Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Biochem J ; 441(1): 367-77, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21883094

RESUMO

MTF-1 (metal-responsive transcription factor 1) is an essential mammalian protein for embryonic development and modulates the expression of genes involving in zinc homoeostasis and responding to oxidative stress. We report in the present paper that PTEN (phosphatase and tensin homologue deleted on chromosome 10) associates with MTF-1 in the cells. These two proteins interact via the acidic domain of MTF-1 and the phosphatase/C2 domain of PTEN. Depletion of PTEN reduced MT (metallothionein) gene expression and increased cellular sensitivity to cadmium toxicity. PTEN did not alter the nuclear translocation, protein stability or DNA-binding activity of MTF-1. Zinc increased MTF-1-PTEN interaction in a dose-dependent manner. The interaction elevated within 2 h of zinc addition and declined afterwards in the cells. The enhanced binding activity occurred mainly in the cytoplasm and reduced after translocating the MTF-1 into the nucleus. Blocking signalling through the PI3K (phosphoinositide 3-kinase) pathway did not alter the zinc-induced MT expression. Analysis of enzymatically inactive PTEN mutants demonstrated that protein but not lipid phosphatase activity of PTEN was involved in the regulation of MTF-1 activity. The same regulatory role of PTEN was also noted in the regulation of ZnT1 (zinc transporter 1), another target gene of MTF-1.


Assuntos
Proteínas de Ligação a DNA/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia , Animais , Células CHO , Cádmio , Proteínas de Transporte de Cátions , Cricetinae , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Homeostase , Humanos , Metalotioneína/genética , Metalotioneína/metabolismo , Camundongos , PTEN Fosfo-Hidrolase/genética , Fatores de Transcrição/genética , Zinco/metabolismo , Fator MTF-1 de Transcrição
2.
Cell Death Discov ; 9(1): 340, 2023 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-37696858

RESUMO

The tumor suppressor p53 primarily functions as a mediator of DNA damage-induced cell death, thereby contributing to the efficacy of genotoxic anticancer therapeutics. Here, we show, on the contrary, that cancer cells can employ genotoxic stress-induced p53 to acquire treatment resistance through the production of the pleiotropic cytokine interleukin (IL)-6. Mechanistically, DNA damage, either repairable or irreparable, activates p53 and stimulates Caspase-2-mediated cleavage of its negative regulator mouse double minute 2 (MDM2) creating a positive feedback loop that leads to elevated p53 protein accumulation. p53 transcriptionally controls the major adenosine triphosphate (ATP) release channel pannexin 1 (Panx1), which directs IL-6 induction via a mechanism dependent on the extracellular ATP-activated purinergic P2 receptors as well as their downstream intracellular calcium (iCa2+)/PI3K/Akt/NF-ĸB signaling pathway. Thus, p53 silencing impairs Panx1 and IL-6 expression and renders cancer cells sensitive to genotoxic stress. Moreover, we confirm that IL-6 hampers the effectiveness of genotoxic anticancer agents by mitigating DNA damage, driving the expression of anti-apoptotic Bcl-2 family genes, and maintaining the migratory and invasive properties of cancer cells. Analysis of patient survival and relevant factors in lung cancer and pan-cancer cohorts supports the prognostic and clinical values of Panx1 and IL-6. Notably, IL-6 secreted by cancer cells during genotoxic treatments promotes the polarization of monocytic THP-1-derived macrophages into an alternative (M2-like) phenotype that exhibits impaired anti-survival activities but enhanced pro-metastatic effects on cancer cells as compared to nonpolarized macrophages. Our study reveals the precise mechanism for genotoxic-induced IL-6 and suggests that targeting p53-mediated IL-6 may improve the responsiveness of cancer cells to genotoxic anticancer therapy.

3.
Redox Biol ; 64: 102791, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37385076

RESUMO

Snake venom l-amino acid oxidases (svLAAOs) have been recognized as promising candidates for anticancer therapeutics. However, multiple aspects of their catalytic mechanism and the overall responses of cancer cells to these redox enzymes remain ambiguous. Here, we present an analysis of the phylogenetic relationships and active site-related residues among svLAAOs and reveal that the previously proposed critical catalytic residue His 223 is highly conserved in the viperid but not the elapid svLAAO clade. To gain further insight into the action mechanism of the elapid svLAAOs, we purify and characterize the structural, biochemical, and anticancer therapeutic potentials of the Thailand elapid snake Naja kaouthia LAAO (NK-LAAO). We find that NK-LAAO, with Ser 223, exhibits high catalytic activity toward hydrophobic l-amino acid substrates. Moreover, NK-LAAO induces substantial oxidative stress-mediated cytotoxicity with the magnitude relying on both the levels of extracellular hydrogen peroxide (H2O2) and intracellular reactive oxygen species (ROS) generated during the enzymatic redox reactions, but not being influenced by the N-linked glycans on its surface. Unexpectedly, we discover a tolerant mechanism deployed by cancer cells to dampen the anticancer activities of NK-LAAO. NK-LAAO treatment amplifies interleukin (IL)-6 expression via the pannexin 1 (Panx1)-directed intracellular calcium (iCa2+) signaling pathway to confer adaptive and aggressive phenotypes on cancer cells. Accordingly, IL-6 silencing renders cancer cells vulnerable to NK-LAAO-induced oxidative stress together with abrogating NK-LAAO-stimulated metastatic acquisition. Collectively, our study urges caution when using svLAAOs in cancer treatment and identifies the Panx1/iCa2+/IL-6 axis as a therapeutic target for improving the effectiveness of svLAAOs-based anticancer therapies.


Assuntos
Interleucina-6 , Neoplasias , Humanos , Interleucina-6/genética , L-Aminoácido Oxidase/química , L-Aminoácido Oxidase/metabolismo , L-Aminoácido Oxidase/farmacologia , Peróxido de Hidrogênio/metabolismo , Filogenia , Venenos de Serpentes , Neoplasias/tratamento farmacológico , Aminoácidos
4.
Front Bioeng Biotechnol ; 11: 1236429, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38094898

RESUMO

Micron-scale structure biphasic calcium phosphate (BCP) materials have demonstrated promising clinical outcomes in the field of bone tissue repair. However, research on biphasic calcium phosphate materials at the nanoscale level remains limited. In this study, we synthesize granular-shaped biphasic calcium phosphate nanomaterials with multiple desirable characteristics, including negatively charged surfaces, non-cytotoxicity, and the capability to penetrate cells, using a nanogrinding dispersion process with a polymeric carboxylic acid as the dispersant. Our results reveal that treating human osteoblasts with 0.5 µg/mL biphasic calcium phosphate nanomaterials results in a marked increase in alkaline phosphatase (ALP) activity and the upregulation of osteogenesis-related genes. Furthermore, these biphasic calcium phosphate nanomaterials exhibit immunomodulatory properties. Treatment of THP-1-derived macrophages with BCP nanomaterials decreases the expression of various inflammatory genes. Biphasic calcium phosphate nanomaterials also mitigate the elevated inflammatory gene expression and protein production triggered by lipopolysaccharide (LPS) exposure in THP-1-derived macrophages. Notably, we observe that biphasic calcium phosphate nanomaterials have the capacity to reverse the detrimental effects of LPS-stimulated macrophage-conditioned medium on osteoblastic activity and mineralization. These findings underscore the potential utility of biphasic calcium phosphate nanomaterials in clinical settings for the repair and regeneration of bone tissue. In conclusion, this study highlights the material properties and positive effects of biphasic calcium phosphate nanomaterials on osteogenesis and immune regulation, opening a promising avenue for further research on inflammatory osteolysis in patients undergoing clinical surgery.

5.
J Biol Chem ; 286(50): 42818-29, 2011 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-22021037

RESUMO

Metal-responsive transcription factor 1 (MTF-1) is an essential protein required for mouse embryonic development. We report here the occurrence of sumoylation on MTF-1. Mutational studies demonstrated that sumoylation occurs on the lysine residue at position 627 (Lys(627)) of mouse MTF-1. Small ubiquitin-like modifier (SUMO)-1 was fused to the C terminus of MTF-1 to mimic the sumoylated form of the protein and it suppressed the transcriptional activity of MTF-1. The nuclear translocation activity, DNA-binding activity, and protein stability of SUMO-fused MTF-1 are similar to that of wild type MTF-1. The level of sumoylation was reduced by metal in a dose- and time-dependent manner. The fact that zinc reduces MTF-1 sumoylation makes the suppressive role of sumoylated MTF-1 in transcription physiologically less significant because the SUMO moiety of MTF-1 is removed when MTF-1 translocates into nucleus. We further identified a SUMO-interacting motif (SIM) on MTF-1. Remarkably, MTF-1 binds sumoylated MTF-1 and/or other cellular factors in a SIM-dependent manner. This interaction was disrupted by treating cells with zinc. Gel permeation chromatography demonstrated that MTF-1 forms SIM-dependent complexes. This cross-interaction transpires in the cytoplasm and markedly reduces upon nuclear translocation. It can therefore be concluded that SUMO conjugation and the SIM on MTF-1 do not play a critical role in suppressing transcriptional activity. Instead, MTF-1 forms complexes with cellular factors through SIM and SUMO moiety in the cytoplasm. The result explores a new understanding for the mode of MTF-1 assembly and regulation in cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína SUMO-1/metabolismo , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Animais , Células CHO , Cádmio/farmacologia , Linhagem Celular , Imunoprecipitação da Cromatina , Cromatografia de Afinidade , Cricetinae , Proteínas de Ligação a DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Imunoprecipitação , Ligação Proteica/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Proteína SUMO-1/genética , Relação Estrutura-Atividade , Sumoilação/efeitos dos fármacos , Fatores de Transcrição/genética , Zinco/farmacologia , Fator MTF-1 de Transcrição
6.
Oncogenesis ; 11(1): 26, 2022 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-35589683

RESUMO

Tumor suppressor p53 plays a central role in preventing tumorigenesis. Here, we unravel how p53 modulates mitochondrial dynamics to restrain the metastatic properties of cancer cells. p53 inhibits the mammalian target of rapamycin complex 1 (mTORC1) signaling to attenuate the protein level of mitochondrial fission process 1 (MTFP1), which fosters the pro-fission dynamin-related protein 1 (Drp1) phosphorylation. This regulatory mechanism allows p53 to restrict cell migration and invasion governed by Drp1-mediated mitochondrial fission. Downregulating p53 expression or elevating the molecular signature of mitochondrial fission correlates with aggressive tumor phenotypes and poor prognosis in cancer patients. Upon p53 loss, exaggerated mitochondrial fragmentation stimulates the activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling resulting in epithelial-to-mesenchymal transition (EMT)-like changes in cell morphology, accompanied by accelerated matrix metalloproteinase 9 (MMP9) expression and invasive cell migration. Notably, blocking the activation of mTORC1/MTFP1/Drp1/ERK1/2 axis completely abolishes the p53 deficiency-driven cellular morphological switch, MMP9 expression, and cancer cell dissemination. Our findings unveil a hitherto unrecognized mitochondria-dependent molecular mechanism underlying the metastatic phenotypes of p53-compromised cancers.

7.
Toxicol Appl Pharmacol ; 235(2): 153-62, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19135076

RESUMO

Cadmium (Cd) induces necrotic death in Chinese hamster ovary (CHO) K1 cells and we have established the responsible signaling pathway. Reportedly, necrostatin-1 (Nec-1) rescues cells from necrotic death by mediating through the death domain receptor (DR) signaling pathway. We show here that Nec-1 also effectively attenuates necrotic death triggered by Cd. Two other treatments that cause necrotic cell death, one can (z-VAD-fmk/TNF-alpha on U937 cells) and the other cannot (etherynic acid (EA) on DLD-1 cells) be rescued by Nec-1, were also studied in parallel for comparison. Results show that Nec-1 is ineffectual in modulating intracellular calcium contents, calpain activity (a downstream protease), or reactive oxygen species production. It can counteract the reduction in mitochondrial membrane potential (MMP) caused by treating CHO K1 or U937 cells with necrosis-inducing agent. However, this effect was not found in EA-treated DLD-1 cells. Notably, Nec-1 elevates NF-kappaB activity in the presence or absence of necrosis-inducing agents. Our study shows that, in addition to DR-mediated necrosis, Nec-1 is effective in attenuating Cd-induced necrosis. It rescues cells with reduced MMP implying that mitochondrion is its major acting site.


Assuntos
Apoptose/efeitos dos fármacos , Intoxicação por Cádmio/prevenção & controle , Morte Celular/efeitos dos fármacos , Imidazóis/farmacologia , Indóis/farmacologia , Animais , Células CHO , Intoxicação por Cádmio/patologia , Sinalização do Cálcio/efeitos dos fármacos , Calpaína/metabolismo , Linhagem Celular , Quelantes/farmacologia , Cricetinae , Cricetulus , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , NF-kappa B/genética , NF-kappa B/fisiologia , Necrose , Propídio , Espécies Reativas de Oxigênio/metabolismo , Receptores de Droga/efeitos dos fármacos , Transfecção , Células U937
8.
Int J Radiat Biol ; 85(3): 214-26, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19296338

RESUMO

PURPOSE: The aim of this work is to compare the radiosensitizing effect between organic and inorganic germanium compounds and to investigate whether nanometer-sized germanium particles can act as radiosensitizers. MATERIALS AND METHODS: Bis (2-carboxyethylgermanium) sesquioxide (Ge-132), germanium oxide (GeO(2)) and germanium nanoparticles were used in this study. Cell viability was determined by clonogenic survival assay. Cellular DNA damage was evaluated by alkaline comet assay, confocal microscopy and the cellular level of phospho-histone H2AX (gamma-H2AX). RESULTS: Nanometer-sized germanium particles were fabricated. They have a similar radiosensitizing effect as that of GeO(2). Conversely, Ge-132 did not enhance the radiosensitivity of cells. Comet assay was employed to evaluate the level of DNA damage and confirmed that inorganic germanium compounds enhanced cellular radiosensitivity. Notably, the comet assay indicated that the nanoparticle itself caused a higher level of DNA damage. The possibility that germanium nanoparticles per se caused DNA damage was ruled out when the cellular level of gamma-H2AX was examined. CONCLUSIONS: We demonstrated that inorganic but not organic germanium compounds exerted radiosensitizing effect in cells. Nanometer-sized germanium particles were fabricated and were able to enhance the radiosensitivity of cells. Confounding effect may occur when comet assay is used to estimate the level of DNA damage in the presence of germanium nanoparticles.


Assuntos
Germânio/química , Nanopartículas , Compostos Organometálicos/química , Radiossensibilizantes/química , Animais , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cricetinae , Cricetulus , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Germânio/farmacologia , Histonas/metabolismo , Compostos Organometálicos/farmacologia , Fosforilação , Propionatos , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia
9.
Biochem Pharmacol ; 166: 203-211, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31129049

RESUMO

Leptin is an adipokine predominantly secreted by adipocytes and has many physiological roles, including in energy homeostasis. We identified that AM630, a cannabinoid receptor 2 (CB2) antagonist, down-regulated leptin expression in mature adipocytes differentiated from either stromal vascular fractions isolated from inguinal fat pads of C57BL/6J mice or 3T3-L1 preadipocytes. However, the leptin-suppressive effects of AM630 preserved in CB2-deficient adipocytes indicated the off-target activity of AM630 in leptin expression. Pharmacological and genetic studies, cheminformatics, and docking simulation were applied to identify the potential protein target of AM630 that modulates leptin expression in differentiated primary preadipocytes. Screening of the reported off-targets of AM630 identified a synthetic cannabinoid WIN55212-2 exerting the same function. Target deconvolution and docking simulation suggested that AM630 and WIN55212-2 were both inhibitors of lipocalin-type prostaglandin D2 synthase (L-PGDS). Further studies showed that L-PGDS positively regulates leptin expression. Although glucocorticoid and aldosterone were previously reported to induce expression of both L-PGDS and leptin, our data demonstrated that L-PGDS mediates only glucocorticoid-induced leptin expression in differentiated primary preadipocytes. No effect was observed after aldosterone treatment. This newly discovered glucocorticoid - L-PGDS - leptin pathway may provide insights into current clinical use of glucocorticoid and management of their undesired effects such as obesity.


Assuntos
Adipócitos/metabolismo , Adipogenia/fisiologia , Glucocorticoides/farmacologia , Oxirredutases Intramoleculares/metabolismo , Leptina/biossíntese , Lipocalinas/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Expressão Gênica , Indóis/metabolismo , Indóis/farmacologia , Leptina/agonistas , Leptina/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Secundária de Proteína , Relação Estrutura-Atividade
10.
Toxicol Res (Camb) ; 8(1): 101-111, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30713663

RESUMO

Arsenic and its compounds are toxic environmental pollutants and known carcinogens. We investigated here the mechanism of arsenite-induced damage in renal cells. Treating human embryonic kidney cells (HEK293) with sodium arsenite reduces cell viability in a dose- and time-dependent manner. The decline of cell viability is due to apoptotic death since arsenite treatment reduces Akt activity and the Bcl2 level but increases caspase 3 activity and the cytochrome c level. These effects can be reverted by the addition of an apoptosis inhibitor. PTEN, the upstream negative regulator of Akt activity, was also reduced with arsenite treatment. Noticeably, PTEN markedly increased in the insoluble fraction of the cells, suggesting a cell failure in removing the damaged proteins. Arsenite treatment activates a variety of signaling factors. Among them, ERK and JNK are associated with autophagy via regulating the levels of LC3 and p62. With arsenite administration, the LC3 and p62 levels increased. However, lysosomal activity was decreased and led to the decline of autophagic activity. The addition of rapamycin, the mTOR inhibitor, activated the autophagic pathway that accelerated the removal of damaged proteins. The recovery of autophagy increased the viability of arsenite-treated cells. Similar to rapamycin treatment, the knockdown of mTOR expression also enhanced the viability of arsenite-treated cells. Both rapamycin treatment and mTOR knockdown enhanced ERK activity further, but reduced JNK activity and the p62 level in arsenite-treated cells. Lysosomal activity increased with the depletion of mTOR, indicating an increase of autophagic activity. These results reveal the critical role of mTOR in regulating the cell fate of arsenite-exposed renal cells.

11.
PLoS One ; 13(1): e0191971, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29381770

RESUMO

Zinc finger (ZF) motifs on proteins are frequently recognized as a structure for DNA binding. Accumulated reports indicate that ZF motifs contain nuclear localization signal (NLS) to facilitate the transport of ZF proteins into nucleus. We investigated the critical factors that facilitate the nuclear transport of triple C2H2 ZF proteins. Three conserved basic residues (hot spots) were identified among the ZF sequences of triple C2H2 ZF proteins that reportedly have NLS function. Additional basic residues can be found on the α-helix of the ZFs. Using the ZF domain (ZFD) of Egr-1 as a template, various mutants were constructed and expressed in cells. The nuclear transport activity of various mutants was estimated by analyzing the proportion of protein localized in the nucleus. Mutation at any hot spot of the Egr-1 ZFs reduced the nuclear transport activity. Changes of the basic residues at the α-helical region of the second ZF (ZF2) of the Egr-1 ZFD abolished the NLS activity. However, this activity can be restored by substituting the acidic residues at the homologous positions of ZF1 or ZF3 with basic residues. The restored activity dropped again when the hot spots at ZF1 or the basic residues in the α-helix of ZF3 were mutated. The variations in nuclear transport activity are linked directly to the binding activity of the ZF proteins with importins. This study was extended to other triple C2H2 ZF proteins. SP1 and KLF families, similar to Egr-1, have charged amino acid residues at the second (α2) and the third (α3) positions of the α-helix. Replacing the amino acids at α2 and α3 with acidic residues reduced the NLS activity of the SP1 and KLF6 ZFD. The reduced activity can be restored by substituting the α3 with histidine at any SP1 and KLF6 ZFD. The results show again the interchangeable role of ZFs and charge residues in the α-helix in regulating the NLS activity of triple C2H2 ZF proteins.


Assuntos
Aminoácidos/metabolismo , Núcleo Celular/metabolismo , Dedos de Zinco , Sequência de Aminoácidos , Transporte Proteico
12.
PLoS One ; 11(1): e0147011, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26751215

RESUMO

Cadmium is a known environmental carcinogen. Exposure of Cd leads to the activation of several proto-oncogenes in cells. We investigated here the mechanism of c-Myc expression in hepatic cells under Cd treatment. The c-Myc protein and mRNA levels increased in dose- and time-dependent manners in HepG2 cells with Cd treatment. This increase was due to an increase in c-Myc mRNA stability. To explore the mechanism involved in enhancing the mRNA stability, several cellular signaling factors that evoked by Cd treatment were analyzed. PI3K, p38, ERK and JNK were activated by Cd. However, ERK did not participate in the Cd-induced c-Myc expression. Further analysis revealed that mTORC2 was a downstream factor of p38. PI3K, JNK and mTORC2 coordinately activated Akt. Akt was phosphorylated at Thr450 in the untreated cells. Cd treatment led to additional phosphorylation at Thr308 and Ser473. Blocking any of the three signaling factors resulted in the reduction of phosphorylation level at all three Akt sites. The activated Akt phosphorylated Foxo1 and allowed the modified protein to translocate into the cytoplasm. We conclude that Cd-induced accumulation of c-Myc requires the activation of several signaling pathways. The signals act coordinately for Akt activation and drive the Foxo1 from the nucleus to the cytoplasm. Reduction of Foxo1 in the nucleus reduces the transcription of its target genes that may affect c-Myc mRNA stability, resulting in a higher accumulation of the c-Myc proteins.


Assuntos
Cádmio/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Estabilidade de RNA , Transdução de Sinais/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Citosol/metabolismo , Relação Dose-Resposta a Droga , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Fígado/metabolismo , Fosforilação/genética , Plasmídeos/metabolismo , RNA Mensageiro/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina
13.
Toxicol Sci ; 85(1): 551-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15689422

RESUMO

Cadmium (Cd) stimulates the production of reactive oxygen species (ROS) and causes cell damage. We investigated here the feasibility of using a cell permeable superoxide dismutase/catalase mimetic, EUK-8, to reduce the Cd-induced ROS and cytotoxicity in Chinese hamster ovary cells. EUK-8 reduces the ROS level caused by Cd treatment. EUK-8 also curtails propidium iodide (PI) influx and increases the viability of Cd-treated cells. The efficacy of EUK-8 as a Cd antidote diminishes gradually when added at a later stage of Cd treatment. EUK-8 blocks Cd transport into cells. It is ineffective in accelerating the efflux of metals from the cells. EUK-8 is a Mn-salen complex. Mn decreases the uptake and cytotoxicity of Cd, while salen perturbs the membrane integrity and increases the uptake and cytotoxicity of Cd. Salen is able to bind Cd, and the Cd-salen complex formed does not perturb the integrity of cell membranes and thus the influx of metal is not enhanced. Our results reveal a differential effect of salen and Mn-salen complex on the transport of Cd with subsequent different levels of cell damage.


Assuntos
Cloreto de Cádmio/toxicidade , Quelantes/farmacologia , Etilenodiaminas/farmacologia , Compostos Organometálicos/farmacologia , Animais , Células CHO , Cloreto de Cádmio/farmacocinética , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Espécies Reativas de Oxigênio/metabolismo
14.
J Nutr Biochem ; 26(4): 351-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25618524

RESUMO

Zinc transporter 2 (ZnT2) is one of the cellular factors responsible for Zn homeostasis. Upon Zn overload, ZnT2 reduces cellular Zn by transporting it into excretory vesicles. We investigated the molecular mechanism that regulates human ZnT2 (hZnT2) gene expression. Zn induces hZnT2 expression in dose- and time-dependent manners. Overexpression of metal-responsive transcription factor 1 (MTF-1) increases hZnT2 transcription, whereas depletion of MTF-1 reduces hZnT2 expression. There are five putative metal response elements (MREs) within 1kb upstream of the hZnT2 gene. A serial deletion of the hZnT2 promoter region (from 5' to 3') shows that the two MREs proximal to the gene are essential for Zn-induced promoter activity. Further mutation analysis concludes that the penultimate MRE (MREb) supports the metal-induced promoter activity. The hZnT2 promoter has also a zinc finger E-box binding homeobox (ZEB) binding element. Mutation or deletion of this ZEB binding element elevates the basal and Zn-induced hZnT2 promoter activities. Knockdown of ZEB1 mRNA enhances the hZnT2 transcript level in HEK-293 cells. In MCF-7 (ZEB-deficient) cells, expression of ZEB proteins attenuates the Zn-induced hZnT2 expression. However, expressions of MTF-1 target genes such as human ZnT1 and metallothionein IIA were not affected. Our study shows the expression of the hZnT2 gene is coordinately regulated via active and suppressive modulators.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição/metabolismo , Animais , Células CHO , Cádmio/metabolismo , Proteínas de Transporte de Cátions/genética , Cricetulus , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Genes Reporter , Células HEK293 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células MCF-7 , Metalotioneína/genética , Metalotioneína/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Elementos de Resposta , Análise de Sequência de DNA , Fatores de Transcrição/genética , Transcrição Gênica , Transfecção , Zinco/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco , Fator MTF-1 de Transcrição
15.
Radiat Res ; 159(3): 391-400, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12600242

RESUMO

We investigated here the combined effect of GeO(2) and radiation on cell viability. Cells were treated with 0 to 22 mM GeO(2) for 12 h followed by 1 Gy X irradiation. A synergistic cytotoxic effect was observed for the combined treatment with a dose-dependent reduction of cell viability. Complete survival curves showed a 2.3- and 2.75-fold increase in radiosensitivity for 50% cell death in the presence of 5 and 15 mM GeO(2), respectively. The increased radiosensitivity also occurred when GeO(2) was given either 4 h prior to irradiation or immediately after radiation exposure. GeO(2) did not affect the total soluble thiol content or the activities of catalase and glutathione S-transferase. Analysis of the production of reactive oxygen species (ROS) revealed that the combined treatment dramatically increased the synthesis of ROS. Addition of N-acetyl cysteine (NAC, 20 mM) decreased the production of ROS in cells. NAC, however, increased cell viability only slightly after treatment with GeO(2) and radiation. Thus increased production of ROS makes little or no contribution to the observed death. The combination of GeO(2) and X radiation, however, significantly increased the frequency of DNA double-strand breaks (DSBs). Notably, the presence of GeO(2) also reduced the efficiency of DNA repair. We conclude that treatment with GeO(2) followed by X irradiation increases DNA DSBs and cell death.


Assuntos
Sobrevivência Celular/efeitos da radiação , Dano ao DNA/efeitos da radiação , Germânio/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Antimutagênicos/farmacologia , Células CHO , Catalase/metabolismo , Cricetinae , Reparo do DNA , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Sequestradores de Radicais Livres/farmacologia , Glutationa Transferase/metabolismo , Espécies Reativas de Oxigênio , Compostos de Sulfidrila/metabolismo , Compostos de Sulfidrila/farmacologia , Fatores de Tempo , Raios X
16.
Biosens Bioelectron ; 20(4): 864-71, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15522603

RESUMO

A biosensor based on mammalian metallothionein (MT) for the detection of metal ions was developed and characterized. MT was immobilized onto a carboxymethylated dextran matrix as a biosensor for the detection of metal ions by surface plasmon resonance (SPR). The optimal pH for the immobilization step was determined to be 4. The temperature for the analysis was also defined, and the highest interaction was observed at 30 degrees C. The MT sensor chip binds cadmium (Cd), zinc (Zn) or nickel (Ni), but not magnesium (Mg), manganese (Mn) and calcium (Ca). Calibration curves for the quantification of metal ions showed excellent linearity. The sensitivity for metal detection is at the micromolar level. The interaction between the metal ions and the sensor chip is influenced significantly by the presence of NaCl, Tween 20 and the pH of the reaction buffer. By decreasing the NaCl in the reaction buffer to 1 mM, the MT chip effectively differentiates cadmium from zinc and nickel. Kinetic parameters of the metal-MT interactions were also determined by using this chip. The binding affinity between the metal ions and the immobilized MT follows the order of cadmium > zinc > nickel, which is the same as that determined for MT in solution. Thus, the MT chip can be an effective biosensor for the detection and measurement of several metal ions.


Assuntos
Técnicas Biossensoriais/instrumentação , Metalotioneína/química , Metais Pesados/análise , Metais Pesados/química , Ressonância de Plasmônio de Superfície/instrumentação , Adsorção , Técnicas Biossensoriais/métodos , Materiais Revestidos Biocompatíveis/química , Ligação Proteica , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Ressonância de Plasmônio de Superfície/métodos
17.
Chem Biol Interact ; 149(2-3): 125-36, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15501434

RESUMO

Chinese hamster ovary K1 (CHO K1) cells are very sensitive to cadmium (Cd) toxicity. They were used to investigate the effect of Cd on cell cycle progression. Cells were cultured with 0.1, 0.4, 1 or 4 microM Cd for various time intervals. There was no difference in growth rate when less than 0.4 microM Cd was given within 24 h. A dose-dependent reduction of cell proliferation was observed when more than 0.4 microM of Cd was given. The cells were pulse-labeled with 5-bromodeoxyuridine (BrdU), and the labeled cells were cultured in the presence of increasing concentrations of Cd. Cell cycle progression was retarded as a function of Cd concentration. G2/M arrest was observed when the BrdU-labeled cells were treated with 1 microM Cd for 8h, whereas cells receiving 4 microM Cd stopped at the S phase within 4 h. Cell cycle analysis of cells treated with Cd for 24 h showed that G2/M arrest occurred only when cells received 0.8 to 2 microM Cd. Despite the occurrence of G2/M arrest in the Cd treatment, only a limited proportion of the cells were blocked in the M phase. However, the increase in M phase cells coincided with an elevation in the cyclin-dependent kinase 1 activity. To examine whether Cd acts on cells at a specific cell stage, they were synchronized at the G1 or G2/M phase then treated with 1 microM Cd for 12 h. The cells were blocked at the G2/M and G1/S phase, respectively. This finding indicates that Cd toxicity is global and not cell phase specific. We also investigated the involvement of Cd-induced reactive oxygen species (ROS) with the occurrence of G2/M block and found a lack of correlation between cell cycle arrest and ROS production. We measured the Cd content that caused G2/M arrest from a series of Cd treatments and determined the ranges of cumulative Cd concentrations that could result in cell cycle arrest.


Assuntos
Células CHO/efeitos dos fármacos , Cádmio/toxicidade , Ciclo Celular/efeitos dos fármacos , Animais , Western Blotting , Bromodesoxiuridina/metabolismo , Proteína Quinase CDC2/metabolismo , Células CHO/citologia , Células CHO/metabolismo , Proliferação de Células/efeitos dos fármacos , Cricetinae , Citometria de Fluxo , Espécies Reativas de Oxigênio/metabolismo
18.
Chem Biol Interact ; 141(3): 211-28, 2002 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-12385720

RESUMO

We report here for the first time that germanium oxide (GeO(2)) blocks cell progression. GeO(2) is not genotoxic to Chinese hamster ovary (CHO) cells and has limited cytotoxicity. However, GeO(2) arrests cells at G2/M phase. The proportion of cells stopped at G2/M phase increased dose-dependently up to 5 mM GeO(2) when treated for 12 h, but decreased at GeO(2) concentration was greater than 5 mM. Analysis of 5-bromodeoxyuridine-labeled cells indicated that GeO(2) delayed S phase progression in a dose-dependent manner, and blocked cells at G2/M phase. Microscopic examination confirmed that GeO(2) treatment arrested cells at G2 phase. Similar to several other events that cause G2 block, the GeO(2)-induced G2 block can also be ameliorated by caffeine in a dose- and time-dependent manner. To explore the mechanism of G2 arrest by GeO(2), cyclin content and cyclin-dependent kinase activity were examined. Cyclin B1 level was not affected after GeO(2) treatment in CHO cells. However, GeO(2) decreased p34(cdc2) kinase (Cdk1) activity. The kinase activity recovered within 9 h after GeO(2) removal and correlated with the transition of G2/M-G1 phase of the cells. This result suggests that GeO(2) treatment reduces Cdk1 activity and causing the G2 arrest in CHO cells.


Assuntos
Fase G2/efeitos dos fármacos , Germânio/toxicidade , Mitose/efeitos dos fármacos , Animais , Proteína Quinase CDC2/metabolismo , Células CHO , Ciclo Celular/efeitos dos fármacos , Colchicina/farmacologia , Cricetinae , Ciclinas/metabolismo , Citotoxinas/farmacologia , Citotoxinas/toxicidade , Relação Dose-Resposta a Droga , Interações Medicamentosas , Germânio/farmacologia , Immunoblotting , Testes para Micronúcleos , Fatores de Tempo
19.
J Biomed Opt ; 19(1): 011004, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23788282

RESUMO

The actin-depolymerizing factor (ADF)/cofilin protein family has been reported to be associated with ischemia-induced renal disorders. We examine whether cofilin-1 is associated with acute kidney injury (AKI) using human urine samples. We exploited a 96-well based high-throughput biosensor that uses gold nanoparticles and a sandwich immunoassay to detect the urine cofilin-1 level of AKI patients. The mean urine cofilin-1 level of the AKI patients (n=37 from 47 cases analyzed) was twofold higher than that of healthy adults (n=21 from 29 cases analyzed). The receiver operating characteristic (ROC) curve showed that cofilin-1 was acceptable for discriminating AKI patients from healthy adults. However, an increase of the sample size is required to conclude the importance of urine cofilin-1 on AKI diagnosis, and the high-throughput ultrasensitive biosensor used in this study would greatly accelerate the measurement of urine cofilin-1 in an increased sample size.


Assuntos
Injúria Renal Aguda/urina , Cofilina 1/urina , Ensaios de Triagem em Larga Escala/métodos , Espectrometria de Fluorescência/métodos , Ressonância de Plasmônio de Superfície/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Técnicas Biossensoriais/métodos , Estudos de Casos e Controles , Linhagem Celular , Feminino , Ouro/química , Humanos , Masculino , Nanopartículas Metálicas/química , Pessoa de Meia-Idade , Estresse Oxidativo , Curva ROC
20.
J Mater Chem B ; 1(20): 2639-2646, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32260951

RESUMO

Development of carbon nanotubes toward commercial antibacterial applications warrants the understanding of their interaction mechanism with bacterial cells. The antibacterial activity and mechanism of acid-functionalized single-walled carbon nanotube (AFSWCNT) coated paper was assessed for gram-positive Staphylococcus aureus and gram-negative Escherichia coli models of bacteria. Better activity towards gram-positive bacteria was observed, whereas the presence of an outer membrane makes gram-negative bacteria more resistant to cell membrane damage caused by AFSWCNTs. Based on measured cytoplasmic efflux materials of bacteria, X-ray photoelectron spectroscopy, and scanning transmission electron microscopy combined with electron energy-loss spectroscopy imaging studies, we found that the better antibacterial activity of AFSWCNTs toward gram-positive bacteria is attributed to not only direct physical contact and piercing action, but also molecular-scale interaction with surface functional groups of bacteria. The novel antibacterial mechanism of AFSWCNTs might bring a promising strategy to design new antibacterial materials against drug-resistant bacteria species.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA