacologically relevant concentrations of specific immunoproteasome inhibitor ONX-0914, even in the presence of stromal cells. Inhibition of multiple active sites of the immunoproteasomes was required to achieve cytotoxicity against ALL. ONX-0914, an inhibitor of LMP7 (ß5i) and LMP2 (ß1i) sites of the immunoproteasome, and LU-102, inhibitor of proteasome ß2 sites, exhibited synergistic cytotoxicity. Treatment with ONX-0914 significantly delayed the growth of orthotopic ALL xenograft tumors in mice. T-cell ALL lines were also sensitive to pharmacologically relevant concentrations of ONX-0914. This study provides a strong rationale for testing clinical stage immunoproteasome inhibitors KZ-616 and M3258 in ALL.


Assuntos
Proteína de Leucina Linfoide-Mieloide/genética , Oligopeptídeos/administração & dosagem , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Inibidores de Proteassoma/administração & dosagem , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Oligopeptídeos/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Inibidores de Proteassoma/farmacologia , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Biol Chem ; 284(46): 31735-45, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19740747

RESUMO

Id2 is a helix-loop-helix transcription factor gene expressed in a circadian manner in multiple tissues with a phase-locked relationship with canonical clock genes. Our previous studies have identified circadian phenotypes in Id2 null mice, including enhanced photo-entrainment and disruption of activity rhythms, and have demonstrated a potent inhibitory effect of ID proteins upon CLOCK-BMAL1 transactivation of clock gene and clock-controlled gene activity. We have now begun to explore the potential role that ID2 may play in specifically regulating clock output. Here we show that ID2 protein is rhythmically expressed in mouse liver. Time-of-day-specific liver gene expression in Id2(+/+) and Id2(-/-) mice under circadian conditions was studied using DNA microarray analysis, identifying 651 differentially expressed genes, including a subset of 318 genes deemed rhythmically expressed in other studies. Examination of individual time courses reveals that these genes are dysregulated in a highly time-specific manner. A cohort of different functional groups were identified, including genes associated with glucose and lipid metabolism, e.g. serum protein Igfbp1 and lipoprotein lipase. We also reveal that the Id2(-/-) mice show a reduction in lipid storage in the liver and white adipose tissue, suggesting that disruption of normal circadian activity of components of lipid metabolism can result in overt physiological alterations. These data reveal a role for the transcriptional repressor ID2 as a circadian output regulator in the periphery.


Assuntos
Ritmo Circadiano/fisiologia , Proteína 2 Inibidora de Diferenciação/fisiologia , Fígado/metabolismo , Proteínas Repressoras/fisiologia , Animais , Western Blotting , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
J Neurosci ; 28(52): 14074-86, 2008 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-19109490

RESUMO

Understanding the biology of adult neural stem cells has important implications for nervous system development and may contribute to our understanding of neurodegenerative disorders and their treatment. We have characterized the process of olfactory neurogenesis in adult mice lacking inhibitor of DNA binding 2(-/-) (Id2(-/-)). We found a diminished olfactory bulb containing reduced numbers of granular and periglomerular neurons with a distinct paucity of dopaminergic periglomerular neurons. While no deficiency of the stem cell compartment was detectable, migrating neuroblasts in Id2(-/-) mutant mice prematurely undergo astroglial differentiation within a disorganized rostral migratory stream. Further, when evaluated in vitro loss of Id2 results in decreased proliferation of neural progenitors and decreased expression of the Hes1 and Ascl1 (Mash1) transcription factors, known mediators of neuronal differentiation. These data support a novel role for sustained Id2 expression in migrating neural progenitors mediating olfactory dopaminergic neuronal differentiation in adult animals.


Assuntos
Dopamina/metabolismo , Proteína 2 Inibidora de Diferenciação/fisiologia , Neurogênese/genética , Neurônios/fisiologia , Bulbo Olfatório/citologia , Células-Tronco Adultas/fisiologia , Animais , Animais Recém-Nascidos , Astrócitos/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Diferenciação Celular/genética , Células Cultivadas , Discriminação Psicológica/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteína 2 Inibidora de Diferenciação/deficiência , Camundongos , Camundongos Knockout , Neurogênese/fisiologia , Bulbo Olfatório/crescimento & desenvolvimento , Condutos Olfatórios/citologia , Condutos Olfatórios/fisiologia , Olfato/genética , Estatísticas não Paramétricas , Fatores de Transcrição HES-1 , Tirosina 3-Mono-Oxigenase/metabolismo
10.
Cancer Res ; 67(6): 2456-68, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363563

RESUMO

MicroRNAs are small noncoding RNAs that function by regulating target gene expression posttranscriptionally. They play a critical role in developmental and physiologic processes and are implicated in the pathogenesis of several human diseases including cancer. We examined the expression profiles of 241 human microRNAs in normal tissues and the NCI-60 panel of human tumor-derived cell lines. To quantify microRNA expression, we employed a highly sensitive technique that uses stem-loop primers for reverse transcription followed by real-time PCR. Most microRNAs were expressed at lower levels in tumor-derived cell lines compared with the corresponding normal tissue. Agglomerative hierarchical clustering analysis of microRNA expression revealed four groups among the NCI-60 cell lines consisting of hematologic, colon, central nervous system, and melanoma tumor-derived cell lines clustered in a manner that reflected their tissue of origin. We identified specific subsets of microRNAs that provide candidate molecular signatures characteristic of the tumor-derived cell lines belonging to these four clusters. We also identified specific microRNA expression patterns that correlated with the proliferation indices of the NCI-60 cell lines, and we developed evidence for the identification of specific microRNAs as candidate oncogenes and tumor suppressor genes in different tumor types. Our results provide evidence that microRNA expression patterns may mark specific biological characteristics of tumors and/or mediate biological activities important for the pathobiology of malignant tumors. These findings call attention to the potential of microRNAs to provide etiologic insights as well as to serve as both diagnostic markers and therapeutic targets for many different tumor types.


Assuntos
MicroRNAs/genética , Neoplasias/genética , Linhagem Celular Tumoral , Análise por Conglomerados , Perfilação da Expressão Gênica , Genes Supressores de Tumor , Humanos , MicroRNAs/biossíntese , Neoplasias/metabolismo , Oncogenes , Regulação para Cima
11.
J Neurosurg ; 132(6): 1706-1714, 2019 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-31125970

RESUMO

OBJECTIVE: 5-aminolevulinic acid (5-ALA)-induced protoporphyrin IX (PpIX) fluorescence is an effective surgical adjunct for the intraoperative identification of tumor tissue during resection of high-grade gliomas. The use of 5-ALA-induced PpIX fluorescence in glioblastoma (GBM) has been shown to double the extent of gross-total resection and 6-month progression-free survival. The heterogeneity of 5-ALA-induced PpIX fluorescence observed during surgery presents a technical and diagnostic challenge when utilizing this tool intraoperatively. While some regions show bright fluorescence after 5-ALA administration, other regions do not, despite that both regions of the tumor may be histopathologically indistinguishable. The authors examined the biological basis of this heterogeneity using computational methods. METHODS: The authors collected both fluorescent and nonfluorescent GBM specimens from a total of 14 patients undergoing surgery and examined their gene expression profiles. RESULTS: In this study, the authors found that the gene expression patterns characterizing fluorescent and nonfluorescent GBM surgical specimens were profoundly different and were associated with distinct cellular functions and different biological pathways. Nonfluorescent tumor tissue tended to resemble the neural subtype of GBM; meanwhile, fluorescent tumor tissue did not exhibit a prominent pattern corresponding to known subtypes of GBM. Consistent with this observation, neural GBM samples from The Cancer Genome Atlas database exhibited a significantly lower fluorescence score than nonneural GBM samples as determined by a fluorescence gene signature developed by the authors. CONCLUSIONS: These results provide a greater understanding regarding the biological basis of differential fluorescence observed intraoperatively and can provide a basis to identify novel strategies to maximize the effectiveness of fluorescence agents.

12.
Cancer Res ; 78(21): 6031-6039, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30333116

RESUMO

Cellular secretion is an important mediator of cancer progression. Secreted molecules in glioma are key components of complex autocrine and paracrine pathways that mediate multiple oncogenic pathologies. In this review, we describe tumor cell secretion in high-grade glioma and highlight potential novel therapeutic opportunities. Cancer Res; 78(21); 6031-9. ©2018 AACR.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Animais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Vesículas Extracelulares/metabolismo , Glioma/metabolismo , Glioma/terapia , Humanos , Camundongos , Transplante de Neoplasias , Oncogenes , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas
13.
Neuro Oncol ; 20(3): 332-342, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29016807

RESUMO

Background: Mouse models of glioblastoma (GBM), the most aggressive primary brain tumor, are critical for understanding GBM pathology and can contribute to the preclinical evaluation of therapeutic agents. Platelet-derived growth factor (PDGF) signaling has been implicated in the development and pathogenesis of GBM, specifically the proneural subtype. Although multiple mouse models of PDGF-driven glioma have been described, they require transgenic mice engineered to activate PDGF signaling and/or impair tumor suppressor genes and typically represent lower-grade glioma. Methods: We designed recombinant lentiviruses expressing both PDGFB and a short hairpin RNA targeting Cdkn2a to induce gliomagenesis following stereotactic injection into the dentate gyrus of adult immunocompetent mice. We engineered these viruses to coexpress CreERT2 with PDGFB, allowing for deletion of floxed genes specifically in transduced cells, and designed another version of this recombinant lentivirus in which enhanced green fluorescent protein was coexpressed with PDGFB and CreERT2 to visualize transduced cells. Results: The dentate gyrus of injected mice showed hypercellularity one week post-injection and subsequently developed bona fide tumors with the pathologic hallmarks of GBM leading to a median survival of 77 days post-injection. Transcriptomic analysis of these tumors revealed a proneural gene expression signature. Conclusion: Informed by the genetic alterations observed in human GBM, we engineered a novel mouse model of proneural GBM. While reflecting many of the advantages of transgenic mice, this model allows for the facile in vivo testing of gene function in tumor cells and makes possible the rapid production of large numbers of immunocompetent tumor-bearing mice for preclinical testing of therapeutics.


Assuntos
Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Glioblastoma/patologia , Lentivirus/genética , Linfocinas/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/genética , Modelos Animais de Doenças , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Linfocinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais , Células Tumorais Cultivadas
14.
Mol Cell Biol ; 24(5): 2083-90, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14966287

RESUMO

We have previously described a transgenic mouse model of epidermal neoplasia wherein expression of a switchable form of c-Myc, MycER(TAM), is targeted to the postmitotic suprabasal keratinocytes of murine epidermis via the involucrin promoter. Sustained activation of c-MycER(TAM) results in a progressive neoplastic phenotype characterized by aberrant ectopic proliferation and delayed differentiation of suprabasal keratinocytes, culminating in papillomatosis. Transcription of the Id2 gene is regulated by Myc family proteins. Moreover, Id2 is implicated as a pivotal determinant of cell fate in multiple lineages and has a demonstrated role in mediating Myc-dependent cell proliferation in vitro through its interaction with retinoblastoma protein. Using Id2 nullizygous mice, we assessed in vivo the requirement for Id2 in mediating Myc-induced papilloma formation in skin. We show that absence of Id2 has no discernible impact on any measurable attribute of Myc function or on the timing or extent of eventual tumor formation. Thus, our data argue against any essential role for Id2 in mediating Myc action in vivo.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Epiderme/metabolismo , Papiloma/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/genética , Células Epidérmicas , Epiderme/patologia , Hibridização In Situ , Proteína 1 Inibidora de Diferenciação , Proteína 2 Inibidora de Diferenciação , Queratinócitos/citologia , Queratinócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica , Papiloma/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Neoplasias Cutâneas/patologia , Fatores de Transcrição/genética
15.
Cell Death Dis ; 8(2): e2615, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-28206987

RESUMO

Tumor cells proliferate in cellular environments characterized by a lack of optimal tissue organization resulting oftentimes in compromised cellular metabolism affecting nutrition, respiration, and energetics. The response of tumor cells to adverse environmental conditions is a key feature affecting their pathogenicity. We found that inhibitor of DNA binding 2 (ID2) expression levels significantly correlate with the ability of glioblastoma (GBM)-derived cell lines to survive glucose deprivation. ID2 suppressed mitochondrial oxidative respiration and mitochondrial ATP production by regulating the function of mitochondrial electron transport chain (mETC) complexes, resulting in reduced superoxide and reactive oxygen species (ROS) production from mitochondria. ID2 suppression of ROS production reduced mitochondrial damage and enhanced tumor cell survival during glucose deprivation. Bioinformatics analysis of GBM gene expression data from The Cancer Genome Atlas (TCGA) database revealed that expression of ID2 mRNA is unique among ID gene family members in correlating with the expression of nuclear genes involved in mitochondrial energy metabolism and assembly of mETC. Our data indicate that the expression level of ID2 in GBM cells can predict the sensitivity of GBM-derived tumor cells to decreased glucose levels. Low levels of ID2 expression in human GBM tissues may identify a clinical group in which metabolic targeting of glycolytic pathways can be expected to have the greatest therapeutic efficacy.


Assuntos
Glioblastoma/metabolismo , Proteína 2 Inibidora de Diferenciação/metabolismo , Mitocôndrias/metabolismo , Linhagem Celular Tumoral , Metabolismo Energético , Glioblastoma/genética , Humanos , Proteína 2 Inibidora de Diferenciação/biossíntese , Proteína 2 Inibidora de Diferenciação/genética , Estresse Fisiológico , Análise de Sobrevida
16.
Mol Cancer Ther ; 16(4): 705-716, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28138037

RESUMO

Despite abundant evidence implicating receptor tyrosine kinases (RTK), including the platelet-derived growth factor receptor (PDGFR), in the pathogenesis of glioblastoma (GBM), the clinical use of RTK inhibitors in this disease has been greatly compromised by the rapid emergence of therapeutic resistance. To study the resistance of proneural gliomas that are driven by a PDGFR-regulated pathway to targeted tyrosine kinase inhibitors, we utilized a mouse model of proneural glioma in which mice develop tumors that become resistant to PDGFR inhibition. We found that tumors resistant to PDGFR inhibition required the expression and activation of the insulin receptor (IR)/insulin growth-like factor receptor (IGF1R) for tumor cell proliferation and survival. Cotargeting IR/IGF1R and PDGFR decreased the emergence of resistant clones in vitro Our findings characterize a novel model of glioma recurrence that implicates the IR/IGF1R signaling axis in mediating the development of resistance to PDGFR inhibition and provide evidence that IR/IGF1R signaling is important in the recurrence of the proneural subtype of glioma in which PDGF/PDGFR is most commonly expressed at a high level. Mol Cancer Ther; 16(4); 705-16. ©2017 AACR.


Assuntos
Neoplasias Encefálicas/genética , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/genética , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Esferoides Celulares/patologia , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Cromonas/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Mesilato de Imatinib/farmacologia , Imidazóis/farmacologia , Insulina/metabolismo , Camundongos , Morfolinas/farmacologia , Transplante de Neoplasias , Pirazinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Esferoides Celulares/transplante , Células Tumorais Cultivadas , Tirfostinas/farmacologia
17.
Cancer Res ; 62(13): 3729-35, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12097282

RESUMO

Glioblastoma multiforme, the most common form of malignant brain tumor,is resistant to all forms of therapy and causes death within 9-12 months of diagnosis. Glioblastomas are known to contain numerous genetic and physiological alterations affecting cell survival and proliferation; one of the most common alterations being platelet-derived growth factor (PDGF) autocrine signaling characterized by coexpression of PDGF and its receptor. The PDGF family consists of four members, PDGF-A, -B, -C, and -D, that signal through the alpha and beta PDGF receptor (PDGFR) tyrosine kinases. Numerous studies have demonstrated expression of PDGF-A, PDGF-B, and the PDGFRs in gliomablastomas, but such studies have not been conducted for the newly identified PDGF-C and -D. Therefore, we examined the expression of all PDGF ligands and receptors in 11 glioma cell lines and 5 primary glioblastoma tumor tissues by quantitative reverse transcription-PCR. Expression of PDGF/PDGFR pairs that are known to functionally interact were identified in all of the samples. Interestingly, PDGF-C expression was ubiquitous in brain tumor cells and tissues but was very low or absent in normal adult and fetal brain. PDGF-D was expressed in 10 of 11 brain tumor cell lines and 3 of 5 primary brain tumor samples. As a strategy for blocking PDGFR signaling, CT52923, a potent selective small molecule piperazinyl quinazoline kinase inhibitor of the PDGFR, was identified. In model systems using NIH/3T3 cells, CT52923 blocked PDGF autocrine-mediated phosphorylation of PDGFR, Akt, and mitogen-activated protein kinase (MAPK), while having no effect on v-fms or V12-ras-mediated Akt or extracellular signal-regulated protein kinase (Erk) phosphorylation. More importantly, p.o. administration of CT52923 to nude mice caused a significant 61% reduction (P < 0.006) in tumor growth of NIH/3T3 cells transformed by PDGF, whereas tumor formation by cells expressing v-fms was unaffected. We next characterized PDGF autocrine signaling in five glioblastoma cell lines. In all of the cases, PDGF autocrine signaling was evident because treatment with 1-10 microM CT52923 inhibited PDGFR autophosphorylation when present at a detectable level and blocked downstream Akt and/or Erk phosphorylation. The functional significance of PDGF autocrine signaling in these cells was demonstrated by the fact that the CT52923 inhibited soft agar colony formation, and, when given p.o. to nude mice, it effectively reduced tumor formation by 44% (P < 0.0019) after s.c. injection of C6 glioblastoma cells. This study of glioblastoma cells and primary tissues is the first to implicate PDGF-C and -D in brain tumor formation and confirms the existence of autocrine signaling by PDGF-A and -B. More importantly, treatment with the PDGFR antagonist CT52923 inhibited survival and/or mitogenic pathways in all of the glioblastoma cell lines tested and prevented glioma formation in a nude mouse xenograft model. Together these findings demonstrate the potential therapeutic utility of this class of compounds for the treatment of glioblastoma.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Linfocinas , Piperazinas/farmacologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Serina-Treonina Quinases , Quinazolinas/farmacologia , Células 3T3/citologia , Células 3T3/efeitos dos fármacos , Células 3T3/metabolismo , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Ligantes , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
18.
Cancer Res ; 63(5): 923-8, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12615704

RESUMO

Abnormal hedgehog signaling, most commonly caused by loss of PTCH1 inhibitor activity,drives tumorigenesis of basal cell carcinomas (BCCs). To assess whether other tumors also have abnormal hedgehog signaling, we have assayed RNA from common cancers at nine different sites for levels of expression of hedgehog target genes that are up-regulated uniformly in BCCs. We report here that such dysregulation appears not to be common in the types of non-BCC cancers studied, indicating that the molecular pathogenesis of BCCs, like their frequency and behavior, differs markedly from that of most other cancers.


Assuntos
Neoplasias/genética , Neoplasias/metabolismo , Transativadores/fisiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Criança , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog , Humanos , Masculino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Neoplasias/patologia , Proteínas Oncogênicas/biossíntese , Proteínas Oncogênicas/genética , Receptores Patched , Receptor Patched-1 , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptores de Superfície Celular , Transdução de Sinais , Transativadores/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Proteína GLI1 em Dedos de Zinco
19.
Cancer Res ; 63(7): 1589-95, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12670909

RESUMO

Oligodendrogliomas of all grades overexpress epidermal growth factor receptor (EGFR), whereas deletion of ink4a/arf is found only in high-grade tumors. We used the S100 beta promoter to generate transgenic mice expressing v-erbB, a transforming allele of EGFR. These mice developed low-grade oligodendroglioma. Transgenic animals heterozygous for ink4a/arf or p53 developed high-grade tumors. Comparative genomic hybridization revealed loss of distal mouse chromosome 4, a region orthologous with human chromosome 1p, which is commonly lost in oligodendroglioma. Our results demonstrate that overexpression of EGFR, an epigenetic observation of uncertain significance in human oligodendroglioma, can initiate oligodendroglioma in the mouse. Furthermore, p53 pathway mutations can mediate the transition from low to high grade. These models hold promise for studying tumor lineage, identifying contributing genetic alterations and evaluating preclinical therapies in this important neoplasm.


Assuntos
Neoplasias Encefálicas/genética , Receptores ErbB/biossíntese , Genes erbB-1/genética , Oligodendroglioma/genética , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Aberrações Cromossômicas , Cromossomos de Mamíferos , Inibidor p16 de Quinase Dependente de Ciclina/genética , Modelos Animais de Doenças , Receptores ErbB/genética , Genes p53 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Mutação , Fatores de Crescimento Neural/genética , Oligodendroglioma/metabolismo , Oligodendroglioma/patologia , Proteínas Oncogênicas v-erbB/biossíntese , Proteínas Oncogênicas v-erbB/genética , Regiões Promotoras Genéticas , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/genética , Células Tumorais Cultivadas
20.
Cancer Res ; 76(10): 2964-76, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26951930

RESUMO

Glioblastoma is the most aggressive primary brain tumor and responds poorly to currently available therapies. Transcriptomic characterization of glioblastoma has identified distinct molecular subtypes of glioblastoma. Gain-of-function alterations leading to enhanced platelet-derived growth factor (PDGF) signaling are commonly observed in the proneural subtype of glioblastoma and can drive gliomagenesis. However, little is known about the downstream effectors of PDGF signaling in glioblastoma. Using a mouse model of proneural glioma and comparative transcriptomics, we determined that PDGF signaling upregulated ubiquitin-specific peptidase 1 (Usp1) to promote the survival of murine proneural glioma cells. Mechanistically, we found that PDGF signaling regulated the expression of the E2F transcription factors, which directly bound to and activated Usp1 Furthermore, PDGF-mediated expression of USP1 led to the stabilization of Inhibitor of DNA-binding 2 (ID2), which we found to be required for glioma cell survival. Genetic ablation of Id2 delayed tumor-induced mortality, and pharmacologic inhibition of USP1, resulting in decreased ID2 levels, also delayed tumorigenesis in mice. Notably, decreased USP1 expression was associated with prolonged survival in patients with proneural glioblastoma, but not with other subtypes of glioblastoma. Collectively, our findings describe a signaling cascade downstream of PDGF that sustains proneural glioblastoma cells and suggest that inhibition of the PDGF-E2F-USP1-ID2 axis could serve as a therapeutic strategy for proneural glioblastoma featuring increased PDGF signaling. Cancer Res; 76(10); 2964-76. ©2016 AACR.


Assuntos
Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/patologia , Fator de Transcrição E2F1/metabolismo , Glioma/patologia , Proteína 2 Inibidora de Diferenciação/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Animais , Apoptose , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Fator de Transcrição E2F1/genética , Glioma/genética , Glioma/metabolismo , Humanos , Proteína 2 Inibidora de Diferenciação/genética , Camundongos , Proteínas Proto-Oncogênicas c-sis/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Proteases Específicas de Ubiquitina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA