Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Semin Cancer Biol ; 80: 58-72, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-32070764

RESUMO

The recent advances in cancer immunotherapy confirm the crucial role of the immune system in cancer progression and treatment. Chronic inflammation and reduced immune surveillance are both features of the tumor microenvironment. Strategies aimed at reverting pro-tumor inflammation and stimulating the antitumor immune components are being actively searched, and the anticancer effects of many candidate drugs have been linked to their ability to modulate the immune system. Marine organisms constitute a rich reservoir of new bioactive molecules; some of them have already been exploited for pharmaceutical use, whereas many others are undergoing clinical or preclinical investigations for the treatment of different diseases, including cancer. In this review, we will discuss the immune-modulatory properties of marine compounds for their potential use in cancer prevention and treatment and as possible tools in the context of cancer immunotherapy.


Assuntos
Neoplasias , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Organismos Aquáticos , Humanos , Imunoterapia , Inflamação/tratamento farmacológico , Inflamação/prevenção & controle , Neoplasias/tratamento farmacológico , Microambiente Tumoral
3.
Mar Drugs ; 19(3)2021 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-33800819

RESUMO

Chemical investigation of the South-Pacific marine sponge Suberea clavata led to the isolation of eight new bromotyrosine metabolites named subereins 1-8 (2-9) along with twelve known co-isolated congeners. The detailed configuration determination of the first representative major compound of this family 11-epi-fistularin-3 (11R,17S) (1) is described. Their chemical characterization was achieved by HRMS and integrated 1D and 2D NMR (nuclear magnetic resonance) spectroscopic studies and extensive comparison with literature data. For the first time, a complete assignment of the absolute configurations for stereogenic centers C-11/17 of the known members (11R,17S) 11-epi-fistularin-3 (1) and 17-deoxyfistularin-3 (10) was determined by a combination of chemical modifications, Mosher's technology, and ECD spectroscopy. Consequently, the absolute configurations of all our new isolated compounds 2-9 were determined by the combination of NMR, Mosher's method, ECD comparison, and chemical modifications. Interestingly, compounds 2-7 were obtained by chemical transformation of the major compound 11-epi-fistularin-3 (1). Evaluation for acetylcholinesterase inhibition (AChE), DNA methyltransferase 1 (DNMT1) modulating activity and antifouling activities using marine bacterial strains are also presented.


Assuntos
Poríferos/metabolismo , Tirosina/análogos & derivados , Animais , Incrustação Biológica/prevenção & controle , Inibidores da Colinesterase/química , Inibidores da Colinesterase/isolamento & purificação , Inibidores da Colinesterase/farmacologia , DNA (Citosina-5-)-Metiltransferase 1/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Espectroscopia de Ressonância Magnética , Oceano Pacífico , Tirosina/química , Tirosina/isolamento & purificação , Tirosina/farmacologia
4.
Clin Epigenetics ; 11(1): 68, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-31060628

RESUMO

BACKGROUND: DNA methyltransferases (DNMTs) are epigenetic enzymes involved in embryonic development, cell differentiation, epithelial to mesenchymal transition, and control of gene expression, whose overexpression or enhanced catalytic activity has been widely reported in cancer initiation and progression. To date, two DNMT inhibitors (DNMTi), 5-azacytidine (5-AZA) and 5-aza-2'-deoxycytidine (DAC), are approved for the treatment of myelodysplastic syndromes and acute myeloid leukemia. Nevertheless, they are chemically instable and quite toxic for healthy cells; thus, the discovery of novel DNMTi is urgent. RESULTS: Here, we report the identification of a new quinoline-based molecule, MC3353, as a non-nucleoside inhibitor and downregulator of DNMT. This compound was able, in promoter demethylating assays, to induce enhanced green fluorescence protein (EGFP) gene expression in HCT116 cells and transcription in a cytomegalovirus (CMV) promoter-driven luciferase reporter system in KG-1 cells. Moreover, MC3353 displayed a strong antiproliferative activity when tested on HCT116 colon cancer cells after 48 h of treatment at 0.5 µM. At higher doses, this compound provided a cytotoxic effect in double DNMT knockout HCT116 cells. MC3353 was also screened on a different panel of cancer cells (KG-1 and U-937 acute myeloid leukemia, RAJI Burkitt's lymphoma, PC-3 prostate cancer, and MDA-MB-231 breast cancer), where it arrested cell proliferation and reduced viability after 48 h of treatment with IC50 values ranging from 0.3 to 0.9 µM. Compared to healthy cell models, MC3353 induced apoptosis (e.g., U-937 and KG-1 cells) or necrosis (e.g., RAJI cells) at lower concentrations. Importantly, together with the main DNMT3A enzyme inhibition, MC3353 was also able to downregulate the DNMT3A protein level in selected HCT116 and PC-3 cell lines. Additionally, this compound provided impairment of the epithelial-to-mesenchymal transition (EMT) by inducing E-cadherin while reducing matrix metalloproteinase (MMP2) mRNA and protein levels in PC-3 and HCT116 cells. Last, tested on a panel of primary osteosarcoma cell lines, MC3353 markedly inhibited cell growth with low single-digit micromolar IC50 ranging from 1.1 to 2.4 µM. Interestingly, in Saos-2 osteosarcoma cells, MC3353 induced both expression of genes and mineralized the matrix as evidence of osteosarcoma to osteoblast differentiation. CONCLUSIONS: The present work describes MC3353 as a novel DNMTi displaying a stronger in cell demethylating ability than both 5-AZA and DAC, providing re-activation of the silenced ubiquitin C-terminal hydrolase L1 (UCHL1) gene. MC3353 displayed dose- and time-dependent antiproliferative activity in several cancer cell types, inducing cell death and affecting EMT through E-cadherin and MMP2 modulation. In addition, this compound proved efficacy even in primary osteosarcoma cell models, through the modulation of genes involved in osteoblast differentiation.


Assuntos
Aminoquinolinas/síntese química , Aminoquinolinas/farmacologia , DNA-Citosina Metilases/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Neoplasias/metabolismo , Aminoquinolinas/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Metilação de DNA , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/química , Epigênese Genética , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Neoplasias/tratamento farmacológico , Pirimidinas/química
5.
Free Radic Biol Med ; 134: 177-189, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30639617

RESUMO

Redox changes and generation of reactive oxygen species (ROS) are part of normal cell metabolism. While low ROS levels are implicated in cellular signaling pathways necessary for survival, higher levels play major roles in cancer development as well as cell death signaling and execution. A role for redox changes in apoptosis has been long established; however, several new modalities of regulated cell death have been brought to light, for which the importance of ROS production as well as ROS source and targets are being actively investigated. In this review, we summarize recent findings on the role of ROS and redox changes in the activation and execution of two major forms of regulated cell death, necroptosis and ferroptosis. We also discuss the potential of using modulators of these two forms of cell death to exacerbate ROS as a promising anticancer therapy.


Assuntos
Ferroptose , Necroptose , Neoplasias/patologia , Animais , Humanos , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
6.
Mar Drugs ; 16(12)2018 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-30572618

RESUMO

Treatment of acute myeloid leukemia (AML) patients is still hindered by resistance and relapse, resulting in an overall poor survival rate. Recently, combining specific B-cell lymphoma (Bcl)-2 inhibitors with compounds downregulating myeloid cell leukemia (Mcl)-1 has been proposed as a new effective strategy to eradicate resistant AML cells. We show here that 1(R), 6(S), 1'(R), 6'(S), 11(R), 17(S)-fistularin-3, a bromotyrosine compound of the fistularin family, isolated from the marine sponge Suberea clavata, synergizes with Bcl-2 inhibitor ABT-199 to efficiently kill Mcl-1/Bcl-2-positive AML cell lines, associated with Mcl-1 downregulation and endoplasmic reticulum stress induction. The absolute configuration of carbons 11 and 17 of the fistularin-3 stereoisomer was fully resolved in this study for the first time, showing that the fistularin we isolated from the marine sponge Subarea clavata is in fact the (+)-11(R), 17(S)-fistularin-3 stereoisomer keeping the known configuration 1(R), 6(S), 1'(R), and 6'(S) for the verongidoic acid part. Docking studies and in vitro assays confirm the potential of this family of molecules to inhibit DNA methyltransferase 1 activity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Isoxazóis/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Sulfonamidas/farmacologia , Tirosina/análogos & derivados , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células HL-60 , Humanos , Isoxazóis/administração & dosagem , Isoxazóis/química , Isoxazóis/isolamento & purificação , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Simulação de Acoplamento Molecular , Poríferos/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulfonamidas/administração & dosagem , Tirosina/administração & dosagem , Tirosina/química , Tirosina/isolamento & purificação , Tirosina/farmacologia , Células U937
7.
Oncotarget ; 7(17): 24027-49, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27006469

RESUMO

We characterized the brominated alkaloid Isofistularin-3 (Iso-3), from the marine sponge Aplysina aerophoba, as a new DNA methyltransferase (DNMT)1 inhibitor. Docking analysis confirmed our in vitro DNMT inhibition data and revealed binding of Iso-3 within the DNA binding site of DNMT1. Subsequent increased expression of tumor suppressor gene aryl hydrocarbon receptor (AHR) could be correlated to decreased methylation of CpG sites within the essential Sp1 regulatory region of its promoter. Iso-3 induced growth arrest of cancer cells in G0/G1 concomitant with increased p21 and p27 expression and reduced cyclin E1, PCNA and c-myc levels. Reduced proliferation was accompanied by morphological changes typical of autophagy revealed by fluorescent and transmission electron microscopy and validated by LC3I-II conversion. Furthermore, Iso-3 strongly synergized with tumor-necrosis-factor related apoptosis inducing ligand (TRAIL) in RAJI [combination index (CI) = 0.22] and U-937 cells (CI = 0.21) and increased TRAIL-induced apoptosis via a mechanism involving reduction of survivin expression but not of Bcl-2 family proteins nor X-linked inhibitor of apoptosis protein (XIAP). Iso-3 treatment decreased FLIPL expression and triggered activation of endoplasmatic reticulum (ER) stress with increased GRP78 expression, eventually inducing TRAIL receptor death receptor (DR)5 surface expression. Importantly, as a potential candidate for further anticancer drug development, Iso-3 reduced the viability, colony and in vivo tumor forming potential without affecting the viability of PBMCs from healthy donors or zebrafish development.


Assuntos
Alcaloides/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Alcaloides/química , Animais , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais , Proliferação de Células/efeitos dos fármacos , Descoberta de Drogas , Chaperona BiP do Retículo Endoplasmático , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Poríferos/química , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Células Tumorais Cultivadas , Peixe-Zebra/metabolismo
8.
Curr Top Med Chem ; 16(7): 745-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26303418

RESUMO

Despite considerable scientific progress, the burden of cancer in our society remains a major public health problem. Tumorigenesis is recognized as a complex and multistep process that involves the accumulation of successive transformational events with multi-factorial etiology. Nevertheless, such events result in the acquisition of key hallmark characteristics that are shared by all cancer cells. Accumulating evidence indicates that, besides genetic alterations, epigenetic mechanisms (heritable changes in gene expression caused by modifications in chromatin structure without alterations of DNA sequence) are implicated in the acquisition of malignant phenotype. The potential reversibility of epigenetic alterations linked to tumorigenesis offers a promising avenue for therapeutic intervention. This review focuses on the epigenetic regulation of the cancer hallmarks and the foreseeable use of epigenetic drugs to target these features as a promising strategy for anti-cancer therapy. Based on this body of evidence, we believe that epigenetic deregulations can affect virtually all cell functions and therefore therapeutic approaches with epigenetic drugs could allow multi-target approach against the hallmarks of cancer.


Assuntos
Antineoplásicos/uso terapêutico , Epigênese Genética/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Medicina de Precisão/tendências , Animais , Metilação de DNA , Inibidores de Histona Desacetilases/uso terapêutico , Histonas/efeitos dos fármacos , Histonas/genética , Humanos
10.
J Med Chem ; 57(3): 701-13, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24387159

RESUMO

DNA methyltransferases (DNMTs) are important enzymes involved in epigenetic control of gene expression and represent valuable targets in cancer chemotherapy. A number of nucleoside DNMT inhibitors (DNMTi) have been studied in cancer, including in cancer stem cells, and two of them (azacytidine and decitabine) have been approved for treatment of myelodysplastic syndromes. However, only a few non-nucleoside DNMTi have been identified so far, and even fewer have been validated in cancer. Through a process of hit-to-lead optimization, we report here the discovery of compound 5 as a potent non-nucleoside DNMTi that is also selective toward other AdoMet-dependent protein methyltransferases. Compound 5 was potent at single-digit micromolar concentrations against a panel of cancer cells and was less toxic in peripheral blood mononuclear cells than two other compounds tested. In mouse medulloblastoma stem cells, 5 inhibited cell growth, whereas related compound 2 showed high cell differentiation. To the best of our knowledge, 2 and 5 are the first non-nucleoside DNMTi tested in a cancer stem cell line.


Assuntos
Aminoquinolinas/síntese química , Antineoplásicos/síntese química , Benzamidas/síntese química , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , Pirimidinas/síntese química , Quinolinas/síntese química , Aminoquinolinas/química , Aminoquinolinas/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Benzamidas/química , Benzamidas/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Pirimidinas/química , Pirimidinas/farmacologia , Quinolinas/química , Quinolinas/farmacologia , Relação Estrutura-Atividade
12.
Biochimie ; 94(11): 2264-79, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22627380

RESUMO

Epigenetic alterations are involved in every step of carcinogenesis. The development of chromatin-modifying agents (CMAs) has provided the ability to fight cancer by reversing these alterations. Currently, four CMAs have been approved for cancer treatment; two DNA demethylating agents and two deacetylase inhibitors. A number of promising CMAs are undergoing clinical trials in several cancer types. Moreover, already approved CMAs are still under clinical investigation to improve their efficacy and to extend their use to a broader panel of cancers. Combinatorial treatments with CMAs are already considered a promising strategy to improve clinical benefits and to limit side effects. The real mechanisms by which these CMAs allow the improvement and remission of patients are still obscure. A deeper analysis of the molecular features expressed by responding patients should be performed to reveal this information. In this review, we focus on clinical trials with CMAs, discussing the success and the pitfalls of this new class of anti-cancer drugs.


Assuntos
Antineoplásicos/farmacologia , Cromatina/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Animais , Antineoplásicos/uso terapêutico , Cromatina/genética , Cromatina/metabolismo , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Neoplasias/enzimologia
13.
Epigenomics ; 3(5): 581-609, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22126248

RESUMO

Leukemogenesis is a multistep process in which successive transformational events enhance the ability of a clonal population arising from hematopoietic progenitor cells to proliferate, differentiate and survive. Clinically and pathologically, leukemia is subdivided into four main categories: chronic lymphocytic leukemia, chronic myeloid leukemia, acute lymphocytic leukemia and acute myeloid leukemia. Leukemia has been previously considered only as a genetic disease. However, in recent years, significant advances have been made in the elucidation of the leukemogenesis-associated processes. Thus, we have come to understand that epigenetic alterations including DNA methylation, histone modifications and miRNA are involved in the permanent changes of gene expression controlling the leukemia phenotype. In this article, we will focus on the epigenetic defects associated with leukemia and their implications as biomarkers for diagnostic, prognostic and therapeutic applications.


Assuntos
Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Código das Histonas/fisiologia , Leucemia/fisiopatologia , MicroRNAs/metabolismo , Modelos Biológicos , Fenótipo , Genes Supressores de Tumor/fisiologia , Humanos , Leucemia/diagnóstico , Leucemia/terapia
14.
J Cell Mol Med ; 15(9): 1833-46, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21435179

RESUMO

Histone deacetylases (HDACs) are important regulators of gene expression. Specific structural features and distinct regulative mechanisms rationalize the separation of the 18 different human HDACs into four classes. The class II comprises a heterogeneous group of nuclear and cytosolic HDACs involved in the regulation of several cellular functions, not just limited to transcriptional repression. In particular, HDAC4, 5, 7 and 9 belong to the subclass IIa and share many transcriptional partners, including members of the MEF2 family. Genetic studies in mice have disclosed the fundamental contribution of class IIa HDACs to specific developmental/differentiation pathways. In this review, we discuss about the recent literature, which hints a role of class IIa HDACs in the development, growth and aggressiveness of cancer cells.


Assuntos
Diferenciação Celular , Transformação Celular Neoplásica/patologia , Histona Desacetilases/metabolismo , Animais , Histona Desacetilases/química , Humanos , Modelos Biológicos
15.
Mol Biol Cell ; 22(2): 278-89, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21118993

RESUMO

HDAC4 (histone deacetylase 4) belongs to class IIa of histone deacetylases, which groups important regulators of gene expression, controlling pleiotropic cellular functions. Here we show that, in addition to the well-defined nuclear/cytoplasmic shuttling, HDAC4 activity is modulated by the ubiquitin-proteasome system. Serum starvation elicits the poly-ubiquitination and degradation of HDAC4 in nontransformed cells. Phosphorylation of serine 298 within the PEST1 sequence plays an important role in the control of HDAC4 stability. Serine 298 lies within a glycogen synthase kinase 3ß consensus sequence, and removal of growth factors fails to trigger HDAC4 degradation in cells deficient in this kinase. GSK3ß can phosphorylate HDAC4 in vitro, and phosphorylation of serine 302 seems to play the role of priming phosphate. We have also found that HDAC4 modulates random cell motility possibly through the regulation of KLF2 transcription. Apoptosis, autophagy, cell proliferation, and growth arrest were unaffected by HDAC4. Our data suggest a link between regulation of HDAC4 degradation and the control of cell motility as operated by growth factors.


Assuntos
Movimento Celular , Quinase 3 da Glicogênio Sintase/metabolismo , Histona Desacetilases/metabolismo , Proteínas Repressoras/metabolismo , Ubiquitina/metabolismo , Animais , Linhagem Celular , Meios de Cultura Livres de Soro , Inibidores de Cisteína Proteinase/farmacologia , Glicogênio Sintase Quinase 3 beta , Histona Desacetilases/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Leupeptinas/farmacologia , Camundongos , Mutagênese Sítio-Dirigida , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Ubiquitinação
16.
J Cell Mol Med ; 14(4): 970-81, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20569277

RESUMO

Curcumin is a non-toxic polyphenol with pleiotropic activities and limited bioavailability. We investigated whether a brief exposure to low doses of curcumin would induce in the myogenic C2C12 cell line an endoplasmic reticulum (ER) stress response and protect against oxidative stress. A 3-hr curcumin administration (5-10 microM) increased protein levels of the ER chaperone Grp94, without affecting those of Grp78, calreticulin and haeme-oxygenase-1 (HO-1). Exposure of cells to hydrogen peroxide 24 hrs after the curcumin treatment decreased caspase-12 activation, total protein oxidation and translocation of NF-kappaB to the nucleus, compared with untreated cells. Grp94 overexpression, achieved by means of either stable or transient trasfection, induced comparable cytoprotective effects to hydrogen peroxide. The delayed cytoprotection induced by curcumin acted through Grp94, because the curcumin-induced increase in Grp94 expression was hampered by either stable or transient transfection with antisense cDNA; in these latter cells, the extent of total protein oxidation, as well as the translocation of NF-kappaB to the nucleus, and the percentage of apoptotic cells were comparable to those observed in both curcumin-untreated wild-type and empty vector transfected cells. Defining the mechanism(s) by which Grp94 exerts its antioxidant defence, the determination of cytosolic calcium levels in C2C12 cells by fura-2 showed a significantly reduced amount of releasable calcium from intracellular stores, both in conditions of Grp94 overexpression and after curcumin pre-treatment. Therefore, a brief exposure to curcumin induces a delayed cytoprotection against oxidative stress in myogenic cells by increasing Grp94 protein level, which acts as a regulator of calcium homeostasis.


Assuntos
Antioxidantes/metabolismo , Curcumina/farmacologia , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Membrana/metabolismo , Mioblastos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Caspase 12/metabolismo , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citoproteção/efeitos dos fármacos , DNA Complementar/metabolismo , Regulação para Baixo/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Peróxido de Hidrogênio/farmacologia , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , NF-kappa B/metabolismo , Oxirredução/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Transfecção
17.
Mol Cancer Ther ; 8(11): 3140-50, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19887551

RESUMO

The regulation of the necrotic death and its relevance in anticancer therapy are largely unknown. Here, we have investigated the proapoptotic and pronecrotic activities of two ubiquitin-proteasome system inhibitors: bortezomib and G5. The present study points out that the glioblastoma cell lines U87MG and T98G are useful models to study the susceptibility to apoptosis and necrosis in response to ubiquitin-proteasome system inhibitors. U87MG cells show resistance to apoptosis induced by bortezomib and G5, but they are more susceptible to necrosis induced by G5. Conversely, T98G cells are more susceptible to apoptosis induced by both inhibitors but show some resistance to G5-induced necrosis. No overt differences in the induction of Noxa and Mcl-1 or in the expression levels of other components of the apoptotic machinery were observed between U87MG and T98G cells. Instead, by comparing the transcriptional profiles of the two cell lines, we have found that the resistance to G5-induced necrosis could arise from differences in glutathione synthesis/utilization and in the microenvironment. In particular, collagen IV, which is highly expressed in T98G cells, and fibronectin, whose adhesive function is counteracted by tenascin-C in U87MG cells, can restrain the necrotic response to G5. Collectively, our results provide an initial characterization of the molecular signals governing cell death by necrosis in glioblastoma cell lines.


Assuntos
Ácidos Borônicos/farmacologia , Caspases/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Inibidores de Proteases/farmacologia , Inibidores de Proteassoma , Piranos/farmacologia , Pirazinas/farmacologia , Compostos de Sulfidrila/farmacologia , Ubiquitina/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Bortezomib , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Glioblastoma/enzimologia , Glioblastoma/genética , Glutationa/deficiência , Glutationa/metabolismo , Humanos , Análise em Microsséries , Necrose , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Ubiquitina/metabolismo
18.
Anal Chem ; 81(23): 9590-8, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19873978

RESUMO

Recombinant fluorescent probes allow the detection of molecular events inside living cells. Many of them exploit the intracellular space to provide positional signals and, thus, require detection by single cell imaging. We describe here a novel strategy based on probes capable of encoding the spatial dimension of intracellular signals into "all-or-none" fluorescence intensity changes (differential anchorage probes, DAPs). The resulting signals can be acquired in single cells at high throughput by automated flow cytometry, (i) bypassing image acquisition and analysis, (ii) providing a direct quantitative readout, and (iii) allowing the exploration of large experimental series. We illustrate our purpose with DAPs for Bax and the effector caspases 3 and 7, which are keys players in apoptotic cell death, and show applications in basic research, high content multiplexed library screening, compound characterization, and drug profiling.


Assuntos
Corantes Fluorescentes/metabolismo , Espaço Intracelular/metabolismo , Proteínas Recombinantes/metabolismo , Sequência de Aminoácidos , Animais , Apoptose , Caspase 3/metabolismo , Caspase 7/metabolismo , Ciclo Celular , Morte Celular , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Proliferação de Células , Descoberta de Drogas , Citometria de Fluxo , Ensaios de Triagem em Larga Escala , Humanos , Imagem Molecular , Dados de Sequência Molecular , Peptídeo Hidrolases/metabolismo , Proteínas Recombinantes/química , Proteína X Associada a bcl-2/metabolismo
19.
J Cell Mol Med ; 13(9B): 3358-69, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19382908

RESUMO

We have previously shown that familial Alzheimer's disease mutants of presenilin-2 (PS2) and, to a lesser extent, of presenilin-1 (PS1) lower the Ca(2+) concentration of intracellular stores. We here examined the mechanism by which wild-type and mutant PS2 affect store Ca(2+) handling. By using HeLa, SH-SY5Y and MEFs as model cells, and recombinant aequorins as Ca(2+) probes, we show evidence that transient expression of either wild-type or mutant PS2 increases the passive Ca(2+) leakage: both ryanodine- and IP(3)-receptors contribute to Ca(2+) exit out of the ER, whereas the ribosome translocon complex is not involved. In SH-SY5Y cells and MEFs, wild-type and mutant PS2 potently reduce the uptake of Ca(2+) inside the stores, an effect that can be counteracted by over-expression of SERCA-2B. On this line, in wild-type MEFs, lowering the endogenous level of PS2 by RNA interference, increases the Ca(2+)-loading capability of intracellular stores. Furthermore, we show that in PS double knockout MEFs, reduction of Ca(2+) stores is mimicked by the expression of PS2-D366A, a loss-of-function mutant, uncleaved because also devoid of presenilinase activity but not by co-expression of the two catalytic active fragments of PS2. In summary, both physiological and increased levels of wild-type and mutant PS2 reduce the Ca(2+) uptake by intracellular stores. To exert this newly described function, PS2 needs to be in its full-length form, even if it can subsequently be cleaved.


Assuntos
Cálcio/metabolismo , Presenilina-2/metabolismo , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Células HeLa , Humanos , Cinética , Proteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Mutação , Neurônios/metabolismo , Plasmídeos/metabolismo , Interferência de RNA
20.
Biochim Biophys Acta ; 1783(8): 1551-60, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18424271

RESUMO

gamma-Secretase mediates the intramembranous proteolysis of amyloid precursor protein (APP), Notch and other cellular substrates and is considered a prime pharmacological target in the development of therapeutics for Alzheimer's disease (AD). We describe here an efficient, new, simple, sensitive and rapid assay to quantify gamma-secretase activity in living cells by flow cytometry using two membrane-bound fluorescent probes, APP-GFP or C99-GFP, as substrates for gamma-secretase. The principle of the assay is based on the fact that the soluble intracellular domain of GFP-tagged APP (AICD-GFP) is released from the membrane into the cytosol following gamma-secretase cleavage. Using this feature, enzymatic activity of gamma-secretase could be deduced from the extent of the membrane retention of the probe observed after plasma membrane permeabilization and washout of the cleaved fraction. By applying two well-known gamma-secretase inhibitors (DAPT and L-685,458), we validated our assay showing that the positional GFP-based probes for gamma-secretase activity behave properly when expressed in different cell lines, providing the basis for the further development of a high-throughput and high content screening for AD targeted drug discovery. Moreover, by co-expression of different familial AD-linked mutated forms of presenilin--the key component of the gamma-secretase complex--in cells devoid of any endogenous gamma-secretase, our method allowed us to evaluate in situ the contribution of different presenilin variants to the modulation of the enzyme.


Assuntos
Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/análise , Citometria de Fluxo , Mutação , Presenilinas/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Linhagem Celular , Corantes Fluorescentes/análise , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Proteínas Recombinantes/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...