Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 736
Filtrar
1.
Talanta ; 274: 125987, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38552478

RESUMO

Multidrug resistance (MDR) is a dominant challenge in cancer chemotherapy failure. The over-expression of breast cancer resistance protein (BCRP) in tumorous cells, along with its extensive substrate profile, is a leading cause of tumor MDR. Herein, on the basis of styrene maleic acid (SMA) polymer membrane protein stabilization strategy and surface plasmon resonance (SPR) biosensor, a novel high-throughput screening (HTS) system for BCRP inhibitors has been established. Firstly, LLC-PK1 and LLC-PK1/BCRP cell membranes were co-incubated with SMA polymers to construct SMA lipid particles (SMALPs). PK1-SMALPs were thus immobilized in channel 1 of the L1 chip as the reference channel, and BCRP-SMALPs were immobilized in channel 2 as the detection channel to establish the BCRP-SMALPs-SPR screening system. The methodological investigation demonstrated that the screening system was highly specific and stable. Three active compounds were screened out from 26 natural products and their affinity constants with BCRP were determined. The KD of xanthotoxin, bergapten, and naringenin were 5.14 µM, 4.57 µM, and 3.72 µM, respectively. The in vitro cell verification experiments demonstrated that xanthotoxin, bergapten, and naringenin all significantly increased the sensitivity of LLC-PK1/BCRP cells to mitoxantrone with possessing reversal BCRP-mediated MDR activity. Collectively, the developed BCRP-SMALPs-SPR screening system in this study has the advantages of rapidity, efficiency, and specificity, providing a novel strategy for the in-depth screening of BCRP inhibitors with less side effects and higher efficacy.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Maleatos , Proteínas de Neoplasias , Ressonância de Plasmônio de Superfície , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Ressonância de Plasmônio de Superfície/métodos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/análise , Humanos , Maleatos/química , Maleatos/farmacologia , Animais , Ensaios de Triagem em Larga Escala/métodos , Suínos , Poliestirenos/química , Técnicas Biossensoriais/métodos
2.
Nat Metab ; 4(5): 534-546, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35655026

RESUMO

Although the immunomodulatory and cytoprotective properties of itaconate have been studied extensively, it is not known whether its naturally occurring isomers mesaconate and citraconate have similar properties. Here, we show that itaconate is partially converted to mesaconate intracellularly and that mesaconate accumulation in macrophage activation depends on prior itaconate synthesis. When added to human cells in supraphysiological concentrations, all three isomers reduce lactate levels, whereas itaconate is the strongest succinate dehydrogenase (SDH) inhibitor. In cells infected with influenza A virus (IAV), all three isomers profoundly alter amino acid metabolism, modulate cytokine/chemokine release and reduce interferon signalling, oxidative stress and the release of viral particles. Of the three isomers, citraconate is the strongest electrophile and nuclear factor-erythroid 2-related factor 2 (NRF2) agonist. Only citraconate inhibits catalysis of itaconate by cis-aconitate decarboxylase (ACOD1), probably by competitive binding to the substrate-binding site. These results reveal mesaconate and citraconate as immunomodulatory, anti-oxidative and antiviral compounds, and citraconate as the first naturally occurring ACOD1 inhibitor.


Assuntos
Fumaratos/farmacologia , Interferons , Macrófagos , Maleatos/farmacologia , Antivirais/metabolismo , Antivirais/farmacologia , Carboxiliases , Catálise , Humanos , Inflamação/metabolismo , Macrófagos/metabolismo , Estresse Oxidativo
3.
Chem Pharm Bull (Tokyo) ; 70(6): 427-434, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35418544

RESUMO

In the present study, four novel ginsenosides fatty acid and aromatic acid derivatives were designed and synthesized, and their cytotoxic effects on human ovarian carcinoma cells (SKOV3) were assessed using the 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. The results demonstrated that all derivatives inhibited SKOV3 cell growth, and Compound 3 showed the most outstanding anti-proliferative effect on SKOV3 cells. The IC50 value of Compound 3 was 33.8 ± 2.21 µM, less than half of that of cis-platinum (70.1 ± 7.64 µM). Subsequent analysis revealed that Compound 3 could promote SKOV3 cell apoptosis, and the percentage of apoptotic cell population increased with increasing Compound 3 concentrations. In addition, the expression ratios of Bax/Bcl-2, cleaved-Caspase-3/Caspase-3 and cleaved-Caspase-9/Caspase-9 were gradually elevated in Compound 3-treated SKOV3 cells compared with control cells. Furthermore, translocation of Bax to mitochondria was associated with the release of Cytochrome C. Molecular docking analysis revealed three hydrogen-bonds existed in Compound 3 with poly(ADP-ribose)polymerase (PARP) receptor (PDB code: 5DSY), which may be the target of the anti-ovarian cancer effect of Compound 3. Altogether, our study indicates that Compound 3 induces SKOV3 cell apoptosis via reactive oxygen species (ROS)-dependent mitochondrial pathway, and can serve as an anti-cancer agent for treating ovarian carcinoma.


Assuntos
Mitocôndrias , Neoplasias Ovarianas , Sapogeninas , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Maleatos/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Simulação de Acoplamento Molecular , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Sapogeninas/farmacologia , Proteína X Associada a bcl-2/metabolismo
4.
Acta Ophthalmol ; 100(7): 788-796, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35080812

RESUMO

PURPOSE: Tyrosine kinase inhibitors inhibit VEGF receptors. If delivered to the retina, they might inhibit oedema and neovascularization such as in age-related macular degeneration and diabetic retinopathy. The aim of this study was to formulate cediranib maleate, a potent VEGF inhibitor, as γ-cyclodextrin nanoparticle eye drops and measure the retinal delivery and overall ocular pharmacokinetics after a single-dose administration in rabbits. METHODS: A novel formulation technology with 3% cediranib maleate as γ-cyclodextrin micro-suspension was prepared by autoclaving method. Suitable stabilizers were tested for heat-stable eye drops. The ophthalmic formulation was topically applied to one eye in rabbits. The pharmacokinetics in ocular tissues, tear film and blood samples were studied at 1, 3 and 6 hr after administration. RESULTS: γ-cyclodextrin formed complex with cediranib maleate. The formation of γ-cyclodextrin nanoparticles occurred in concentrated complexing media. Combined stabilizers prevented the degradation of drug during the autoclaving process. Three hours after administration of the eye drops, treated eyes showed cediranib levels of 737 ± 460 nM (mean ± SD) in the retina and 10 ± 6 nM in the vitreous humour. CONCLUSIONS: Cediranib maleate in γ-cyclodextrin nanoparticles were stable to heat in presence of stabilizers. The drug as eye drops reached the retina in concentrations that are more than 100 times higher than the 0.4 nM IC50 value reported for the VEGF type-II receptor and thus, presumably, above therapeutic level. These results suggest that γ-cyclodextrin-based cediranib maleate eye drops deliver effective drug concentrations to the retina in rabbits after a single-dose administration.


Assuntos
Ciclodextrinas , Nanopartículas , gama-Ciclodextrinas , Administração Tópica , Animais , Ciclodextrinas/metabolismo , Ciclodextrinas/farmacologia , Indóis , Maleatos/metabolismo , Maleatos/farmacologia , Soluções Oftálmicas , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas , Coelhos , Retina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , gama-Ciclodextrinas/farmacocinética
5.
Molecules ; 26(12)2021 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-34207832

RESUMO

Triple-negative breast cancer (TNBC) is a heterogeneous subtype of tumors that tests negative for estrogen receptors, progesterone receptors, and excess HER2 protein. The mainstay of treatment remains chemotherapy, but the therapeutic outcome remains inadequate. This paper investigates the potential of a duocarmycin derivative, tafuramycin A (TFA), as a new and more effective chemotherapy agent in TNBC treatment. To this extent, we optimized the chemical synthesis of TFA, and we encapsulated TFA in a micellar system to reduce side effects and increase tumor accumulation. In vitro and in vivo studies suggest that both TFA and SMA-TFA possess high anticancer effects in TNBC models. Finally, the encapsulation of TFA offered a preferential avenue to tumor accumulation by increasing its concentration at the tumor tissues by around four times in comparison with the free drug. Overall, the results provide a new potential strategy useful for TNBC treatment.


Assuntos
Antineoplásicos/farmacologia , Alcaloides Indólicos/farmacologia , Nanopartículas/química , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Alcaloides Indólicos/química , Maleatos/química , Maleatos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Poliestirenos/química , Poliestirenos/farmacologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
6.
Chem Commun (Camb) ; 57(56): 6919-6922, 2021 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-34155490

RESUMO

We demonstrate an intrinsic antitumor effect of polymer nanoparticles (P-NPs), which could re-program tumor-associated macrophages to pro-inflammatory phenotype. The intrinsic effect of P-NPs on macrophage repolarization and its combination with other therapies provide new ideas for drug delivery, macrophage regulation and immunotherapy in cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Maleatos/farmacologia , Nanopartículas/química , Poliestirenos/farmacologia , Polivinil/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Maleatos/química , Maleatos/toxicidade , Camundongos , Nanopartículas/toxicidade , Poliestirenos/química , Poliestirenos/toxicidade , Polivinil/química , Polivinil/toxicidade
7.
Int J Biol Macromol ; 165(Pt B): 2528-2540, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33098901

RESUMO

A new sponge-type hydrogel was obtained by cross-linking hyaluronic acid (HA) and poly(methylvinylether-alt-maleic acid) P(MVE-alt-MA) through a solvent-free thermal method. The sponge-type hydrogel was characterized and checked as a support for cell growth. The influence of concentration and weight ratio of polymers on the morphology and hydrogel stability was investigated. The total polymers concentration of 3% (w/w) and the weight ratio of 1:1 were optimal for the synthesis of a stable hydrogel (HA3P50) and to promote cell proliferation. The swelling measurements revealed a high-water absorption capacity of the hydrogel in basic medium. Diphenhydramine (DPH), lidocaine (Lid) and propranolol (Prop) were loaded within the hydrogel as a model drugs to investigate the ability of drug transport and release. In vitro studies revealed that HA3P50 hydrogel promoted the adhesion and proliferation of human hepatocellular carcinoma cell line HepG2, providing a good support for 3D cell culture to obtain surrogate tumor scaffold suitable for preclinical anti-cancer drug screening.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ácido Hialurônico/química , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Hidrogéis/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , Difenidramina/farmacologia , Células Hep G2 , Humanos , Ácido Hialurônico/farmacologia , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Hidrogéis/química , Lidocaína/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Maleatos/química , Maleatos/farmacologia , Propranolol/farmacologia
8.
ACS Chem Biol ; 15(4): 856-861, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32250583

RESUMO

Metabolites regulate protein function via covalent and noncovalent interactions. However, manipulating these interactions in living cells remains a major challenge. Here, we report a chemical strategy for inducing cysteine S-succination, a nonenzymatic post-translational modification derived from the oncometabolite fumarate. Using a combination of antibody-based detection and kinetic assays, we benchmark the in vitro and cellular reactivity of two novel S-succination "agonists," maleate and 2-bromosuccinate. Cellular assays reveal maleate to be a more potent and less toxic inducer of S-succination, which can activate KEAP1-NRF2 signaling in living cells. By enabling the cellular reconstitution of an oncometabolite-protein interaction with physiochemical accuracy and minimal toxicity, this study provides a methodological basis for better understanding the signaling role of metabolites in disease.


Assuntos
Cisteína/química , Fumaratos/farmacologia , Maleatos/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteoma/metabolismo , Succinatos/farmacologia , Acilação , Linhagem Celular Tumoral , Fumaratos/química , Fumaratos/toxicidade , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Maleatos/química , Maleatos/toxicidade , Fenóis/química , Proteoma/química , Proteômica/métodos , Succinatos/química , Succinatos/toxicidade , Compostos de Sulfidrila/química
9.
Cells ; 9(4)2020 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-32235396

RESUMO

The co-chaperone HspBP1 interacts with members of the hsp70 family, but also provides chaperone-independent functions. We report here novel biological properties of HspBP1 that are relevant to the formation of cytoplasmic stress granules (SGs). SG assembly is a conserved reaction to environmental or pathological insults and part of the cellular stress response. Our study reveals that HspBP1 (1) is an integral SG constituent, and (2) a regulator of SG assembly. Oxidative stress relocates HspBP1 to SGs, where it co-localizes with granule marker proteins and polyA-RNA. Mass spectrometry and co-immunoprecipitation identified novel HspBP1-binding partners that are critical for SG biology. Specifically, HspBP1 associates with the SG proteins G3BP1, HuR and TIA-1/TIAR. HspBP1 also interacts with polyA-RNA in vivo and binds directly RNA homopolymers in vitro. Multiple lines of evidence and single-granule analyses demonstrate that HspBP1 is crucial for SG biogenesis. Thus, HspBP1 knockdown interferes with stress-induced SG assembly. By contrast, HspBP1 overexpression promotes SG formation in the absence of stress. Notably, the hsp70-binding domains of HspBP1 regulate SG production in unstressed cells. Taken together, we identified novel HspBP1 activities that control SG formation. These features expand HspBP1's role in the cellular stress response and provide new mechanistic insights into SG biogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Grânulos Citoplasmáticos/metabolismo , Chaperonas Moleculares/metabolismo , Estresse Fisiológico , Animais , Grânulos Citoplasmáticos/efeitos dos fármacos , DNA Helicases/metabolismo , Proteína Semelhante a ELAV 1/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Células HeLa , Humanos , Cinética , Maleatos/farmacologia , Camundongos , Proteínas Mutantes/metabolismo , Células NIH 3T3 , Gambás , Oxidantes/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Poli A/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Ligação Proteica/efeitos dos fármacos , RNA Helicases/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Antígeno-1 Intracelular de Células T/metabolismo
10.
Pharmacol Res Perspect ; 7(6): e00540, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31832203

RESUMO

Dimethylfumarate (DMF) has long been used as part of a fixed combination of fumaric acid esters (FAE) in some European countries and is now available as an oral monotherapy for psoriasis. The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF's regulatory commitments. Although referred to in the available product labels/summary of product characteristics, the actual data have not yet been made publicly available. In vitro inhibition experiments using CYP-selective substrates with human liver microsomes showed 50% inhibitory concentrations (IC50) of >666 µmol/L for DMF and >750 µmol/L for MMF. MMF (≤250 µmol/L; 72 hours) was not cytotoxic in cultured human hepatocyte experiments and mRNA expression data indicated no CYP induction by MMF (1-250 µmol/L). DMF (≤6.66 mmol/L) showed moderate-to-high absorption (apparent permeability [Papp] ≥2.3-29.7 x 10-6 cm/s) across a Caucasian colon adenocarcinoma (Caco-2) cell monolayer, while MMF (≤7.38 mmol/L) demonstrated low-to-moderate permeability (Papp 1.2-8.9 × 10-6 cm/s). DMF was not a substrate for P-gp (net efflux ratios ≤1.22) but was a weak inhibitor of P-gp at supratherapeutic concentrations (estimated IC50 relative to solvent control of 1.5 mmol/L; [3H]digoxin efflux in Caco-2 cells). This inhibition is unlikely to be clinically relevant. MMF was not a substrate or inhibitor of P-gp. Thus, DMF and MMF should not affect the absorption, distribution, metabolism or excretion of coadministered drugs that are CYP and P-gp substrates.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Fumarato de Dimetilo/farmacologia , Fumaratos/farmacologia , Maleatos/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/antagonistas & inibidores , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Células CACO-2 , Permeabilidade da Membrana Celular , Fumarato de Dimetilo/uso terapêutico , Relação Dose-Resposta a Droga , Interações Medicamentosas , Fumaratos/uso terapêutico , Hepatócitos , Humanos , Concentração Inibidora 50 , Fígado/metabolismo , Maleatos/uso terapêutico , Microssomos Hepáticos , Psoríase/tratamento farmacológico
11.
Sci Rep ; 9(1): 16408, 2019 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-31767876

RESUMO

Detection of protein biomarkers is an important tool for medical diagnostics, typically exploiting concentration of particular biomarkers or biomarker release from tissues. We sought to establish whether proteins not normally released by living cells can be extracted without harming cells, with a view to extending this into biomarker harvest for medical diagnosis and other applications. Styrene maleic acid (SMA) is a polymer that extracts nanodiscs of biological membranes (containing membrane proteins) from cells. Hitherto it has been used to harvest SMA-lipid-membrane protein particles (SMALP) for biochemical study, by destroying the living cellular specimen. In this study, we applied SMA at low concentration to human primary cardiovascular cells and rat vascular tissue, to 'biopsy' cell proteins while avoiding significant reductions in cell viability. SMA at 6.25 parts per million harvested proteins from cells and tissues without causing significant release of cytosolic dye (calcein) or reduction in cell viability at 24 and 72 hours post-SMA (MTT assay). A wide range of proteins were recovered (20-200 kDa) and a number identified by mass spectrometry: this confirmed protein recovery from plasma membrane, intracellular membranes and cell cytosol without associated cell death. These data demonstrate the feasibility of non-lethally sampling proteins from cells, greatly extending our sampling capability, which could yield new physiological and/or pathological biomarkers.


Assuntos
Proteínas Ligadas a Lipídeos/análise , Maleatos/farmacologia , Músculo Liso Vascular/citologia , Miocárdio/citologia , Estireno/química , Animais , Morte Celular , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Coração/efeitos dos fármacos , Humanos , Maleatos/química , Espectrometria de Massas , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miocárdio/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Cultura Primária de Células , Ratos
12.
Mol Cells ; 42(8): 597-603, 2019 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-31387164

RESUMO

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a core enzyme of the aerobic glycolytic pathway with versatile functions and is associated with cancer development. Recently, Kornberg et al . published the detailed correlation between GAPDH and di- or monomethyl fumarate (DMF or MMF), which are well-known GAPDH antagonists in the immune system. As an extension, herein, we report the crystal structure of MMF-bound human GAPDH at 2.29 Å. The MMF molecule is covalently linked to the catalytic Cys152 of human GAPDH, and inhibits the catalytic activity of the residue and dramatically reduces the enzymatic activity of GAPDH. Structural comparisons between NAD+bound GAPDH and MMF-bound GAPDH revealed that the covalently linked MMF can block the binding of the NAD+ cosubstrate due to steric hindrance of the nicotinamide portion of the NAD+ molecule, illuminating the specific mechanism by which MMF inhibits GAPDH. Our data provide insights into GAPDH antagonist development for GAPDH-mediated disease treatment.


Assuntos
Fumaratos/química , Gliceraldeído-3-Fosfato Desidrogenases/química , Maleatos/química , Domínio Catalítico , Proliferação de Células/efeitos dos fármacos , Fumaratos/farmacologia , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Maleatos/farmacologia , Ligação Proteica , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos
13.
Mol Imaging Biol ; 21(6): 1107-1116, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30838549

RESUMO

PURPOSE: The system xC- transporter is upregulated in cancer cells in response to oxidative stress (OS). 5-[18F]fluoroaminosuberic acid ([18F]FASu) has been reported as a novel positron emission tomography (PET) imaging agent, targeting system xC-. The goal of this study was to evaluate the utility of [18F]FASu in monitoring cellular response to diethyl maleate (DEM) and radiation-induced OS fluctuations. PROCEDURES: [18F]FASu uptake by breast cancer cells was studied in correlation to OS biomarkers: glutathione (GSH) and reactive oxygen species (ROS), as well as transcriptional and translational levels of xCT (the functional subunit of xC-). System xC- inhibitor, sulfasalazine (SSZ), and small interfering RNA (siRNA) knockdown were used as negative controls. Radiotracer uptake was evaluated in three breast cancer models: MDA-MB-231, MCF-7, and ZR-75-1, at two-time points (1 h and 16 h) following OS induction. In vivo [18F]FASu imaging and biodistribution were performed using MDA-MB-231 xenograft-bearing mice at 16 and 24 h post-radiation treatment. RESULTS: [18F]FASu uptake was positively correlated to intracellular GSH and SLC7A11 expression levels, and radiotracer uptake was induced both by radiation treatment and by DEM at time points longer than 3 h. In an in vivo setting, there was no statistically significant uptake difference between irradiated and control tumors. CONCLUSION: [18F]FASu is a specific system xC- PET radiotracer and as such it can be used to monitor system xC- activity due to OS. As such, [18F]FASu has the potential to be used in therapy response monitoring by PET. Further optimization is required for in vivo application.


Assuntos
Sistema y+ de Transporte de Aminoácidos/metabolismo , Aminoácidos Dicarboxílicos/farmacocinética , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/radioterapia , Maleatos/farmacologia , Tomografia por Emissão de Pósitrons , Sistema y+ de Transporte de Aminoácidos/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Glutationa/metabolismo , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Fatores de Tempo , Distribuição Tecidual , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cell Death Differ ; 26(9): 1545-1565, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30770874

RESUMO

In the presence of aggregation-prone proteins, the cytosol and endoplasmic reticulum (ER) undergo a dramatic shift in their respective redox status, with the cytosol becoming more oxidized and the ER more reducing. However, whether and how changes in the cellular redox status may affect protein aggregation is unknown. Here, we show that C. elegans loss-of-function mutants for the glutathione reductase gsr-1 gene enhance the deleterious phenotypes of heterologous human, as well as endogenous worm aggregation-prone proteins. These effects are phenocopied by the GSH-depleting agent diethyl maleate. Additionally, gsr-1 mutants abolish the nuclear translocation of HLH-30/TFEB transcription factor, a key inducer of autophagy, and strongly impair the degradation of the autophagy substrate p62/SQST-1::GFP, revealing glutathione reductase may have a role in the clearance of protein aggregates by autophagy. Blocking autophagy in gsr-1 worms expressing aggregation-prone proteins results in strong synthetic developmental phenotypes and lethality, supporting the physiological importance of glutathione reductase in the regulation of misfolded protein clearance. Furthermore, impairing redox homeostasis in both yeast and mammalian cells induces toxicity phenotypes associated with protein aggregation. Together, our data reveal that glutathione redox homeostasis may be central to proteostasis maintenance through autophagy regulation.


Assuntos
Autofagia/genética , Caenorhabditis elegans/genética , Glutationa Redutase/metabolismo , Glutationa/metabolismo , Peptídeos/toxicidade , Agregação Patológica de Proteínas/metabolismo , Proteostase/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Linhagem Celular , Retículo Endoplasmático/metabolismo , Glutationa/genética , Glutationa Redutase/genética , Homeostase/efeitos dos fármacos , Homeostase/genética , Humanos , Maleatos/farmacologia , Células Musculares/metabolismo , Neurônios/metabolismo , Oxirredução/efeitos dos fármacos , Peptídeos/antagonistas & inibidores , Fenótipo , Proteólise/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
15.
J Drug Target ; 27(8): 903-916, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30615483

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that differs in progression, recurrence, and prognosis from other forms of breast cancer. The heterogeneity of TNBC has remained a challenge as no targeted therapy is currently available. Previously, we and others have demonstrated that raloxifene, a selective oestrogen receptor modulator, was also acting independently of the oestrogen receptor-α. However, raloxifene is characterised by a low bioavailability in vivo. Thus, we encapsulated raloxifene into a styrene-maleic acid (SMA) micelle to improve its pharmacokinetics. The micellar raloxifene had higher cytotoxicity when compared to the free formulation, promoted a higher cellular uptake and affected critical signalling pathways. Furthermore, SMA-raloxifene reduced TNBC tumour growth more efficiently than free raloxifene. Finally, we showed that this effect was partially mediated through oestrogen receptor-ß. In conclusion, we have provided new insight into the role of raloxifene nanoformulation in improving the management of TNBC.


Assuntos
Receptor beta de Estrogênio/metabolismo , Nanopartículas/química , Cloridrato de Raloxifeno/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Receptor beta de Estrogênio/antagonistas & inibidores , Estrogênios/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Maleatos/farmacologia , Camundongos SCID , Micelas , Distribuição Aleatória , Neoplasias de Mama Triplo Negativas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Sci Transl Med ; 10(425)2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29367345

RESUMO

Resistance to oncolytic virotherapy is frequently associated with failure of tumor cells to get infected by the virus. Dimethyl fumarate (DMF), a common treatment for psoriasis and multiple sclerosis, also has anticancer properties. We show that DMF and various fumaric and maleic acid esters (FMAEs) enhance viral infection of cancer cell lines as well as human tumor biopsies with several oncolytic viruses (OVs), improving therapeutic outcomes in resistant syngeneic and xenograft tumor models. This results in durable responses, even in models otherwise refractory to OV and drug monotherapies. The ability of DMF to enhance viral spread results from its ability to inhibit type I interferon (IFN) production and response, which is associated with its blockade of nuclear translocation of the transcription factor nuclear factor κB (NF-κB). This study demonstrates that unconventional application of U.S. Food and Drug Administration-approved drugs and biological agents can result in improved anticancer therapeutic outcomes.


Assuntos
Fumarato de Dimetilo/farmacologia , NF-kappa B/metabolismo , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Animais , Linhagem Celular Tumoral , Citocinas/biossíntese , Ésteres/farmacologia , Fumaratos/farmacologia , Glutationa/metabolismo , Humanos , Interferon Tipo I/farmacologia , Maleatos/farmacologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus Oncolíticos/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Am Chem Soc ; 140(1): 277-284, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29236489

RESUMO

The ring-opening copolymerization of maleic anhydride and propylene oxide, using a functionalized primary alcohol initiator and magnesium 2,6-di-tert-butyl phenoxide as a catalyst, was investigated in order to produce high end-group fidelity poly(propylene maleate). Subsequent isomerization of the material into 3D printable poly(propylene fumarate) was utilized to produce thin films and scaffolds possessing groups that can be modified with bioactive groups postpolymerization and postprinting. The surface concentration of these modifiable groups was determined to be 30.0 ± 3.3 pmol·cm-2, and copper-mediated azide-alkyne cycloaddition was used to attach a small molecule dye and cell adhesive GRGDS peptides to the surface as a model system. The films were then studied for cytotoxicity and found to have high cell viability before and after surface modification.


Assuntos
Fumaratos/química , Magnésio/química , Maleatos/química , Polipropilenos/química , Impressão Tridimensional , Células 3T3 , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Catálise , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fumaratos/farmacologia , Maleatos/farmacologia , Camundongos , Estrutura Molecular , Polimerização , Polipropilenos/farmacologia , Relação Estrutura-Atividade
19.
Bioorg Med Chem Lett ; 27(16): 3716-3722, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28705644

RESUMO

Hepcidin has emerged as the central regulatory molecule of systemic iron homeostasis. Inhibition of hepcidin could be a strategy favorable to treating anemia of chronic disease (ACD). We report herein the synthesis and structure-activity relationships (SARs) of a series of benzisoxazole compounds as orally active hepcidin production inhibitors. The optimization study of multi kinase inhibitor 1 led to a potent and bioavailable hepcidin production inhibitor 38 (DS79182026), which showed serum hepcidin lowering effects in a mouse IL-6 induced acute inflammatory model.


Assuntos
Benzoxazóis/química , Benzoxazóis/farmacologia , Carbamatos/química , Carbamatos/farmacologia , Hepcidinas/antagonistas & inibidores , Administração Oral , Animais , Benzoxazóis/administração & dosagem , Benzoxazóis/farmacocinética , Carbamatos/administração & dosagem , Carbamatos/farmacocinética , Regulação da Expressão Gênica/efeitos dos fármacos , Meia-Vida , Hepcidinas/genética , Hepcidinas/metabolismo , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Concentração Inibidora 50 , Interleucina-6 , Maleatos/administração & dosagem , Maleatos/química , Maleatos/farmacocinética , Maleatos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Modelos Animais , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Relação Estrutura-Atividade
20.
Biochem Biophys Res Commun ; 485(4): 802-806, 2017 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-28257840

RESUMO

Glutathione (GSH) is the most abundant non-protein thiol that buffers reactive oxygen species in the brain. GSH does not reduce nitroxides directly, but in the presence of ascorbates, addition of GSH increases ascorbate-induced reduction of nitroxides. In this study, we used electron paramagnetic resonance (EPR) imaging and the nitroxide imaging probe, 3-methoxycarbonyl-2,2,5,5-tetramethyl-piperidine-1-oxyl (MCP), to non-invasively obtain spatially resolved redox data from mouse brains depleted of GSH with diethyl maleate compared to control. Based on the pharmacokinetics of the reduction reaction of MCP in the mouse heads, the pixel-based rate constant of its reduction reaction was calculated as an index of the redox status in vivo and mapped as a "redox map". The obtained redox maps from control and GSH-depleted mouse brains showed a clear change in the brain redox status, which was due to the decreased levels of GSH in brains as measured by a biochemical assay. We observed a linear relationship between the reduction rate constant of MCP and the level of GSH for both control and GSH-depleted mouse brains. Using this relationship, the GSH level in the brain can be estimated from the redox map obtained with EPR imaging.


Assuntos
Antioxidantes/metabolismo , Encéfalo/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Glutationa/metabolismo , Animais , Ácido Ascórbico/metabolismo , Encéfalo/diagnóstico por imagem , Óxidos N-Cíclicos/administração & dosagem , Óxidos N-Cíclicos/química , Óxidos N-Cíclicos/farmacocinética , Glutationa/antagonistas & inibidores , Imageamento por Ressonância Magnética/métodos , Masculino , Maleatos/administração & dosagem , Maleatos/farmacologia , Camundongos Endogâmicos C57BL , Estrutura Molecular , Oxirredução/efeitos dos fármacos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA