Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(16)2021 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-34445392

RESUMO

Oral cancer is a major global health problem with high incidence and low survival rates. The oral cavity contains biofilms as dental plaques that harbour both Gram-negative and Gram-positive bacterial antigens, lipopolysaccharide (LPS) and lipoteichoic acid (LTA), respectively. LPS and LTA are known to stimulate cancer cell growth, and the bioactive phytochemical capsaicin has been reported to reverse this effect. Here, we tested the efficacy of oral cancer chemotherapy treatment with capsaicin in the presence of LPS, LTA or the combination of both antigens. LPS and LTA were administered to Cal 27 oral cancer cells prior to and/or concurrently with capsaicin, and the treatment efficacy was evaluated by measuring cell proliferation and apoptotic cell death. We found that while capsaicin inhibits oral cancer cell proliferation and metabolism (MT Glo assay) and increases cell death (Trypan blue exclusion assay and Caspase 3/7 expression), its anti-cancer effect was significantly reduced on cells that are either primed or exposed to the bacterial antigens. Capsaicin treatment significantly increased oral cancer cells' suppressor of cytokine signalling 3 gene expression. This increase was reversed in the presence of bacterial antigens during treatment. Our data establish a rationale for clinical consideration of bacterial antigens that may interfere with the treatment efficacy of oral cancer.


Assuntos
Antígenos de Bactérias/efeitos adversos , Capsaicina/farmacologia , Neoplasias Bucais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lipopolissacarídeos/efeitos adversos , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/microbiologia , Ácidos Teicoicos/efeitos adversos
2.
Apoptosis ; 25(1-2): 120-134, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31863325

RESUMO

Macrophages play a crucial role in several diseases' development and progression, such as in cancer and arthritis through ROS generation and inflammation. This makes macrophages a therapeutic target in these diseases. While silver nanoparticles (AgNP) have been widely used as an antibacterial and investigated as anticancer, its potential against macrophages may be limited due to its inherent oxidative mechanism. Here we encapsulated AgNP in a dipalmitoyl-phosphatidyl choline (DPPC) liposome (forming Lipo-AgNP) to suppress AgNP-induced ROS and enhance its cytotoxicity against THP1-differentiated macrophages (TDM). Our findings showed that while Lipo-AgNP had significantly more of a cytotoxic effect on TDMs (p < 0.01), it also significantly suppressed AgNP induced ROS generation and unexpectedly suppressed reduced glutathione (GSH) levels (p < 0.05) resulting in a redox imbalance in comparison to the unexposed control TDMs. Lipo-AgNP was also found to cause an increase DNA damage through H2AX histone phosphorylation and inhibition of Bcl-2 protein expression. This increased the Bax/Bcl2 ratio causing possible release of cytochrome C and subsequent caspase 3/7-dependent apoptosis. It was found that the difference between the mechanism of AgNP and Lipo-AgNP cytotoxicity may have been through the significantly increased Lipo-AgNP uptake by the TDMs as early as 30 min post-exposure (p < 0.05), changing the nanoparticle pharmacokinetic. In conclusion, the improved uptake of AgNP within the liposome caused ROS-independent caspase activation induced by Lipo-AgNP and this was facilitated by increased DNA damage, the induced redox imbalance and an increased Bax/Bcl-2 ratio.


Assuntos
Apoptose , Lipossomos/química , Macrófagos/citologia , Macrófagos/metabolismo , Nanopartículas Metálicas/química , Prata/metabolismo , Apoptose/efeitos dos fármacos , Caspase 3/genética , Caspase 3/metabolismo , Caspase 7/genética , Caspase 7/metabolismo , Linhagem Celular , Dano ao DNA/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Lipossomos/metabolismo , Macrófagos/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Prata/química , Prata/farmacologia , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
3.
Mol Pharm ; 16(2): 618-631, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30608696

RESUMO

Amphipathic, nonionic, surfactants are widely used in pharmaceutical, food, and agricultural industry to enhance product features; as pharmaceutical excipients, they are also aimed at increasing cell membrane permeability and consequently improving oral drugs absorption. Here, we report on the concentration- and time-dependent succession of events occurring throughout and subsequent exposure of Caco-2 epithelium to a "typical" nonionic surfactant (Kolliphor HS15) to provide a molecular explanation for nonionic surfactant cytotoxicity. The study shows that the conditions of surfactant exposure, which increase plasma membrane fluidity and permeability, produced rapid (within 5 min) redox and mitochondrial effects. Apoptosis was triggered early during exposure (within 10 min) and relied upon an initial mitochondrial membrane hyperpolarization (5-10 min) as a crucial step, leading to its subsequent depolarization and caspase-3/7 activation (60 min). The apoptotic pathway appears to be triggered prior to substantial surfactant-induced membrane damage (observed ≥60 min). We hence propose that the cellular response to the model nonionic surfactant is triggered via surfactant-induced increase in plasma membrane fluidity, a phenomenon akin to the stress response to membrane fluidization induced by heat shock, and consequent apoptosis. Therefore, the fluidization effect that confers surfactants the ability to enhance drug permeability may also be intrinsically linked to the propagation of their cytotoxicity. The reported observations have important implications for the safety of a multitude of nonionic surfactants used in drug delivery formulations and to other permeability enhancing compounds with similar plasma membrane fluidizing mechanisms.


Assuntos
Excipientes/efeitos adversos , Polietilenoglicóis/farmacologia , Estearatos/farmacologia , Apoptose/efeitos dos fármacos , Células CACO-2 , Caspase 3/metabolismo , Caspase 7/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Humanos , Oxirredução/efeitos dos fármacos , Polietilenoglicóis/efeitos adversos , Estearatos/efeitos adversos
4.
Drug Chem Toxicol ; 41(2): 182-187, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28669220

RESUMO

This study aimed at comparing the cytotoxicity of a new octenidine mouth rinse (MR) on gingival fibroblasts and epithelial cells using different established MRs. Octenidol (OCT), Chlorhexidine 0.2% (CHX), Meridol (MER), Oral B (OB), and control (PBS only) were used. Human primary gingival fibroblasts (HGFIBs) and human primary nasal epithelial cells (HNEPCs) were cultivated in cell-specific media (2 × 105 cells/well) and treated with a MR or PBS for 1, 5, and 15 min. All tests were performed in duplicate and repeated 12 times. The apoptosis and necrosis were determined using a Caspase-3/7 assay and LDH assay, respectively. The data were analyzed using two-way analysis of variance with subsequent Mann-Whitney U-test. No significant differences could be found between the incubation times of the MR, neither for apoptosis nor necrosis (p > 0.05). Regarding apoptosis of HGFIBs, MRs had no influence at all. In HNEPCs, OCT induced relevantly lower apoptosis than CHX (p = 0.01). Considering necrosis, MER showed the lowest numbers of necrotic HGFIBs and HNEPCs, whereas OB induced the highest number of necrotic cells. The differences between both MR were statistically relevant (p < 0.01). OCT did neither differ from the other MRs nor from the control (PBS) in induction of necrosis in both cell types. In conclusion, the slightly negative effect of OCT considering apoptosis and necrosis of HGFIBs and HNEPCs is nearly the same or even lower compared to the established MRs included in this study. The results confirm that OCT is a potential alternative to CHX.


Assuntos
Anti-Infecciosos Locais/toxicidade , Apoptose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Antissépticos Bucais/toxicidade , Mucosa Nasal/efeitos dos fármacos , Piridinas/toxicidade , Biomarcadores/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Gengiva/metabolismo , Gengiva/patologia , Humanos , Iminas , L-Lactato Desidrogenase/metabolismo , Mucosa Nasal/metabolismo , Mucosa Nasal/patologia , Necrose , Medição de Risco
5.
J Periodontal Res ; 52(3): 522-531, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27624546

RESUMO

BACKGROUND AND OBJECTIVE: Bacteria in the dental biofilm surrounding marginal gingival grooves cause periodontal diseases. Numerous bacteria within the biofilm consume nutrients from the gingival crevicular fluid. Furthermore, some gram-negative bacteria in mature dental biofilms produce butyrate. Thus, gingival epithelial cells in close proximity to mature dental biofilms are at risk of both starvation and exposure to butyrate. In the present study, we determined the combined effects of starvation and butyrate exposure on gingival epithelial cell death and the underlying mechanisms. MATERIAL AND METHODS: The Ca9-22 cell line was used as an in vitro counterpart of gingival epithelial cells. Cell death was measured as the amount of total DNA in the dead cells using SYTOX Green dye, which penetrates through membranes of dead cells and emits fluorescence when it intercalates into double-stranded DNA. AMP-activated protein kinase (AMPK) activity, the amount of autophagy, and acetylation of histone H3 were determined using western blot. Gene expression levels of microtubule-associated protein 1 light chain 3b (lc3b) were determined using quantitative reverse transcription-polymerase chain reaction. RESULTS: Butyrate-induced cell death occurred in a dose-dependent manner whether cells were starved or fed. However, the induction of cell death was two to four times higher when cells were placed under starvation conditions compared to when they were fed. Moreover, both starvation and butyrate exposure induced AMPK activity and autophagy. While AMPK inactivation resulted in decreased autophagy and butyrate-induced cell death under conditions of starvation, AMPK activation resulted in butyrate-induced cell death when cells were fed. Combined with the results of our previous report, which demonstrated butyrate-induced autophagy-dependent cell death, the results of this study suggest that the combination of starvation and butyrate exposure activates AMPK inducing autophagy and subsequent cell death. Notably, this combination markedly induced LC3B production and the induction was attenuated by AMPK inhibition. LC3B knockdown, in turn, significantly decreased butyrate-induced cell death. Therefore, AMPK-dependent LC3B induction apparently plays an important role in butyrate-induced cell death. There was a lack of correspondence between the levels of AMPK activation and LC3B induction; this may reflect the histone deacetylase-inhibitory capacity of butyrate on histone proteins. CONCLUSION: Taken together, starvation and butyrate exposure promote autophagy via AMPK signaling, while the histone deacetylase-inhibitory effects of butyrate alter chromatin to transcriptionally active state, resulting in strong LC3B induction and subsequent cell death. These findings may help improve the understanding of the cellular processes underlying periodontal disease initiation.


Assuntos
Autofagia , Butiratos/farmacologia , Células Epiteliais/fisiologia , Gengiva/fisiopatologia , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Western Blotting , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inanição/fisiopatologia
6.
Int J Mol Sci ; 18(7)2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28696356

RESUMO

Osteoarthritis (OA) is hallmarked by a progressive degradation of articular cartilage. Besides risk factors including trauma, obesity or genetic predisposition, inflammation has a major impact on the development of this chronic disease. During the course of inflammation, cytokines such as tumor necrosis factor-alpha(TNF-α) and interleukin (IL)-1ß are secreted by activated chondrocytes as well as synovial cells and stimulate the production of other inflammatory cytokines and matrix degrading enzymes. The mTORC1 inhibitor rapamycin is a clinical approved immunosuppressant and several studies also verified its chondroprotective effects in OA. However, the effect of blocking the mechanistic target of rapamycin complex (mTORC)1 on the inflammatory status within OA is not well studied. Therefore, we aimed to investigate if inhibition of mTORC1 by rapamycin can preserve and sustain chondrocytes in an inflammatory environment. Patient-derived chondrocytes were cultured in media supplemented with or without the mTORC1 inhibitor rapamycin. To establish an inflammatory environment, either TNF-α or IL-1ß was added to the media (=OA-model). The chondroprotective and anti-inflammatory effects of rapamycin were evaluated using sulfated glycosaminoglycan (sGAG) release assay, Caspase 3/7 activity assay, lactate dehydrogenase (LDH) assay and quantitative real time polymerase chain reaction (PCR). Blocking mTORC1 by rapamycin reduced the release and therefore degradation of sGAGs, which are components of the extracellular matrix secreted by chondrocytes. Furthermore, blocking mTORC1 in OA chondrocytes resulted in an enhanced expression of the main chondrogenic markers. Rapamycin was able to protect chondrocytes from cell death in an OA-model shown by reduced Caspase 3/7 activity and diminished LDH release. Furthermore, inhibition of mTORC1 preserved the chondrogenic phenotype of OA chondrocytes, but also reduced inflammatory processes within the OA-model. This study highlights that blocking mTORC1 is a new and promising approach for treating OA. Low side effects make rapamycin an attractive implementation to existing therapeutic strategies. We showed that rapamycin's chondroprotective property might be due to an interference with IL-1ß triggered inflammatory processes.


Assuntos
Condrócitos/efeitos dos fármacos , Citocinas/farmacologia , Sirolimo/farmacologia , Caspase 3/metabolismo , Caspase 7/metabolismo , Células Cultivadas , Colágeno Tipo I/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Imuno-Histoquímica , L-Lactato Desidrogenase/metabolismo , Osteoartrite/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Cytokine ; 75(1): 165-73, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25882870

RESUMO

Periodontitis is the most prevalent infectious disease caused by periodontopathic bacteria and is also a chronic inflammatory disease. Gingival crevicular fluid (GCF) is an inflammatory exudate that seeps into the gingival crevices or periodontal pockets around teeth with inflamed gingiva, and contains various materials including leukocytes and cytokines. Since gingival epithelial cells, which form a barrier against bacterial challenges, are affected by GCF, cytokines or other materials contained within GCF are engaged in the maintenance and disruption of the epithelial barrier. Accordingly, its compositional pattern has been employed as a reliable objective index of local inflammation. Transforming growth factor ß1 (TGF-ß1) levels in GCF were previously shown to be markedly higher in patients with periodontitis than in healthy subject. However, it currently remains unclear how TGF-ß1 affects gingival epithelial cell growth or apoptosis; therefore, elucidating the mechanism responsible may lead to a deeper understanding of pathogenic periodontitis. In the present study, the human gingival epithelial cell line, OBA9 cells were stimulated with recombinant TGF-ß1. Apoptosis-related protein levels were determined by Western blotting. Caspase-3/7 activity was measured with a Caspase-Glo assay kit. Surviving and apoptotic cells were detected using an MTS assay and TUNEL staining, respectively. TGF-ßRI siRNA and smad2 siRNA were transfected into cells using the lipofectamine RNAiMAX reagent. TGF-ß1 elevated caspase-3 activity and the number of TUNEL-positive apoptotic cells in OBA9 cells. Furthermore, while the levels of the pro-apoptotic proteins Bax, Bak, Bim, and Bad were increased in OBA9 cells stimulated with TGF-ß1, the TGF-ß1 treatment also decreased the levels of anti-apoptotic proteins such as Bcl-2 and Bcl-xL in a time-dependent manner. Additionally, TGF-ß1 up-regulated the protein levels of cleaved caspase-9. These results indicated that TGF-ß1-induced apoptosis was involved in a mitochondria-related intrinsic pathway. TGF-ß1 phosphorylated smad2 in OBA9 cells and this phosphorylation was clearly reduced by SB431542 (a TGF-ß type I receptor inhibitor). Consistent with this result, SB431542 or smad2 siRNA-induced reductions in smad2 protein expression levels attenuated TGF-ß1-induced apoptosis. On the other hand, the ligation of TGF-ß1 on its receptor also stimulated the phosphorylation of Erk and Akt, which are smad2-independent pathways. However, the inhibition of Erk/Akt signaling pathways by U0126, a MEK-Erk inhibitor and LY294002, a PI3Kinase-Akt inhibitor, augmented TGF-ß1-induced apoptosis in OBA9 cells. Taken together, the results of present study demonstrated that TGF-ß1 activated both the smad2 and Erk/Akt cascades via its receptor on gingival epithelial cells, even though these two pathways have opposite roles in cell death and survival, and the culmination of these signaling events induced mitochondria-dependent apoptosis in gingival epithelial cells. Based on the results of the present study, we herein proposed for the first time, that TGF-ß1 is a novel target cytokine for monitoring the progression of periodontal disease.


Assuntos
Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gengiva/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Acetilcisteína/metabolismo , Apoptose , Benzamidas/química , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Dioxóis/química , Humanos , Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases , Periodontite/metabolismo , Fosforilação , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
8.
J Mater Sci Mater Med ; 25(4): 1033-44, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24394981

RESUMO

Hydroxylation of carbon nanotubes (CNTs) can enhance their dispersibility in water, and allows the capability to conjugate with other molecules for the expected applications. However, the cytotoxicity of hydroxylated CNTs has not been thoroughly investigated. Here, we compared the cytotoxicity of hydroxylated multi-walled carbon nanotubes (MWCNTs-OH) on a human cell line with that of pristine multi-walled carbon nanotubes (p-MWCNTs). We showed that while both MWCNTs-OH and p-MWCNTs induced apoptosis in a time- and dose-dependent manner, MWCNTs-OH triggered a significantly milder cytotoxic response than that of p-MWCNTs. We further showed that such attenuated response could be attributed to a reduced disruption of the mitochondrial membrane potential (MMP), leading to the attenuation of both cytochrome c (cyt-c) release and activation of caspases. These findings suggest that MWCNTs-OH, could be more biocompatible for in vivo applications than that of p-MWCNTs by limiting the activation of the mitochondrial mediated apoptotic pathway.


Assuntos
Apoptose/efeitos dos fármacos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/toxicidade , Nanotubos de Carbono/química , Nanotubos de Carbono/toxicidade , Apoptose/fisiologia , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocromos c/metabolismo , Ativação Enzimática/efeitos dos fármacos , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Hidroxilação , Teste de Materiais , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/fisiologia , Nanotubos de Carbono/ultraestrutura , Transdução de Sinais/efeitos dos fármacos
9.
Drug Dev Ind Pharm ; 40(4): 470-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23614830

RESUMO

INTRODUCTION: Gemcitabine-loaded TSL (Gem-TSL) was used in combination with hyperthermia (HT) to treat the colon adenocarcinoma-bearing BALB/c mice for improved anticancer effect following intravenous administration. METHODS: A new temperature-sensitive liposome (TSL), composed of DPPC:DMPC:DSPC (4:1:1 molar ratio) releasing the encapsulated gemcitabine (Gem) at 41 °C, was developed and evaluated for enhanced antitumor efficacy both in vitro and in vivo. RESULTS: Drug release from the TSL was sharply increased at 41 °C and in vitro cytotoxicity of Gem-TSL in colon adenocarcinoma cells (CT-26) was 10 times higher than the free drug (IC50 = 0.3 µM versus 3 µM). Apoptosis seemed to be the main mechanism of cell death as the treatment of the cells with Gem-TSL increased the caspse-3/7 activity by 1.5-fold and also caused the fragmentation of chromatin DNA. Gem-TSL suppressed the tumor growth in CT-26-bearing BALB/c mice more stronger than the free gemcitabine after intravenous administration. Moreover, this in vivo antitumor efficacy of Gem-TSL was further increased when HT was added. DISCUSSION: This study suggests that this new TSL-Gem formulation could serve as a new chemotherapy modality together with HT.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Desoxicitidina/análogos & derivados , Hipertermia Induzida/métodos , Adenocarcinoma/patologia , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Cromatina/metabolismo , Neoplasias do Colo/patologia , Terapia Combinada , Fragmentação do DNA/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Concentração Inibidora 50 , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Temperatura , Gencitabina
10.
Dev Growth Differ ; 55(5): 615-21, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23713787

RESUMO

Apoptosis during tooth development appears dependent on the apoptotic executioner caspase-3, but not caspase-7. Instead, activated caspase-7 has been found in differentiated odontoblasts and ameloblasts, where it does not correlate with apoptosis. To further investigate these findings, the mouse incisor was used as a model. Analysis of caspase-7-deficient mice revealed a significant thinner layer of hard tissue in the adult incisor. Micro computed tomography scan confirmed this decrease in mineralized tissues. These data strongly suggest that caspase-7 might be directly involved in functional cell differentiation and regulation of the mineralization of dental matrices.


Assuntos
Ameloblastos/enzimologia , Caspase 7/metabolismo , Diferenciação Celular , Odontoblastos/enzimologia , Ameloblastos/citologia , Ameloblastos/metabolismo , Animais , Caspase 7/genética , Proliferação de Células , Esmalte Dentário/embriologia , Esmalte Dentário/crescimento & desenvolvimento , Esmalte Dentário/metabolismo , Imuno-Histoquímica , Incisivo/embriologia , Incisivo/crescimento & desenvolvimento , Incisivo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Odontoblastos/citologia , Odontoblastos/metabolismo , Odontogênese , Fatores de Tempo , Microtomografia por Raio-X
11.
Eur J Pharmacol ; 933: 175273, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36108738

RESUMO

Cisplatin is one of the most widely used chemotherapeutic agents and induces caspase-9-mediated apoptosis. Here, we examined whether phospholipase C-related catalytically inactive protein (PRIP) enhances cisplatin-induced apoptosis of breast cancer cells. PRIP depletion increased expression of X-linked inhibitor of apoptosis protein (XIAP) by inhibiting protein degradation, which is downstream of the phosphatidylinositol 3-kinase/AKT pathway and inhibits apoptotic signaling by blocking caspase-9 activation. Conversely, the viability of MCF-7 cells transfected with Prip1 was significantly lower than that of control cells in the presence of cisplatin. The number of apoptotic nuclei and expression levels of cleaved caspase-9 and downstream cleaved caspase-7 and poly-ADP ribose polymerase were greater in PRIP1-expressing MCF-7 cells treated with cisplatin than in control cells. XIAP was decreased by expression of pleckstrin homology domain of PRIP1 (PRIP1-PH domain) that blocked phosphatidylinositol 4,5 bisphosphate metabolism. In an orthotopic transplantation model, combined administration of PRIP1-PH domain-containing liposomes and cisplatin reduced the size of MCF-7 tumors compared with cisplatin alone. Our findings demonstrate that PRIP promotes XIAP degradation by inhibiting PI(3,4,5)P3/AKT signaling and enhances cisplatin-induced apoptotic cell death. Therefore, we propose that PRIP1-PH liposomes are a novel agent to avoid cisplatin resistance.


Assuntos
Cisplatino , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X , Adenosina Difosfato Ribose , Apoptose , Caspase 7/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Cisplatino/farmacologia , Humanos , Lipossomos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositóis , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfolipases Tipo C/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
12.
J Pharmacol Exp Ther ; 339(3): 779-89, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21890508

RESUMO

Many diseases and pathological conditions, including ischemia/reperfusion (I/R) injury, are the consequence of the actions of reactive oxygen species (ROS). Controlling ROS generation or its level may thus hold promise as a standard therapeutic modality for ROS-related diseases. Here, we assessed heme oxygenase-1 (HO-1), which is a crucial antioxidative, antiapoptotic molecule against intracellular stresses, for its therapeutic potential via its inducer, hemin. To improve the solubility and in vivo pharmacokinetics of hemin for clinical applications, we developed a micellar hemin by conjugating it with poly(ethylene glycol) (PEG) (PEG-hemin). PEG-hemin showed higher solubility in water and significantly prolonged plasma half-life than free hemin, which resulted from its micellar nature with molecular mass of 126 kDa in aqueous media. In a rat I/R model, administration of PEG-hemin significantly elevated HO-1 expression and enzymatic activity. This induction of HO-1 led to significantly improved liver function, reduced apoptosis and thiobarbituric acid reactive substances of the liver, and decreased inflammatory cytokine production. PEG-hemin administration also markedly improved hepatic blood flow. These results suggest that PEG-hemin exerted a significant cytoprotective effect against I/R injury in rat liver by inducing HO-1 and thus seems to be a potential therapeutic for ROS-related diseases, including I/R injury.


Assuntos
Cardiotônicos/farmacologia , Heme Oxigenase-1/biossíntese , Hemina/análogos & derivados , Hemina/uso terapêutico , Hepatopatias/tratamento farmacológico , Fígado/metabolismo , Polietilenoglicóis/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Monóxido de Carbono/sangue , Caspase 3/metabolismo , Caspase 7/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CCL2/sangue , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Indução Enzimática , Hemina/química , Hemina/farmacologia , Hepatócitos/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Circulação Hepática , Hepatopatias/metabolismo , Hepatopatias/fisiopatologia , Masculino , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Transaminases/sangue
13.
Chem Pharm Bull (Tokyo) ; 59(11): 1386-92, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22041075

RESUMO

Histone deacetylase inhibitor (HDACI), suberoylanilide hydroxamic acid (SAHA), approved by the Food and Drug Administration (FDA) for the treatment of cutaneous T cell lymphoma, is a promising new treatment strategy for various cancers. In this study, we hypothesized that a liposomal formulation of HDACI might efficiently deliver HDACI into tumors. To incorporate HDACI efficiently into the liposomal membrane, we synthesized six HDACI-lipid conjugates, in which polyethylene glycol(2000) (PEG(2000))-lipid or cholesterol (Chol) was linked with a potent hydroxamic acid, HDACI, SAHA or K-182, by cleavable linkers, such as ester, carbamide and disulfide bonds. Liposomal HDACI-lipid conjugates were prepared with distearoylphosphatidylcholine (DSPC) and HDACI-Chol conjugate or with DSPC, Chol and HDACI-PEG-lipid conjugates, and their cytotoxicities were evaluated for human cervix tumor HeLa and mouse colon tumor Colon 26 cells. Among the liposomes, liposomal oleyl-PEG(2000)-SAHA conjugated with SAHA and oleyl-PEG(2000) via a carbamate linker showed higher cytotoxicity via hyperacetylation of histone H3 and induction of caspase 3/7 activity. These results suggested that liposomal HDACI-lipid conjugates may be a potential tool for cancer therapy.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Inibidores de Histona Desacetilases/química , Lipídeos/química , Lipossomos/química , Animais , Antineoplásicos/síntese química , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Colesterol/química , Histona Desacetilases/química , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/química , Camundongos , Neoplasias , Fosfatidilcolinas/química , Polietilenoglicóis/química
14.
Toxicol In Vitro ; 69: 105005, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32956835

RESUMO

Human gingival fibroblasts (HGF) play a vital role in wound healing, oral cancer, and are among the first cells being exposed to e-cigarette vapor (eCV) or cigarette smoke (CS) during inhalation. Although the cell-damaging effect of CS has been well studied, the effects of eCV on gingival cells are still unclear. The aim of this in vitro study was to compare the effects of eCV and CS on HGF in terms of proliferation, metabolic activity, cell death, and formation of reactive oxygen species (ROS). After 24 h cell numbers in CS-exposed cells in contrast to eCV-exposed cells were significantly decreased compared to the control. At later points in time, such differences could no longer be observed. Compared to the control, HGF stimulated with eCV showed a significantly higher metabolic activity 1 h, 24 h, and 48 h after exposure. 24 h after exposure, the metabolic activity was increased in both test groups. No caspase 3/7 activation nor significant differences in the amount of apoptosis/necrosis among the groups were seen. Only in CS-exposed cells ROS formation was increased at 1 h, 3 h, and 6 h after exposition. In conclusion, when compared to conventional CS, a less harmful effect of eCV on HGF can be assumed.


Assuntos
Vapor do Cigarro Eletrônico/toxicidade , Fibroblastos/efeitos dos fármacos , Gengiva/citologia , Nicotiana , Fumaça/efeitos adversos , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Necrose/induzido quimicamente , Espécies Reativas de Oxigênio/metabolismo
15.
Chem Res Toxicol ; 22(1): 97-105, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19105591

RESUMO

We have evaluated the toxicity of four glyphosate (G)-based herbicides in Roundup formulations, from 10(5) times dilutions, on three different human cell types. This dilution level is far below agricultural recommendations and corresponds to low levels of residues in food or feed. The formulations have been compared to G alone and with its main metabolite AMPA or with one known adjuvant of R formulations, POEA. HUVEC primary neonate umbilical cord vein cells have been tested with 293 embryonic kidney and JEG3 placental cell lines. All R formulations cause total cell death within 24 h, through an inhibition of the mitochondrial succinate dehydrogenase activity, and necrosis, by release of cytosolic adenylate kinase measuring membrane damage. They also induce apoptosis via activation of enzymatic caspases 3/7 activity. This is confirmed by characteristic DNA fragmentation, nuclear shrinkage (pyknosis), and nuclear fragmentation (karyorrhexis), which is demonstrated by DAPI in apoptotic round cells. G provokes only apoptosis, and HUVEC are 100 times more sensitive overall at this level. The deleterious effects are not proportional to G concentrations but rather depend on the nature of the adjuvants. AMPA and POEA separately and synergistically damage cell membranes like R but at different concentrations. Their mixtures are generally even more harmful with G. In conclusion, the R adjuvants like POEA change human cell permeability and amplify toxicity induced already by G, through apoptosis and necrosis. The real threshold of G toxicity must take into account the presence of adjuvants but also G metabolism and time-amplified effects or bioaccumulation. This should be discussed when analyzing the in vivo toxic actions of R. This work clearly confirms that the adjuvants in Roundup formulations are not inert. Moreover, the proprietary mixtures available on the market could cause cell damage and even death around residual levels to be expected, especially in food and feed derived from R formulation-treated crops.


Assuntos
Apoptose , Glicina/análogos & derivados , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Feminino , Glicina/metabolismo , Glicina/toxicidade , Humanos , Rim/citologia , Rim/embriologia , Necrose , Placenta/citologia , Polietilenoglicóis/química , Polietilenoglicóis/toxicidade , Células-Tronco , Succinato Desidrogenase/metabolismo , Veias Umbilicais/citologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/química , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/toxicidade , Glifosato
16.
Artigo em Inglês | MEDLINE | ID: mdl-30934888

RESUMO

Because of their nephrotoxicity and presence in the environment, uranium (U) and fluoride (F) represent risks to the global population. There is a general lack of knowledge regarding the mechanisms of U and F nephrotoxicity and the underlying molecular pathways. The present study aims to compare the threshold of the appearance of renal impairment and to study apoptosis and inflammation as mechanisms of nephrotoxicity. C57BL/6J male mice were intraperitoneally treated with a single dose of U (0, 2, 4 and 5 mg/kg) or F (0, 2, 5, 7.5 and 10 mg/kg) and euthanized 72 h after. Renal phenotypic characteristics and biological mechanisms were evaluated by urine biochemistry, gene/protein expression, enzyme activity, and (immuno)histological analyses. U and F exposures induced nephrotoxicity in a dose-dependent manner, and the highest concentrations induced severe histopathological alterations as well as increased gene expression and urinary excretion of nephrotoxicity biomarkers. KIM-1 gene expression was induced starting at 2 mg/kg U and 7.5 mg/kg F, and this increase in expression was confirmed through in situ detection of this biomarker of nephrotoxicity. Both treatments induced inflammation as evidenced by cell adhesion molecule expression and in situ levels, whereas caspase 3/7-dependent apoptosis was increased only after U treatment. Overall, a single dose of F or U induced histopathologic evidence of nephrotoxicity renal impairment and inflammation in mice with thresholds under 7.5 mg/kg and 4 mg/kg, respectively.


Assuntos
Rim/efeitos dos fármacos , Fluoreto de Sódio/toxicidade , Nitrato de Uranil/toxicidade , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Receptor Celular 1 do Vírus da Hepatite A/genética , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Rim/metabolismo , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL
17.
J Med Chem ; 50(21): 5109-17, 2007 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-17887662

RESUMO

A range of substituted N-alkylisatins were synthesized and their cytotoxicity evaluated against several cancer cell lines in vitro. SAR studies indicated that the introduction of an aromatic ring with a one or three carbon atom linker at N1 enhanced the activity from that of the allyl, 2'-methoxyethyl, and 3'-methylbutyl N-substituted isatins. Furthermore, electron-withdrawing groups substituted at the meta or para position of the ring were favored over the ortho orientation. Of the 24 compounds screened, nine displayed sub-micromolar IC50 values and in general demonstrated greater selectivity toward leukemia and lymphoma cell lines over any of the carcinoma cell lines tested. 5,7-Dibromo-N-(p-methylbenzyl)isatin (6) was the most active compound, inhibiting the metabolic activity of both U937 and Jurkat cancer cell lines at 0.49 muM. Various N-alkylisatins were also found to dramatically alter lymphocyte morphology, destabilize microtubules, inhibit tubulin polymerization, induce G2/M cell cycle arrest, and activate the effector caspase-3 and -7.


Assuntos
Antineoplásicos/síntese química , Isatina/análogos & derivados , Isatina/síntese química , Antineoplásicos/farmacologia , Apoptose , Biopolímeros , Caspase 3/metabolismo , Caspase 7/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática , Humanos , Isatina/farmacologia , Linfócitos/efeitos dos fármacos , Linfócitos/patologia , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura , Estereoisomerismo , Relação Estrutura-Atividade , Tubulina (Proteína)/química
18.
J Biomed Mater Res A ; 81(1): 24-39, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17109414

RESUMO

Novel pentablock copolymers of poly(diethylaminoethylmethacrylate) (PDEAEM), poly(ethylene oxide) (PEO), and poly(propylene oxide) (PPO), (PDEAEM-b-PEO-b-PPO-b-PEO-b-PDEAEM), were synthesized as vectors for gene delivery, and were tested for their biocompatibility on SKOV3 (human ovarian carcinoma) and A431 (human epidermoid cancer) cell lines under different in vitro conditions using various assays to elucidate the mechanism of cell death. These copolymers form micelles in aqueous solutions and can be tuned for their cytotoxicity by tailoring the weight percentage of their cationic component, PDEAEM. Copolymers with higher PDEAEM content were found to be more cytotoxic, though their polyplexes were less toxic than the polycations alone. Pentablock copolymers displayed higher cell viability than commercially available ExGen 500 at similar N:P ratios. While cell death with ExGen was found to be accompanied by an early loss of cell membrane integrity, pentablock copolymers caused very little membrane leakage. Caspase-3/7 assay confirmed that none of these polymers induced apoptosis in the cells. These pentablock copolymers form thermo-reversible gels at physiological temperatures, thereby enabling controlled gene delivery. Toxicity of the polymer gels was tested using an agarose-matrix, simulating an in vivo tumor model where injected polyplex gels would dissolve to release polyplexes, diffusing through tumor mass to reach the target cells. Twenty five weight percent of copolymer gels were found to be nontoxic or mildly cytotoxic after 24 h incubation. Transfection efficiency of the copolymers was found to be critically correlated to cytotoxicity and depended on DNA dose, polymer concentration, and N:P ratios. Transgene expression obtained was comparable to that of ExGen, but ExGen exhibited greater cell death.


Assuntos
Técnicas de Transferência de Genes , Teste de Materiais , Metacrilatos/química , Micelas , Polietilenoglicóis/química , Polímeros/química , Propilenoglicóis/química , Caspase 3/metabolismo , Caspase 7/metabolismo , Morte Celular , Linhagem Celular Tumoral , DNA/química , Humanos , Metacrilatos/síntese química , Polietilenoglicóis/síntese química , Polímeros/síntese química , Propilenoglicóis/síntese química
19.
Chem Commun (Camb) ; 53(40): 5519-5522, 2017 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-28466932

RESUMO

A photo-active luminescent rhenium carbonyl complex namely, [Re(CO)3(phen)(pyAl)](CF3SO3) was grafted on a biocompatible carboxymethyl chitosan (CMC) matrix through Schiff base condensation reaction. The light-induced CO delivery from ReCMC has been shown to eradicate human colorectal adenocarcinoma cells (HT-29) very efficiently in a dose-dependent fashion. The onset of CO-induced apoptosis was realized by caspase-3,-7 detection aided by fluorescence confocal microscopy. ReCMC represents a unique example of a biocompatible and biodegradable antineoplastic agent that could find its use in cancer photopharmacology.


Assuntos
Apoptose/efeitos dos fármacos , Materiais Biocompatíveis/química , Monóxido de Carbono/toxicidade , Quitosana/análogos & derivados , Monóxido de Carbono/química , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Quitosana/química , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Complexos de Coordenação/química , Complexos de Coordenação/metabolismo , Cristalografia por Raios X , Células HT29 , Humanos , Microscopia Confocal , Conformação Molecular , Rênio/química , Espectroscopia de Infravermelho com Transformada de Fourier
20.
Oncotarget ; 8(21): 35124-35137, 2017 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-28410232

RESUMO

Glioblastomas (GBM) are the most malignant brain tumors in humans and have a very poor prognosis. New therapeutic options are urgently needed. A novel drug, Vacquinol-1 (Vac), a quinolone derivative, displays promising properties by inducing rapid cell death in GBM but not in non-transformed tissues. Features of this type of cell death are compatible with a process termed methuosis. Here we tested Vac on a highly malignant glioma cell line observed by long-term video microscopy. Human dental-pulp stem cells (DPSCs) served as controls. A major finding was that an exogenous ATP concentration of as little as 1 µM counter regulated the Vac-induced cell death. Studies using carvacrol, an inhibitor of transient receptor potential cation channel, subfamily M, member 7 (TRPM7), demonstrated that the ATP-inducible inhibitory effect is likely to be via TRPM7. Exogenous ATP is of relevance in GBM with large necrotic areas. Our results support the use of GBM cultures with different grades of malignancy to address their sensitivity to methuosis. The video-microscopy approach presented here allows decoding of signaling pathways as well as mechanisms of chemotherapeutic resistance by long-term observation. Before implementing Vac as a novel therapeutic drug in GBM, cells from each individual patient need to be assessed for their ATP sensitivity. In summary, the current investigation supports the concept of methuosis, described as non-apoptotic cell death and a promising approach for GBM treatment. Tissue-resident ATP/necrosis may interfere with this cell-death pathway but can be overcome by a natural compound, carvacrol that even penetrates the blood-brain barrier.


Assuntos
Trifosfato de Adenosina/farmacologia , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Piperidinas/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Quinolinas/farmacologia , Canais de Cátion TRPM/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Caspase 3/metabolismo , Caspase 7/metabolismo , Morte Celular , Linhagem Celular Tumoral , Cimenos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Humanos , Monoterpenos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA