Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Cancer ; 136(7): 1546-58, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25175359

RESUMO

Glioblastoma multiforme is the most aggressive primary tumor of the central nervous system. Glioma stem cells (GSCs), a small population of tumor cells with stem-like properties, are supposedly responsible for glioblastoma multiforme relapse after current therapies. In approximately thirty percent of glioblastoma multiforme tumors, telomeres are not maintained by telomerase but through an alternative mechanism, termed alternative lengthening of telomere (ALT), suggesting potential interest in developing specific therapeutic strategies. However, no preclinical model of ALT glioma was available until the isolation of TG20 cells from a human ALT glioma. Herein, we show that TG20 cells exhibit a high level of telomeric recombination but a stable karyotype, indicating that their telomeres retain their protective function against chromosomal instability. TG20 cells possess all of the characteristic features of GSCs: the expression of neural stem cell markers, the generation of intracerebral tumors in NOD-SCID-IL2Rγ (NSG) mice as well as in nude mice, and the ability to sustain serial intracerebral transplantations without expressing telomerase, demonstrating the stability of the ALT phenotype in vivo. Furthermore, we also demonstrate that 360B, a G-quadruplex ligand of the pyridine derivative series that impairs telomere replication and mitotic progression in cancer cells, prevents the development of TG20 tumors. Together, our results show that intracerebral grafts of TG20 cells in immunodeficient mice constitute an efficient preclinical model of ALT glioblastoma multiforme and that G-quadruplex ligands are a potential therapy for this specific type of tumor.


Assuntos
Glioma/genética , Telômero/genética , Adulto , Animais , Linhagem Celular Tumoral , Metilação de DNA , Modelos Animais de Doenças , Quadruplex G , Regulação da Expressão Gênica , Glioma/metabolismo , Xenoenxertos , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Fenótipo , Troca de Cromátide Irmã , Telomerase/genética , Telomerase/metabolismo , Telômero/metabolismo , Homeostase do Telômero
2.
Stem Cells ; 29(3): 440-51, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21425407

RESUMO

Cancer stem cells are increasingly recognized as major therapeutic targets. We report here the isolation of glioma stem cells (GSCs) maintaining telomere length through a telomerase-independent mechanism known as alternative lengthening of telomeres (ALTs). TG20 cells were isolated from a glioblastoma multiforme, which had the ALT phenotype. They have no detectable telomerase activity and extremely long and heterogeneous telomeres colocalizing with promyelocytic leukemia bodies. The cancer stem cell potential of TG20 cells was confirmed based on their expression of neural stem cell markers, their capacity of in vitro long-term proliferation and to form intracranial tumors in immune-deficient mice. Interestingly, we found that both in vitro and in vivo TG20 cells were significantly more resistant to ionizing radiation than GSCs with telomerase activity. Analysis of DNA damage foci, DNA double-strand breaks repair, and chromosome instability suggest that radiation resistance was related to interference of ALT pathway with DNA damage response. Therefore, our data show for the first time that the ALT pathway can confer to cancer stem cells the capacity to sustain long-term proliferation as telomerase activity and importantly may also affect treatment efficiency. TG20 cells are thus the first cellular model of GSCs displaying ALT and should prove to be useful for the development of specific treatment strategies.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Células-Tronco Neoplásicas/metabolismo , Telômero/metabolismo , Adulto , Idoso , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Feminino , Glioma/genética , Glioma/metabolismo , Humanos , Hibridização in Situ Fluorescente , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/patologia , Telômero/genética , Transplante Heterólogo , Células Tumorais Cultivadas
3.
Neurochem Res ; 37(6): 1364-71, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22476983

RESUMO

Some 20 years ago c-Fos was identified as a member of the AP-1 family of inducible transcription factors (Angel and Karin in Biochim Biophys Acta 1072:129-157, 1991). More recently, an additional activity was described for this protein: it associates to the endoplasmic reticulum and activates the biosynthesis of phospholipids (Bussolino et al. in FASEB J 15:556-558, 2001), (Gil et al. in Mol Biol Cell 15:1881-1894, 2004), the quantitatively most important components of cellular membranes. This latter activity of c-Fos determines the rate of membrane genesis and consequently of growth in differentiating PC12 cells (Gil et al. in Mol Biol Cell 15:1881-1894, 2004). In addition, it has been shown that c-Fos is over-expressed both in PNS and CNS tumors (Silvestre et al. in PLoS One 5(3):e9544, 2010). Herein, it is shown that c-Fos-activated phospholipid synthesis is required to support membrane genesis during the exacerbated growth characteristic of brain tumor cells. Specifically blocking c-Fos-activated phospholipid synthesis significantly reduces proliferation of tumor cells in culture. Blocking c-Fos expression also prevents tumor progression in mice intra-cranially xeno-grafted human brain tumor cells. In NPcis mice, an animal model of the human disease Neurofibromatosis Type I (Cichowski and Jacks in Cell 104:593-604, 2001), animals spontaneously develop tumors of the PNS and the CNS, provided they express c-Fos (Silvestre et al. in PLoS One 5(3):e9544, 2010). Treatment of PNS tumors with an antisense oligonucleotide that specifically blocks c-Fos expression also blocks tumor growth in vivo. These results disclose cytoplasmic c-Fos as a new target for effectively controlling brain tumor growth.


Assuntos
Proliferação de Células/efeitos dos fármacos , Neoplasias do Sistema Nervoso Central/patologia , Neoplasias do Sistema Nervoso Periférico/patologia , Fosfolipídeos/biossíntese , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Linhagem Celular Tumoral , Sistema Nervoso Central/metabolismo , Neoplasias do Sistema Nervoso Central/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Camundongos , Oligonucleotídeos Antissenso/metabolismo , Células PC12 , Neoplasias do Sistema Nervoso Periférico/metabolismo , Ratos
4.
Cancers (Basel) ; 14(2)2022 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-35053470

RESUMO

Identifying new therapeutic strategies for triple-negative breast cancer (TNBC) patients is a priority as these patients are highly prone to relapse after chemotherapy. Here, we found that protein arginine methyltransferase 1 (PRMT1) is highly expressed in all breast cancer subtypes. PRMT1 depletion decreases cell survival by inducing DNA damage and apoptosis in various breast cancer cell lines. Transcriptomic analysis and chromatin immunoprecipitation revealed that PRMT1 regulates the epidermal growth factor receptor (EGFR) and the Wnt signaling pathways, reported to be activated in TNBC. PRMT1 enzymatic activity is also required to stimulate the canonical Wnt pathway. Type I PRMT inhibitors decrease breast cancer cell proliferation and show anti-tumor activity in a TNBC xenograft model. These inhibitors display synergistic interactions with some chemotherapies used to treat TNBC patients as well as erlotinib, an EGFR inhibitor. Therefore, targeting PRMT1 in combination with these chemotherapies may improve existing treatments for TNBC patients.

5.
J Neurosci Res ; 87(4): 857-65, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18951474

RESUMO

Although the molecular and cellular basis of particular events that lead to the biogenesis of membranes in eukaryotic cells has been described in detail, understanding of the intrinsic complexity of the pleiotropic response by which a cell adjusts the overall activity of its endomembrane system to accomplish these requirements is limited. Here we carried out an immunocytochemical and biochemical examination of the content and quality of the endoplasmic reticulum (ER) and Golgi apparatus membranes in two in vivo situations characterized by a phase of active cell proliferation followed by a phase of declination in proliferation (rat brain tissue at early and late developmental stages) or by permanent active proliferation (gliomas and their most malignant manifestation, glioblastomas multiforme). It was found that, in highly proliferative phases of brain development (early embryo brain cells), the content of ER and Golgi apparatus membranes, measured as total lipid phosphorous content, is higher than in adult brain cells. In addition, the concentration of protein markers of ER and Golgi is also higher in early embryo brain cells and in human glioblastoma multiforme cells than in adult rat brain or in nonpathological human brain cells. Results suggest that the amount of endomembranes and the concentration of constituent functional proteins diminish as cells decline in their proliferative activity.


Assuntos
Encéfalo/citologia , Proliferação de Células , Retículo Endoplasmático/química , Complexo de Golgi/química , Membranas Intracelulares/química , Animais , Western Blotting , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Encéfalo/ultraestrutura , Retículo Endoplasmático/ultraestrutura , Feminino , Glioblastoma/química , Glioblastoma/metabolismo , Glioblastoma/patologia , Complexo de Golgi/ultraestrutura , Humanos , Imuno-Histoquímica , Membranas Intracelulares/ultraestrutura , Lipídeos de Membrana/análise , Proteínas de Membrana/análise , Fósforo/análise , Ratos , Ratos Wistar
6.
Oncotarget ; 8(16): 26269-26280, 2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28412741

RESUMO

Cancer cells can use a telomerase-independent mechanism, known as alternative lengthening of telomeres (ALT), to elongate their telomeres. General control non-derepressible 5 (GCN5) and P300/CBP-associated factor (PCAF) are two homologous acetyltransferases that are mutually exclusive subunits in SAGA-like complexes. Here, we reveal that down regulation of GCN5 and PCAF had differential effects on some phenotypic characteristics of ALT cells. Our results suggest that GCN5 is present at telomeres and opposes telomere recombination, in contrast to PCAF that may indirectly favour them in ALT cells.


Assuntos
Estudos de Associação Genética , Homeostase do Telômero/genética , Telômero/genética , Fatores de Transcrição de p300-CBP/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Expressão Gênica , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Humanos , Corpos de Inclusão Intranuclear/genética , Corpos de Inclusão Intranuclear/metabolismo , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Ligação Proteica , Troca de Cromátide Irmã , Translocação Genética
7.
Oncotarget ; 6(19): 16883-901, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26143639

RESUMO

c-Fos is a proto-oncogene involved in diverse cellular functions. Its deregulation has been associated to abnormal development and oncogenic progression. c-fos-/- mice are viable but present a reduction in their body weight and brain size. We examined the importance of c-Fos during neocortex development at 13.5, 14.5 and 16.5 days of gestation. At E14.5, neocortex thickness, apoptosis, mitosis and expression of markers along the different stages of Neural Stem Progenitor Cells (NSPCs) differentiation in c-fos-/- and wild-type mice were analyzed. A ~15% reduction in the neocortex thickness of c-fos-/- embryos was observed which correlates with a decrease in the number of differentiated cells and an increase in apoptosis at the ventricular zone. No difference in mitosis rate was observed, although the mitotic angle was predominantly vertical in c-fos-/- embryos, suggesting a reduced trend of NSPCs to differentiate. At E13.5, changes in differentiation markers start to be apparent and are still clearly observed at E16.5. A tendency of more AP-1/DNA complexes present in nuclear extracts of cerebral cortex from c-fos-/- embryos with no differences in the lipid synthesis activity was found. These results suggest that c-Fos is involved in the normal development of NSPCs by means of its AP-1 activity.


Assuntos
Diferenciação Celular/genética , Genes fos/genética , Neocórtex/embriologia , Células-Tronco Neurais/citologia , Neurogênese/genética , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Embrião de Mamíferos , Imunofluorescência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Chronobiol Int ; 29(8): 1011-20, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22889436

RESUMO

Retinal ganglion cells (RGCs) contain circadian clocks driving melatonin synthesis during the day, a subset of these cells acting as nonvisual photoreceptors sending photic information to the brain. In this work, the authors investigated the temporal and light regulation of arylalkylamine N-acetyltransferase (AA-NAT) activity, a key enzyme in melatonin synthesis. The authors first examined this activity in RGCs of wild-type chickens and compared it to that in photoreceptor cells (PRs) from animals maintained for 48 h in constant dark (DD), light (LL), or regular 12-h:12-h light-dark (LD) cycle. AA-NAT activity in RGCs displayed circadian rhythmicity, with highest levels during the subjective day in both DD and LL as well as in the light phase of the LD cycle. In contrast, AA-NAT activity in PRs exhibited the typical nocturnal peak in DD and LD, but no detectable oscillation was observed under LL, under which conditions the levels were basal at all times examined. A light pulse of 30-60 min significantly decreased AA-NAT activity in PRs during the subjective night, but had no effect on RGCs during the day or night. Intraocular injection of dopamine (50 nmol/eye) during the night to mimic the effect of light presented significant inhibition of AA-NAT activity in PRs compared to controls but had no effect on RGCs. The results clearly demonstrate that the regulation of the diurnal increase in AA-NAT activity in RGCs of chickens undergoes a different control mechanism from that observed in PRs, in which the endogenous clock, light, and dopamine exhibited differential effects.


Assuntos
Arilalquilamina N-Acetiltransferase/metabolismo , Galinhas/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos da radiação , Luz , Células Ganglionares da Retina/enzimologia , Animais , Arilalquilamina N-Acetiltransferase/genética , Cegueira/genética , Cegueira/metabolismo , Galinhas/genética , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Doenças das Aves Domésticas/genética
9.
PLoS One ; 5(3): e9544, 2010 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-20209053

RESUMO

BACKGROUND: We have previously shown that the transcription factor c-Fos is also capable of associating to endoplasmic reticulum membranes (ER) and activating phospholipid synthesis. Herein we examined phospholipid synthesis status in brain tumors from human patients and from NPcis mice, an animal model of the human disease Neurofibromatosis Type 1 (NF1). PRINCIPAL FINDINGS: In human samples, c-Fos expression was at the limit of detection in non-pathological specimens, but was abundantly expressed associated to ER membranes in tumor cells. This was also observed in CNS of adult tumor-bearing NPcis mice but not in NPcis fos(-/-) KO mice. A glioblastoma multiforme and a malignant PNS tumor from a NF1 patient (MPNST) showed a 2- and 4- fold c-Fos-dependent phospholipid synthesis activation, respectively. MPNST samples also showed increased cell proliferation rates and abundant c-Fos expression. CONCLUSIONS: Results highlight a role of cytoplasmic c-Fos as an activator of phospholipid synthesis in events demanding high rates of membrane biogenesis as occurs for the exacerbated growth of tumors cells. They also disclose this protein as a potential target for controlling tumor growth in the nervous system.


Assuntos
Neoplasias do Sistema Nervoso Central/patologia , Citoplasma/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proliferação de Células , Neoplasias do Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Genótipo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurofibromatose 1/metabolismo , Fosforilação
10.
J Biol Chem ; 283(45): 31163-71, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18784083

RESUMO

It has been demonstrated that c-Fos has, in addition to its well recognized AP-1 transcription factor activity, the capacity to associate to the endoplasmic reticulum and activate key enzymes involved in the synthesis of phospholipids required for membrane biogenesis during cell growth and neurite formation. Because membrane genesis requires the coordinated supply of all its integral membrane components, the question emerges as to whether c-Fos also activates the synthesis of glycolipids, another ubiquitous membrane component. We show that c-Fos activates the metabolic labeling of glycolipids in differentiating PC12 cells. Specifically, c-Fos activates the enzyme glucosylceramide synthase (GlcCerS), the product of which, GlcCer, is the first glycosylated intermediate in the pathway of synthesis of glycolipids. By contrast, the activities of GlcCer galactosyltransferase 1 and lactosylceramide sialyltransferase 1 are essentially unaffected by c-Fos. Co-immunoprecipitation experiments in cells co-transfected with c-Fos and a V5-tagged version of GlcCerS evidenced that both proteins participate in a physical association. c-Fos expression is tightly regulated by specific environmental cues. This strict regulation assures that lipid metabolism activation will occur as a response to cell requirements thus pointing to c-Fos as an important regulator of key membrane metabolisms in membrane biogenesis-demanding processes.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Glucosiltransferases/metabolismo , Glicolipídeos/biossíntese , Neuritos/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Diferenciação Celular/fisiologia , Ativação Enzimática/fisiologia , Células PC12 , Fosfolipídeos/biossíntese , Ligação Proteica/fisiologia , Ratos , Fator de Transcrição AP-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA