Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Front Oncol ; 14: 1359652, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38454929

RESUMO

Background: Glioblastoma is one of the most aggressive primary brain tumors, with a poor outcome despite multimodal treatment. Methylation of the MGMT promoter, which predicts the response to temozolomide, is a well-established prognostic marker for glioblastoma. However, a difference in survival can still be detected within the MGMT methylated group, with some patients exhibiting a shorter survival than others, emphasizing the need for additional predictive factors. Methods: We analyzed DIAPH3 expression in glioblastoma samples from the cancer genome atlas (TCGA). We also retrospectively analyzed one hundred seventeen histological glioblastomas from patients operated on at Saint-Luc University Hospital between May 2013 and August 2019. We analyzed the DIAPH3 expression, explored the relationship between mRNA levels and Patient's survival after the surgical resection. Finally, we assessed the methylation pattern of the DIAPH3 promoter using a targeted deep bisulfite sequencing approach. Results: We found that 36% and 1% of the TCGA glioblastoma samples exhibit copy number alterations and mutations in DIAPH3, respectively. We scrutinized the expression of DIAPH3 at single cell level and detected an overlap with MKI67 expression in glioblastoma proliferating cells, including neural progenitor-like, oligodendrocyte progenitor-like and astrocyte-like states. We quantitatively analyzed DIAPH3 expression in our cohort and uncovered a positive correlation between DIAPH3 mRNA level and patient's survival. The effect of DIAPH3 was prominent in MGMT-methylated glioblastoma. Finally, we report that the expression of DIAPH3 is at least partially regulated by the methylation of three CpG sites in the promoter region. Conclusion: We propose that combining the DIAPH3 expression with MGMT methylation could offer a better prediction of survival and more adapted postsurgical treatment for patients with MGMT-methylated glioblastoma.

2.
Acta Neurochir (Wien) ; 165(4): 1075-1085, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36920664

RESUMO

BACKG ROUND: Glioblastoma is an aggressive tumor that has a dismal prognosis even with multimodal treatment. However, some patients survive longer than expected. The objective of this study was to revisit patients diagnosed with glioblastoma according to the 2021 WHO classification and analyze clinical and molecular characteristics associated with long-term survival (LTS). METHODS: We retrospectively analyzed 120 IDH-wildtype glioblastomas operated on at our institution between 2013 and 2018. We divided them into LTS patients, surviving more than 3 years, and non-LTS patients, and then compared their features. Additionally, we performed DNA methylation-based brain tumor classification in LTS patients. RESULTS: Sixteen patients were long-term survivors. Age < 70 years, MGMT promoter methylation, extent of resection ≥ 95%, and administration of radiochemotherapy were associated with LTS (P = 0.005, P < 0.001, P = 0.048, and P = 0.008, respectively). In addition, when these factors were combined, the probability of LTS was 74% (95% CI: 62--84). The methylome analysis confirmed the diagnosis of glioblastoma in the majority of the tested LTS patients. Regarding subtypes, 29% of cases were mesenchymal (MES), 43% were RTK1, and 29% were RTK2. Interestingly, RTK1 and RTK2 cases tended to have longer overall survival than MES cases (P = 0.057). Moreover, the only tested LTS patient with an unmethylated MGMT promoter had an "adult-type diffuse high-grade glioma, IDH-wildtype, subtype E" rather than a glioblastoma. This tumor was characterized by multinucleated giant cells and a somatic mutation in POLE. CONCLUSIONS: We suggest that glioblastoma patients with a combination of favorable prognostic factors can achieve LTS in 74% of cases. In addition, methylome analysis is important to ascertain the type of glioma in LTS patients, especially when the MGMT promoter is unmethylated.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Adulto , Humanos , Idoso , Glioblastoma/terapia , Glioblastoma/tratamento farmacológico , Estudos Retrospectivos , Glioma/genética , Prognóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patologia , Metilação de DNA/genética , Isocitrato Desidrogenase/genética , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética
3.
Elife ; 102021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33899739

RESUMO

Diaphanous (DIAPH) three (DIAPH3) is a member of the formin proteins that have the capacity to nucleate and elongate actin filaments and, therefore, to remodel the cytoskeleton. DIAPH3 is essential for cytokinesis as its dysfunction impairs the contractile ring and produces multinucleated cells. Here, we report that DIAPH3 localizes at the centrosome during mitosis and regulates the assembly and bipolarity of the mitotic spindle. DIAPH3-deficient cells display disorganized cytoskeleton and multipolar spindles. DIAPH3 deficiency disrupts the expression and/or stability of several proteins including the kinetochore-associated protein SPAG5. DIAPH3 and SPAG5 have similar expression patterns in the developing brain and overlapping subcellular localization during mitosis. Knockdown of SPAG5 phenocopies DIAPH3 deficiency, whereas its overexpression rescues the DIAHP3 knockdown phenotype. Conditional inactivation of Diaph3 in mouse cerebral cortex profoundly disrupts neurogenesis, depleting cortical progenitors and neurons, leading to cortical malformation and autistic-like behavior. Our data uncover the uncharacterized functions of DIAPH3 and provide evidence that this protein belongs to a molecular toolbox that links microtubule dynamics during mitosis to aneuploidy, cell death, fate determination defects, and cortical malformation.


Assuntos
Comportamento Animal , Córtex Cerebral/metabolismo , Forminas/deficiência , Microtúbulos/metabolismo , Mitose , Neurogênese , Neurônios/metabolismo , Fuso Acromático/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Comportamento Alimentar , Forminas/genética , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Humanos , Locomoção , Aprendizagem em Labirinto , Camundongos , Camundongos Knockout , Microtúbulos/genética , Microtúbulos/patologia , Células NIH 3T3 , Neurônios/patologia , Fenótipo , Comportamento Social , Fuso Acromático/genética , Fuso Acromático/patologia
5.
Cancers (Basel) ; 11(3)2019 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-30917547

RESUMO

Cisplatin (CDDP) is one of the principal chemotherapeutic agents used for the first-line treatment of many malignancies, including non-small cell lung carcinoma (NSCLC). Despite its use for over 40 years, its mechanism of action is not yet fully understood. Store-operated calcium entry (SOCE), the main pathway allowing Ca2+ entry in non-excitable cells, is involved in tumorogenesis, cancer progression and chemoresistance. It has become an attractive target in cancer treatment. In this study, we showed that siRNA-mediated depletion of stromal interaction molecule 1 (STIM1) and transient receptor potential channel 1 (TRPC1), two players of the store-operated calcium entry, dramatically reduced CDDP cytotoxicity in NSCLC cells. This was associated with an inhibition of the DNA damage response (DDR) triggered by CDDP. Moreover, STIM1 depletion also reduced CDDP-dependent oxidative stress. In parallel, SOCE activation induced Ca2+ entry into the mitochondria, a major source of reactive oxygen species (ROS) within the cell. This effect was highly decreased in STIM1-depleted cells. We then conclude that mitochondrial Ca2+ peak associated to the SOCE contributes to CDDP-induced ROS production, DDR and subsequent apoptosis. To the best of our knowledge, this is the first time that it is shown that Ca2+ signalling constitutes an initial step in CDDP-induced apoptosis.

6.
Oncotarget ; 9(22): 16059-16073, 2018 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-29662626

RESUMO

SOCE (Store-Operated Calcium Entry) is the main mechanism by which external Ca2+ enters into non-excitable cells after endoplasmic reticulum emptying. It is implicated in several processes such as proliferation and migration. Alterations in SOCE could initiate or support the development of hallmarks of cancer. In this project, we showed that disruption of the EGFR/ErbB2-dependent signalling by lapatinib and CP-724714, two inhibitors of the receptor tyrosine kinase (RTK), dramatically reduced the amplitude of the SOCE in breast cancer cells. LY294002 and MK2206, two inhibitors of the PI3K/Akt pathway, mimicked the effect of the inhibition of EGFR/ErbB2. In contrast, inhibitors of the MAPK pathway had no effect on SOCE. The involvement of EGFR/ErbB2 receptors and the PI3K/Akt pathway in the regulation of SOCE was confirmed in other cell lines derived from various cancer types. All these results showed that SOCE is positively regulated by the PI3K/Akt pathway and that this effect may be suppressed by the inhibition of the upstream RTKs. Inhibition of SOCE might therefore contribute to the anticancer effects of RTK inhibitors.

7.
Cell Calcium ; 60(6): 373-383, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27638096

RESUMO

TRP channels are involved in the control of a broad range of cellular functions such as cell proliferation and motility. We investigated the gating mechanism of TRPC1 channel and its role in U251 glioblastoma cells migration in response to chemotaxis by platelet-derived growth factor (PDGF). PDGF induced an influx of Ca2+ that was partially inhibited after pretreatment of the cells with SKI-II, a specific inhibitor of sphingosine kinase producing sphingosine-1-P (S1P). S1P by itself also induced an entry of Ca2+. Interestingly, PDGF- and S1P-induced entries of Ca2+ were lost in siRNA-TRPC1 treated cells. PDGF-induced chemotaxis of U251 cells was dramatically inhibited in cells treated with SKI-II. This effect was almost completely rescued by addition of synthetic S1P. Chemotaxis was also completely lost in siRNA-TRPC1 treated cells and interestingly, the rescue of migration of cells treated with SKI-II by S1P was dependent on the expression of TRPC1. Immunocytochemistry revealed that, in response to PDGF, TRPC1 translocated from inside of the cell to the front of migration (lamellipodes). This effect seemed PI3K dependent as it was inhibited by cell pre-treatment with LY294002, a PI3-kinase inhibitor. Our results thus identify S1P as a potential activator of TRPC1, a channel involved in cell orientation during chemotaxis by PDGF.


Assuntos
Quimiotaxia/efeitos dos fármacos , Glioblastoma/metabolismo , Lisofosfolipídeos/farmacologia , Esfingosina/análogos & derivados , Canais de Cátion TRPC/metabolismo , Cálcio/análise , Cálcio/metabolismo , Movimento Celular/efeitos dos fármacos , Glioblastoma/patologia , Humanos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Esfingosina/farmacologia , Células Tumorais Cultivadas
8.
Biochim Biophys Acta ; 1860(6): 1079-88, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26922832

RESUMO

BACKGROUND: Mitochondrial membrane permeabilisation (MMP) is classically considered as a point of no return in several forms of cell death and is involved in numerous diseases such as cancer, neurodegenerative disorders or ischemia/reperfusion injuries. Many studies established that reactive oxygen species (ROS) and Ca(2+) were the prominent inducers of MMP. However, the mechanisms connecting ROS and Ca(2+) to the players of MMP are still a matter of debate. SCOPE OF REVIEW: The aim of this review is to summarise the various studies related to the mechanisms of ROS- and Ca(2+)-induced MMP. Several lines of evidence suggest that ROS and Ca(2+) cooperate to induce MMP but the molecular details of the ROS-Ca(2+)-MMP network remain controversial. We then discuss recent data depicting this topic. MAJOR CONCLUSIONS: Cytotoxic stimuli may be transduced within the cell by ROS and Ca(2+) increases. In most models, Ca(2+) and ROS can cooperate to induce MMP. Moreover, several data suggest that MMP increases mitochondrial Ca(2+) and ROS which therefore amplify the cytotoxic signal. Intriguingly, many reports have identified players of MMP as direct ROS targets. On the contrary, direct targets of Ca(2+) remain elusive. At the same time, the mechanisms by which mitochondrial Ca(2+) overload induces ROS generation are well documented. Upon these observations, we hypothesise that Ca(2+) cannot directly induce MMP and requires ROS production as a mandatory step. GENERAL SIGNIFICANCE: Given the importance of Ca(2+)- and ROS-induced MMP in diseases, we expect that a better understanding of this process will lead to the development of novel therapies.


Assuntos
Cálcio/metabolismo , Membranas Mitocondriais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Humanos , Permeabilidade
9.
Cell Calcium ; 57(4): 312-20, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25740420

RESUMO

Reference treatment of advanced prostate cancer (PCa) relies on pharmacological or surgical androgen deprivation therapy. However, it is only temporarily efficient as tumor cells inevitably adapt to the low testosterone environment and become hormone-refractory (HRPCa). We observed that androgen removal in HRPCa-derived LNCaP cells causes different alterations in their Ca(2+) homeostasis among which a reduction of ER Ca(2+) content. We show that the decrease in [Ca(2+)]ER is due to a modest overexpression of type 1 IP3R and a threefold increased phosphorylation of IP3R1 on Ser-1716, a protein kinase A (PKA) consensus site, both implicated in ER Ca(2+) leak. Accordingly, ER Ca(2+) content was restored by siRNA-mediated down-regulation of IP3R1 or by inhibition of its phosphorylation by competition with a permeant TAT-peptide containing the Ser-1716 consensus phosphorylation sequence or by treatment with the PKA inhibitor H89. Moreover, inhibition of the IP3R1 phosphorylation by both methods sensitized the LNCaP cells to androgen deprivation-induced apoptosis. In addition, SERCA2b overexpression precluded the effect of androgen deprivation on ER Ca(2+) store content and reduced resistance to androgen deprivation. Taken together, these results indicate that lowering the ER Ca(2+)-store content by increasing IP3R1 levels and IP3R1 phosphorylation by PKA is a protective mechanism by which HRPCa-derived cells escape cell death in the absence of androgenic stimulation.


Assuntos
Androgênios/metabolismo , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Isoquinolinas/farmacologia , Masculino , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Sulfonamidas/farmacologia , Testosterona/metabolismo , Transgenes/genética , Evasão Tumoral
10.
Am J Physiol Heart Circ Physiol ; 307(8): H1120-33, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25128166

RESUMO

Exposure of cardiomyocytes to high glucose concentrations (HG) stimulates reactive oxygen species (ROS) production by NADPH oxidase (NOX2). NOX2 activation is triggered by enhanced glucose transport through a sodium-glucose cotransporter (SGLT) but not by a stimulation of glucose metabolism. The aim of this work was to identify potential therapeutic approaches to counteract this glucotoxicity. In cultured adult rat cardiomyocytes incubated with 21 mM glucose (HG), AMP-activated protein kinase (AMPK) activation by A769662 or phenformin nearly suppressed ROS production. Interestingly, glucagon-like peptide 1 (GLP-1), a new antidiabetic drug, concomitantly induced AMPK activation and prevented the HG-mediated ROS production (maximal effect at 100 nM). α2-AMPK, the major isoform expressed in cardiomyocytes (but not α1-AMPK), was activated in response to GLP-1. Anti-ROS properties of AMPK activators were not related to changes in glucose uptake or glycolysis. Using in situ proximity ligation assay, we demonstrated that AMPK activation prevented the HG-induced p47phox translocation to caveolae, whatever the AMPK activators used. NOX2 activation by either α-methyl-d-glucopyranoside, a glucose analog transported through SGLT, or angiotensin II was also counteracted by GLP-1. The crucial role of AMPK in limiting HG-mediated NOX2 activation was demonstrated by overexpressing a constitutively active form of α2-AMPK using adenoviral infection. This overexpression prevented NOX2 activation in response to HG, whereas GLP-1 lost its protective action in α2-AMPK-deficient mouse cardiomyocytes. Under HG, the GLP-1/AMPK pathway inhibited PKC-ß2 phosphorylation, a key element mediating p47phox translocation. In conclusion, GLP-1 induces α2-AMPK activation and blocks HG-induced p47phox translocation to the plasma membrane, thereby preventing glucotoxicity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Glucose/farmacologia , Hipoglicemiantes/farmacologia , Miócitos Cardíacos/metabolismo , NADPH Oxidases/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Compostos de Bifenilo , Células Cultivadas , Masculino , Glicoproteínas de Membrana/metabolismo , Metilglucosídeos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , NADPH Oxidase 2 , NADPH Oxidases/genética , Fenformin/farmacologia , Proteína Quinase C/metabolismo , Transporte Proteico , Pironas/farmacologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Tiofenos/farmacologia
11.
Prostate ; 73(10): 1090-102, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23532738

RESUMO

BACKGROUND: Treatment of advanced prostate cancer (PCa) relies on pharmacological or surgical androgen deprivation. However, it is only temporarily efficient. After a few months or years, the tumor relapses despite the absence of androgenic stimulation: a state referred to as hormone-refractory prostate cancer (HRPCa). Although autophagy confers chemoresistance in some cancers, its role in the development of HRPCa remains unknown. METHODS: Autophagic flux was assayed by GFP-LC3 clustering, by LC3-I to LC3-II conversion and transmission electron microscopy. Cell death was detected by sub-G1 quantification and concomitant measurement of transmembrane mitochondrial potential and plasma membrane permeabilization. Inhibition of autophagy was achieved by siRNAs and pharmacological inhibitors. RESULTS: Androgen deprivation or treatment with the anti-androgen bicalutamide promoted autophagy in HRPCa-derived LNCaP cells. This effect was dramatically reduced after depletion of Atg5 and Beclin-1, two canonical autophagy genes, and was associated with an inhibition of the androgen-induced mTOR pathway. The depletion of Atg5 and Beclin-1 significantly increased the level of cell death induced by androgen deprivation or bicalutamide. Finally, the safe anti-malarial drug chloroquine, an inhibitor of autophagy, dramatically increased cell death after androgen deprivation or bicalutamide treatment. CONCLUSION: Taken together, our data suggest that autophagy is a protective mechanism against androgen deprivation in HRPCa cells and that chloroquine could restore hormone dependence. This set of data could lead to the development of new therapeutic strategy against HRPCa.


Assuntos
Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Nitrilas/farmacologia , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Compostos de Tosil/farmacologia , Antagonistas de Androgênios/uso terapêutico , Anilidas/uso terapêutico , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Cloroquina/farmacologia , Humanos , Masculino , Nitrilas/uso terapêutico , Próstata/efeitos dos fármacos , Próstata/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Serina-Treonina Quinases TOR/metabolismo , Compostos de Tosil/uso terapêutico
12.
Cell Rep ; 2(2): 257-69, 2012 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-22854025

RESUMO

Patients with non-small cell lung cancer (NSCLC) are routinely treated with cytotoxic agents such as cisplatin. Through a genome-wide siRNA-based screen, we identified vitamin B6 metabolism as a central regulator of cisplatin responses in vitro and in vivo. By aggravating a bioenergetic catastrophe that involves the depletion of intracellular glutathione, vitamin B6 exacerbates cisplatin-mediated DNA damage, thus sensitizing a large panel of cancer cell lines to apoptosis. Moreover, vitamin B6 sensitizes cancer cells to apoptosis induction by distinct types of physical and chemical stress, including multiple chemotherapeutics. This effect requires pyridoxal kinase (PDXK), the enzyme that generates the bioactive form of vitamin B6. In line with a general role of vitamin B6 in stress responses, low PDXK expression levels were found to be associated with poor disease outcome in two independent cohorts of patients with NSCLC. These results indicate that PDXK expression levels constitute a biomarker for risk stratification among patients with NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Vitamina B 6/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Cisplatino/administração & dosagem , Estudos de Coortes , Intervalo Livre de Doença , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Estudo de Associação Genômica Ampla , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Piridoxal Quinase/biossíntese , Piridoxal Quinase/genética , Taxa de Sobrevida , Vitamina B 6/genética
13.
Eur J Med Chem ; 54: 834-44, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22809560

RESUMO

A series of chroman derivatives previously reported as potassium channel openers, as well as some newly synthesized simplified structures, were examined for their in vitro effects on the growth of three human high-grade glioma cell lines: U373, T98G, and Hs683. Significant in vitro growth inhibitory activity was observed with 2,2-dimethylchroman-type nitro-substituted phenylthioureas, such as compounds 4o and 4p. Interestingly, most tested phenylureas were found to be slightly less active, but more cell selective (normal versus tumor glial cells, such as 3d, 3e, and 3g), thus less toxic, than the corresponding phenylthioureas. No significant differences were observed in terms of chroman-derivative-induced growth inhibitory effects between glioma cells sensitive to pro-apoptotic stimuli (Hs683 glioma cells) and glioma cells associated with various levels of resistance to pro-apoptotic stimuli (U373 and T98G glioma cells), a feature that suggests non-apoptotic-mediated growth inhibition. Flow cytometry analyses confirmed the absence of pro-apoptotic effects for phenylthioureas and phenylureas when analyzed in U373 glioma cells and demonstrated U373 cell cycle arrest in the G0/G1 phase. Computer-assisted phase-contrast videomicroscopy revealed that 3d and 3g displayed cytostatic effects, while 3e displayed cytotoxic ones. As a result, this work identified phenylurea-type 2,2-dimethylchromans as a new class of antitumor agents to be further explored for an innovative therapeutic approach for high-grade glioma and/or for a possible new mechanism of action.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Glioblastoma/patologia , Tioureia/síntese química , Tioureia/farmacologia , Antineoplásicos/química , Antineoplásicos/toxicidade , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Canais KATP/metabolismo , Relação Estrutura-Atividade , Tioureia/química , Tioureia/toxicidade
14.
J Biol Chem ; 287(20): 16146-57, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22451676

RESUMO

TRP channels have been associated with cell proliferation and aggressiveness in several cancers. In particular, TRPC1 regulates cell proliferation and motility, two processes underlying cancer progression. We and others have described the mechanisms of TRPC1-dependent cell migration. However, the involvement of TRPC1 in cell proliferation remains unexplained. In this study, we show that siRNA-mediated TRPC1 depletion in non small cell lung carcinoma cell lines induced G(0)/G(1) cell cycle arrest resulting in dramatic decrease in cell growth. The expression of cyclins D1 and D3 was reduced after TRPC1 knockdown, pointing out the role of TRPC1 in G(1)/S transition. This was associated with a decreased phosphorylation and activation of EGFR and with a subsequent disruption of PI3K/Akt and MAPK downstream pathways. Stimulation of EGFR by its natural ligand, EGF, induced Ca(2+) release from the endoplasmic reticulum and Ca(2+) entry through TRPC1. Ca(2+) entry through TRPC1 conversely activated EGFR, suggesting that TRPC1 is a component of a Ca(2+)-dependent amplification of EGF-dependent cell proliferation.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Ciclo Celular/fisiologia , Receptores ErbB/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Canais de Cátion TRPC/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina D3/genética , Ciclina D3/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/agonistas , Receptores ErbB/genética , Técnicas de Silenciamento de Genes , Humanos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Canais de Cátion TRPC/genética
15.
Cardiovasc Res ; 92(2): 237-46, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21859816

RESUMO

AIMS: Exposure to high glucose (HG) stimulates reactive oxygen species (ROS) production by NADPH oxidase in cardiomyocytes, but the underlying mechanism remains elusive. In this study, we have dissected the link between glucose transport and metabolism and NADPH oxidase activation under hyperglycaemic conditions. METHODS AND RESULTS: Primary cultures of adult rat cardiomyocytes were exposed to HG concentration (HG, 21 mM) and compared with the normal glucose level (LG, 5 mM). HG exposure activated Rac1GTP and induced p47phox translocation to the plasma membrane, resulting in NADPH oxidase (NOX2) activation, increased ROS production, insulin resistance, and eventually cell death. Comparison of the level of O-linked N-acetylglucosamine (O-GlcNAc) residues in LG- and HG-treated cells did not reveal any significant difference. Inhibition of the pentose phosphate pathway (PPP) by 6-aminonicotinamide counteracted ROS production in response to HG but did not prevent Rac-1 upregulation and p47phox translocation leading to NOX2 activation. Modulation of glucose uptake barely affected oxidative stress and toxicity induced by HG. More interestingly, non-metabolizable glucose analogues (i.e. 3-O-methyl-D-glucopyranoside and α-methyl-D-glucopyranoside) reproduced the toxic effect of HG. Inhibition of the sodium/glucose cotransporter SGLT1 by phlorizin counteracted HG-induced NOX2 activation and ROS production. CONCLUSION: Increased glucose metabolism by itself does not trigger NADPH oxidase activation, although PPP is required to provide NOX2 with NADPH and to produce ROS. NOX2 activation results from glucose transport through SGLT1, suggesting that an extracellular metabolic signal transduces into an intracellular ionic signal.


Assuntos
Glucose/metabolismo , Hiperglicemia/enzimologia , Glicoproteínas de Membrana/metabolismo , Miócitos Cardíacos/enzimologia , NADPH Oxidases/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , 6-Aminonicotinamida/farmacologia , Acetilglucosamina/metabolismo , Animais , Morte Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Glucose/análogos & derivados , Glicosilação , Hiperglicemia/patologia , Hiperglicemia/fisiopatologia , Resistência à Insulina , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , NADPH Oxidase 2 , Via de Pentose Fosfato/efeitos dos fármacos , Florizina/farmacologia , Processamento de Proteína Pós-Traducional , Transporte Proteico , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/metabolismo
16.
Autophagy ; 7(8): 883-91, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21460612

RESUMO

Autophagic flux can be measured by determining the declining abundance of autophagic substrates such as sequestosome 1 (SQSTM1, better known as p62), which is sequestered in autophagosomes upon its direct interaction with LC3. However, the total amount of p62 results from two opposed processes, namely its synthesis (which can be modulated by some cellular stressors including autophagy inducers) and its degradation. To avoid this problem, we generated a stable cell line expressing a chimeric protein composed by p62 and the HaloTag (®) protein, which serves as a receptor for fluorescent HaloTag (®) ligands. Upon labeling with HaloTag (®) ligands (which form covalent, near-to-undissociable bonds with the Halotag (®) receptor) and washing, the resulting fluorescent labeling is not influenced by de novo protein synthesis, therefore allowing for the specific monitoring of the fusion protein decline without any interference by protein synthesis. We demonstrate that a HaloTag (®) -p62 fusion protein stably expressed in suitable cell lines can be used to monitor autophagy by flow cytometry and automated fluorescence microscopy. We surmise that this system could be adapted to high-throughput applications.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia , Citometria de Fluxo/métodos , Microscopia de Fluorescência/métodos , Linhagem Celular Tumoral , Humanos , Organelas/metabolismo , Frações Subcelulares/metabolismo , Especificidade por Substrato
17.
EMBO J ; 29(7): 1272-84, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20186124

RESUMO

Tetraploidy can constitute a metastable intermediate between normal diploidy and oncogenic aneuploidy. Here, we show that the absence of p53 is not only permissive for the survival but also for multipolar asymmetric divisions of tetraploid cells, which lead to the generation of aneuploid cells with a near-to-diploid chromosome content. Multipolar mitoses (which reduce the tetraploid genome to a sub-tetraploid state) are more frequent when p53 is downregulated and the product of the Mos oncogene is upregulated. Mos inhibits the coalescence of supernumerary centrosomes that allow for normal bipolar mitoses of tetraploid cells. In the absence of p53, Mos knockdown prevents multipolar mitoses and exerts genome-stabilizing effects. These results elucidate the mechanisms through which asymmetric cell division drives chromosomal instability in tetraploid cells.


Assuntos
Carcinoma/metabolismo , Neoplasias do Colo/metabolismo , Genes mos , Mitose , Poliploidia , Proteína Supressora de Tumor p53/metabolismo , Aneuploidia , Animais , Carcinoma/genética , Linhagem Celular Tumoral , Centrossomo/metabolismo , Instabilidade Cromossômica , Neoplasias do Colo/genética , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Proteína Supressora de Tumor p53/genética
18.
Biochem Pharmacol ; 79(9): 1221-30, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20006589

RESUMO

Increase in cytosolic calcium concentration ([Ca2+](c)), release of endoplasmic reticulum (ER) calcium ([Ca2+](er)) and ER stress have been proposed to be involved in oxidative toxicity. Nevertheless, their relative involvements in the processes leading to cell death are not well defined. In this study, we investigated whether oxidative stress generated during ascorbate-driven menadione redox cycling (Asc/Men) could trigger these three events, and, if so, whether they contributed to Asc/Men cytoxicity in MCF-7 cells. Using microspectrofluorimetry, we demonstrated that Asc/Men-generated oxidative stress was associated with a slow and moderate increase in [Ca2+](c), largely preceding permeation of propidium iodide, and thus cell death. Asc/Men treatment was shown to partially deplete ER calcium stores after 90 min (decrease by 45% compared to control). This event was associated with ER stress activation, as shown by analysis of eIF2 phosphorylation and expression of the molecular chaperone GRP94. Thapsigargin (TG) was then used to study the effect of complete [Ca2+](er) emptying during the oxidative stress generated by Asc/Men. Surprisingly, the combination of TG and Asc/Men increased ER stress to a level considerably higher than that observed for either treatment alone, suggesting that [Ca2+](er) release alone is not sufficient to explain ER stress activation during oxidative stress. Finally, TG-mediated [Ca2+](er) release largely potentiated ER stress, DNA fragmentation and cell death caused by Asc/Men, supporting a role of ER stress in the process of Asc/Men cytotoxicity. Taken together, our results highlight the involvement of ER stress and [Ca2+](er) decrease in the process of oxidative stress-induced cell death in MCF-7 cells.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Estresse Oxidativo/fisiologia , Ácido Ascórbico/farmacologia , Morte Celular , Linhagem Celular Tumoral , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Retículo Endoplasmático , Humanos , Vitamina K 3/farmacologia
19.
Biochem Pharmacol ; 76(11): 1417-25, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18617157

RESUMO

Erlotinib and gefitinib, two inhibitors of the epidermal growth factor receptor (EGFR), can stimulate apoptosis and differentiation of myeloid cell lines that lack EGFR, unveiling a novel, therapeutically exploitable off-target effect of tyrosine kinase inhibitors. Here, we performed a side-by-side comparison of erlotinib and gefitinib effects on a broad spectrum of malignant myeloid cell lines, as well as on primary myeloblasts freshly purified from the bone marrow of patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Both erlotinib and gefitinib induce apoptosis of a cell line (KG-1) that represents AML, and differentiation in another cell line (P39) derived from a patient with high-risk MDS. In this setting, erlotinib was more efficient than gefitinib. Erlotinib and gefitinib were equipotent in inducing apoptosis of primary CD34+ myeloblasts from MDS and AML patients, yet had no toxic effect on CD34+ progenitor cells from healthy donors. Although the response of individual MDS and AML patients in vitro was highly heterogeneous, the pro-apoptotic effects of erlotinib and gefitinib correlated significantly. These results suggest that erlotinib and gefitinib share a mechanistically related off-target effect that may be taken advantage of for the therapy of MDS and AML.


Assuntos
Leucemia Mieloide Aguda/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Cloridrato de Erlotinib , Feminino , Gefitinibe , Humanos , Masculino , Pessoa de Meia-Idade , Quinazolinas/farmacologia
20.
Cell Cycle ; 7(13): 1949-55, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18642442

RESUMO

Although the tumor suppressor protein p53 is a major senescence- and cell death-inducing transcription factor, recent work has clearly demonstrated that p53 has additional, extranuclear effects that contribute to its cell cycle-arresting and proapoptotic functions. Mitochondrial outer membrane permeabilization (MOMP) is (one of) the most prominent apoptotic checkpoint(s), and cytoplasmic p53 can induce MOMP by direct interactions with multidomain proteins from the Bcl-2 family present at the mitochondrial outer membrane (OM). Since MOMP is commonly disabled in cancer cells, its pharmacological induction constitutes a therapeutic goal, and this has stimulated the design of mitochondriotropic inducers of apoptosis, both inhibitors of antiapoptotic Bcl-2 family proteins (e.g., Bcl-2, Bcl-XL) or activators of their proapoptotic counterparts (e.g., Bak, Bax). Moreover, novel approaches of gene therapy have been designed in which p53 is specifically targeted to mitochondria and have been demonstrated to inhibit the growth of human cancer xenografts in immunodeficient mice. Thus, a number of distinct strategies can be employed to achieve the therapeutic induction of MOMP in cancer cells.


Assuntos
Genes p53 , Terapia Genética , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Neoplasias/terapia , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Humanos , Proteínas Mitocondriais/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Permeabilidade , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA