Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-37873235

RESUMO

Telomeres protect chromosome ends and determine the replication potential of dividing cells. The canonical telomere sequence TTAGGG is synthesized by telomerase holoenzyme, which maintains telomere length in proliferative stem cells. Although the core components of telomerase are well-defined, mechanisms of telomerase regulation are still under investigation. We report a novel role for the Src family kinase Fyn, which disrupts telomere maintenance in stem cells by phosphorylating the scaffold protein Menin. We found that Fyn knockdown prevented telomere erosion in human and mouse stem cells, validating the results with four telomere measurement techniques. We show that Fyn phosphorylates Menin at tyrosine 603 (Y603), which increases Menin's SUMO1 modification, C-terminal stability, and importantly, its association with the telomerase RNA component (TR). Using mass spectrometry, immunoprecipitation, and immunofluorescence experiments we found that SUMO1-Menin decreases TR's association with telomerase subunit Dyskerin, suggesting that Fyn's phosphorylation of Menin induces telomerase subunit mislocalization and may compromise telomerase function at telomeres. Importantly, we find that Fyn inhibition reduces accelerated telomere shortening in human iPSCs harboring mutations for dyskeratosis congenita.

2.
Inflamm Res ; 62(7): 681-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23603994

RESUMO

OBJECTIVE: Natural polyamines are some of the most abundant polycationic molecules in eukaryotic cells, regulating gene expression. Polyamines have been reported to possess anti-inflammatory activities in many model inflammation systems. However, there is no report on their role in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced dermal edema. METHODS: Mouse ear edema was induced by TPA. Edema biopsies were investigated using H&E staining. Levels of nitric oxide (NO) were determined using the Griess reaction. Tumor necrosis factor α (TNFα) and interleukin (IL)-1ß levels in cell supernatants were measured by TNFα and IL-1ß ELISA kits. RESULTS: Spermidine and spermine caused significant decreases in ear thickness, water content, and neutrophil infiltrations in comparison with negative control (p < 0.05). External polyamines reduced the levels of inflammatory mediators such as NO, TNFα, and IL-1ß in lipopolysaccharide (LPS)-stimulated RAW264.7 murine macrophages. Spermine had a higher inhibitory effect on the production of cytokines such as IL-1ß and TNFα in LPS-stimulated murine macrophages compared to other polyamines. CONCLUSION: Our findings clearly demonstrated that polyamines are involved in the anti-inflammatory effect by reducing dermal edema thickness and other inflammatory mediators like NO and cytokines in a dose-dependent manner.


Assuntos
Anti-Inflamatórios/uso terapêutico , Edema/tratamento farmacológico , Ativação de Macrófagos/efeitos dos fármacos , Espermidina/uso terapêutico , Espermina/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Linhagem Celular , Edema/induzido quimicamente , Edema/imunologia , Interleucina-1beta/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/imunologia , Pele/efeitos dos fármacos , Pele/imunologia , Pele/patologia , Espermidina/farmacologia , Espermina/farmacologia , Acetato de Tetradecanoilforbol , Fator de Necrose Tumoral alfa/imunologia
3.
Int J Oncol ; 61(5)2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36129143

RESUMO

The aim of the present study was to decipher the mechanism of glutathione­S­transferase Ω­1 (GSTO1)­induced drug resistance in colon cancer cells. Cisplatin is used widely as a therapeutic drug in cancer, but colon cancer is the most susceptible to acquired drug resistance. Autophagy is recognized as one of the contributors to drug resistance in cancers. Phase II detoxifying enzymes, such as GSTO1, serve important roles in autophagy­apoptosis cross talk. The present study revealed a novel interaction between GSTO1 and TNFα­induced protein 3/zinc­finger protein A20 (TNFαIP3/A20) as a prime target for cisplatin sensitization in drug­resistant cells. GSTO1 and ATP­binding cassette subfamily B member 1 (ABCB1) were both expressed at higher levels in multidrug­resistant (MDR) HCT­116 cells compared with the wild­type (WT) HCT­116 cells, suggesting they may serve vital roles in multidrug resistance. MDR cells showed autophagy induction, which is dependent on calcium signaling­dependent endoplasmic stress. In WT cells, the mitochondria­dependent pathway leads to apoptosis, which was not observed in MDR cells. The MDR conditions were mimicked by transfecting WT cells with the GSTO1­activation CRISPR plasmid, which induced autophagy. Similarly, MDR cells with GSTO1­knockdown (KD) CRISPR/Cas9 transfection showed reduced autophagy with increased apoptosis. These data revealed a potentially important role of GSTO1 in drug resistance. A GSTO1 pull­down assay detected TNFαIP3/A20 as a binding partner in MDR cells. The data suggested that the expression of TNFαIP3/A20 may be dependent on GSTO1 expression in MDR cells. Targeting either GSTO1 or TNFαIP3/A20 by CRISPR/Cas9 sensitized the MDR cells to cisplatin. GSTO1 and TNFαIP3/A20 dual­KD cells were more sensitive to cisplatin compared with single­gene KD cells. These data highlight the importance of the GSTO1­TNFαIP3/A20 interaction during drug resistance.


Assuntos
Cisplatino , Neoplasias do Colo , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Glutationa Transferase , Trifosfato de Adenosina , Cisplatino/farmacologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Glutationa , Glutationa Transferase/genética , Células HCT116 , Humanos , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fator de Necrose Tumoral alfa/farmacologia , Zinco
4.
Nat Commun ; 13(1): 7074, 2022 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-36400785

RESUMO

Centromere defects in Systemic Sclerosis (SSc) have remained unexplored despite the fact that many centromere proteins were discovered in patients with SSc. Here we report that lesion skin fibroblasts from SSc patients show marked alterations in centromeric DNA. SSc fibroblasts also show DNA damage, abnormal chromosome segregation, aneuploidy (only in diffuse cutaneous (dcSSc)) and micronuclei (in all types of SSc), some of which lose centromere identity while retaining centromere DNA sequences. Strikingly, we find cytoplasmic "leaking" of centromere proteins in limited cutaneous SSc (lcSSc) fibroblasts. Cytoplasmic centromere proteins co-localize with antigen presenting MHC Class II molecules, which correlate precisely with the presence of anti-centromere antibodies. CENPA expression and micronuclei formation correlate highly with activation of the cGAS-STING/IFN-ß pathway as well as markers of reactive oxygen species (ROS) and fibrosis, ultimately suggesting a link between centromere alterations, chromosome instability, SSc autoimmunity, and fibrosis.


Assuntos
Esclerodermia Difusa , Escleroderma Sistêmico , Humanos , Escleroderma Sistêmico/metabolismo , Instabilidade Cromossômica , Fibrose , Nucleotidiltransferases/genética
5.
Food Chem Toxicol ; 154: 112326, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34111490

RESUMO

Fumonisin B1 (FB1) is a well-known mycotoxin produced by Fusarium spp. and has a wide range of dose-dependent toxic effects, including nephrotoxicity, hepatotoxicity, and neurotoxicity. This research illustrated that FB1 exerts its toxicity in the neuroblastoma cell line through a distinct cell-death pathway called parthanatos. FB1 can cause excessive DNA strand breaks, leading to poly (ADP-ribose) polymerase-1 (PARP-1) overactivation and cell death. In this study, we used 50 µM FB1-treated SH-SY5Y neuroblastoma cells to elucidate the signaling pathway of FB1-induced parthanatos. We observed that FB1-induced cell death is caspase-independent and accompanied by rapid activation of PARP-1, c-Jun N-terminal kinase activation, reactive oxygen species (ROS) generation, and intracellular calcium increase. FB1 treatment also increased endoplasmic reticulum stress due to the rapid increase of calcium ions and ROS levels. In addition, FB1 induced massive DNA damage and chromatin decondensation. We also observed that apoptosis-inducing factor nuclear translocation and PAR accumulation were associated with the necroptosis signal.


Assuntos
Fumonisinas/farmacologia , Neuroblastoma/metabolismo , Parthanatos/efeitos dos fármacos , Cálcio/metabolismo , Linhagem Celular Tumoral , Cromatina/metabolismo , DNA/metabolismo , Dano ao DNA/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Necroptose/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo
6.
Cancer Res ; 81(4): 945-955, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33184107

RESUMO

The Wilms' tumor 1 (WT1) gene is well known as a chameleon gene. It plays a role as a tumor suppressor in Wilms' tumor but also acts as an oncogene in other cancers. Previously, our group reported that a canonical AUG starting site for the WT1 protein (augWT1) acts as a tumor suppressor, whereas a CUG starting site for the WT1 protein (cugWT1) functions as an oncogene. In this study, we report an oncogenic role of cugWT1 in the AOM/DSS-induced colon cancer mouse model and in a urethane-induced lung cancer model in mice lacking cugWT1. Development of chemically-induced tumors was significantly depressed in cugWT1-deficient mice. Moreover, glycogen synthase kinase 3ß promoted phosphorylation of cugWT1 at S64, resulting in ubiquitination and degradation of the cugWT1 associated with the F-box-/- WD repeat-containing protein 8. Overall, our findings suggest that inhibition of cugWT1 expression provides a potential candidate target for therapy. SIGNIFICANCE: These findings demonstrate that CUG-translated WT1 plays an oncogenic role in vivo, and GSK3ß-mediated phosphorylation of cugWT1 induces its ubiquitination and degradation in concert with FBXW8.


Assuntos
Glicogênio Sintase Quinase 3 beta/fisiologia , Neoplasias Renais/patologia , Proteínas WT1/genética , Tumor de Wilms/patologia , Células A549 , Animais , Células Cultivadas , Códon de Iniciação/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HEK293 , Células HeLa , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oncogenes/genética , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteólise , Ubiquitinação/genética , Proteínas WT1/química , Proteínas WT1/metabolismo , Tumor de Wilms/genética , Tumor de Wilms/metabolismo
7.
Int Immunopharmacol ; 65: 268-278, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30359933

RESUMO

In this study, the immunomodulatory effects of astemizole (AST) against lipopolysaccharide (LPS) mediated T cell proliferation and induction of inflammation in RAW macrophages (in vitro), and zebrafish larvae (in vivo) were determined. AST significantly suppressed the phagocytic activity of macrophages (3.303 ±â€¯0.115) and inhibited lysosomal enzyme secretion (13.27 ±â€¯2.52) induced by LPS (100 ng/ml). Moreover, AST subdued the morphological deformities such as yolk sac edema (YSE) and spinal curvature curving (SC) by inhibiting ROS generation in zebrafish larvae 24 h after microinjection of LPS (0.5 mg/ml). AST was also shown to inhibit the production of the major cytokines TNF-α (150.8 ±â€¯0.6), IL-1ß (276.5 ±â€¯1.6), and PGE2 (194.6 ±â€¯0.6) pg/ml in RAW macrophages. It also subdued the ROS induced iNOS and COX-2 generated in response to LPS mediated immune dysfunctions in zebrafish larvae. These results suggested the immunosuppression effect of AST. Furthermore, induction of immune-suppression due to AST resulted in significant down-regulation of innate immunity directed by MAPK (p38, ERK and JNK), which was found to be associated with decreased production of acute inflammatory mediators both in vitro and in vivo. To confirm its activity, splenocytes were prepared using BALB/c mice and a mitogen activated splenocyte proliferation assay was also performed. Our findings suggest that AST has the ability to inhibit T cell proliferation and cytokine secretion both in vitro and in vivo by interfering with MAPK signaling pathway. Taken together, our results showed the potential of AST as a countermeasure to immune dysfunction and suggest its use as immunosuppressant compound in inflammatory disease.


Assuntos
Astemizol/farmacologia , Imunossupressores/farmacologia , Macrófagos/imunologia , Baço/imunologia , Linfócitos T/imunologia , Peixe-Zebra/imunologia , Animais , Proliferação de Células , Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Peixes/metabolismo , Larva , Lipopolissacarídeos/imunologia , Ativação Linfocitária , Camundongos , Células RAW 264.7 , Transdução de Sinais , Baço/patologia , Linfócitos T/efeitos dos fármacos
8.
Anticancer Agents Med Chem ; 17(14): 1942-1950, 2018 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-28356011

RESUMO

BACKGROUND: Colon cancer is one of the most deadly and common carcinomas occurring worldwide and there have been many attempts to treat this cancer. The present work was designed in order to evaluate thymol as a potent drug against colon cancer. MATERIALS AND METHODS: Cytotoxicity of thymol at different concentrations was evaluated against a human colon carcinoma cell line (HCT-116 cells). Fluorescent staining was carried out to evaluate the level of ROS as well as mitochondrial and DNA fragmentation and immunoblot analysis were performed to confirm apoptosis and mitoptosis. RESULTS AND CONCLUSION: Results of the study demonstrated that thymol efficiently created an oxidative stress environment inside HCT-116 cells, a colorectal carcinoma cell line, through induction of ROS production along with intense damage to DNA and mitochondria, as observed through Hoechst and rhodamine 123 staining, respectively. Moreover, expression of PARP-1, p-JNK, cytochrome-C and caspase-3 proteins was up-regulated, suggesting HCT-116 cells underwent mitoptotic cell death. Therefore, thymol could be used as a potent drug against colon cancer due to its lower toxicity and prevalence in natural medicinal plants.


Assuntos
Antineoplásicos/farmacologia , Timol/farmacologia , Antineoplásicos/química , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colorimetria , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células HCT116 , Humanos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade , Timol/química , Células Tumorais Cultivadas
9.
Oncotarget ; 9(3): 3278-3291, 2018 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-29423046

RESUMO

Cancer treatment is limited due to the diverse multidrug resistance acquired by cancer cells and the collateral damage caused to adjacent normal cells by chemotherapy. The flavonoid compound vitexin exhibits anti-oxidative, anti-inflammatory and anti-tumor activity. This study elucidated the antitumor effects of vitexin and its underlying mechanisms in a multi-drug resistant human colon cancer cell line (HCT-116DR), which exhibits higher levels of multidrug-resistant protein 1 (MDR1) expression as compared with its parental cell line (HCT-116). Here, we observed that vitexin suppressed MDR-1 expression and activity in HCT-116DR cells and showed cytotoxic effect in HCT-116DR cells by inhibiting autophagy and inducing apoptosis in a concentration-dependent manner. Additionally, vitexin treatment caused cleavage of caspase-9 and caspase-3, and upregulated the expression of the pro-apoptotic proteins, BID and Bax. Moreover, the expression of autophagy-related proteins, such as ATG5, Beclin-1 and LC3-II, was markedly reduced by vitexin treatment. Furthermore, in vivo experiments showed that vitexin induced apoptosis and suppressed tumor growth in HCT-116DR xenograft model. These results revealed that vitexin induced apoptosis through suppression of autophagy in vitro and in vivo and provide insight into the therapeutic potential of vitexin for the treatment of chemo-resistant colorectal cancer.

10.
Oncotarget ; 8(68): 112426-112441, 2017 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-29348836

RESUMO

Heat shock transcription factor-1 (HSF-1) guards the cancerous cells proteome against the alterations in protein homeostasis generated by their hostile tumor microenvironment. Contrasting with the classical induction of heat shock proteins, the pro-oncogenic activities of HSF-1 remains to be explored. Therefore, cancer's fragile proteostatic pathway governed by HSF-1 could be a potential therapeutic target and novel biomarker by natural compounds. Vitexin, a natural flavonoid has been documented as a potent anti-tumor agent on various cell lines. However, in the present study, when human colorectal carcinoma HCT-116 cells were exposed to vitexin, the induction of HSF-1 downstream target proteins, such as heat shock proteins were suppressed. We identified HSF-1 as a potential molecular target of vitexin that interact with DNA-binding domain of HSF-1, which inhibited HSF-1 oligomerization and activation (in silico). Consequently, HSF-1 hyperphosphorylation mediated by JNK operation causes transcriptional inactivation of HSF-1, and supported ROS-mediated autophagy induction. Interestingly, in HSF-1 immunoprecipitated and silenced HCT-116 cells, co-expression of apolipoprotein 1 (ApoL1) and JNK was observed which promoted the caspase independent autophagic cell death accompanied by p62 downregulation and increased LC3-I to LC3-II conversion. Finally, in vivo findings confirmed that vitexin suppressed tumor growth through activation of autophagic cascade in HCT-116 xenograft model. Taken together, our study insights a probable novel association between HSF-1 and ApoL-1 was established in this study, which supports HSF-1 as a potential target of vitexin to improve treatment outcome in colorectal cancer.

11.
Cancer Lett ; 372(1): 89-100, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26739061

RESUMO

Apoptosis and autophagy are genetically regulated, evolutionarily conserved processes that can jointly seal cancer cell fates, and numerous death stimuli are capable of activating either pathway. Although crosstalk between apoptosis and autophagy is quite complex and sometimes contradictory, it remains a key factor determining the outcomes of death-related pathologies such as cancer. In the present study, exposure of MCF-7 breast cancer cells to HIS and the H1 receptor antagonist AST both alone and together with HIS (AST-HIS) led to generation of intracellular ROS, which induced massive cellular vacuolization through dilation of the ER and mitochondria. Consequently, apoptosis by Bax translocation, cytochrome c release, and caspase activation were triggered. In addition, AST-HIS caused ER stress-induced autophagy in MCF-7 cells, as evidenced by an increased LC3-II/LC3-I ratio, with surprisingly no changes in Beclin-1 expression. Non-canonical autophagy was induced via p53 phosphorylation, which increased p53-p62 interactions to enhance Beclin-1-independent autophagy as evidenced by immunocytochemistry and immunoprecipitation. In the absence of Beclin-1, enhanced autophagy further activated apoptosis through caspase induction. In conclusion, these findings indicate that AST-HIS-induced apoptosis and autophagy can be regulated by ROS-mediated signaling pathways.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/efeitos dos fármacos , Astemizol/farmacologia , Autofagia/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Antagonistas não Sedativos dos Receptores H1 da Histamina/farmacologia , Histamina/farmacologia , Proteínas de Membrana/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteína Beclina-1 , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspases/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células MCF-7 , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Proteína Sequestossoma-1 , Transdução de Sinais/efeitos dos fármacos , Transfecção , Proteína Supressora de Tumor p53/genética
12.
PLoS One ; 11(4): e0154525, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27116119

RESUMO

Plant-derived compounds are an important source of clinically useful anti-cancer agents. Chrysin, a biologically active flavone found in many plants, has limited usage for cancer chemotherapeutics due to its poor oral bioavailability. 5-Hydroxy-7-methoxyflavone (HMF), an active natural chrysin derivative found in various plant sources, is known to modulate several biological activities. However, the mechanism underlying HMF-induced apoptotic cell death in human colorectal carcinoma cells in vitro is still unknown. Herein, HMF was shown to be capable of inducing cytotoxicity in HCT-116 cells and induced cell death in a dose-dependent manner. Treatment of HCT-116 cells with HMF caused DNA damage and triggered mitochondrial membrane perturbation accompanied by Cyt c release, down-regulation of Bcl-2, activation of BID and Bax, and caspase-3-mediated apoptosis. These results show that ROS generation by HMF was the crucial mediator behind ER stress induction, resulting in intracellular Ca2+ release, JNK phosphorylation, and activation of the mitochondrial apoptosis pathway. Furthermore, time course study also reveals that HMF treatment leads to increase in mitochondrial and cytosolic ROS generation and decrease in antioxidant enzymes expression. Temporal upregulation of IRE1-α expression and JNK phosphorylation was noticed after HMF treatment. These results were further confirmed by pre-treatment with the ROS scavenger N-acetyl-l-cysteine (NAC), which completely reversed the effects of HMF treatment by preventing lipid peroxidation, followed by abolishment of JNK phosphorylation and attenuation of apoptogenic marker proteins. These results emphasize that ROS generation by HMF treatment regulates the mitochondrial-mediated apoptotic signaling pathway in HCT-116 cells, demonstrating HMF as a promising pro-oxidant therapeutic candidate for targeting colorectal cancer.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias do Colo/metabolismo , Flavonoides/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Apoptose , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Relação Dose-Resposta a Droga , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos
13.
Biomed Pharmacother ; 84: 789-799, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27721177

RESUMO

Although quercetin is very well known for its anticancer activity, however it shows some drawbacks. Herein, we have evaluated the apoptotic effect TEF (5, 3'-dihydroxy-3, 7, 4'-triethoxyflavone), a newly synthesized quercetin derivative on HCT-116 colon cancer cells. After 24h of treatment, the proliferation of colon cancer cells was inhibited by TEF. TEF induced apoptosis, as confirmed by the presence of fragmented nuclei, reduced mitochondrial membrane potential, and elevated cytoplasmic and mitochondrial reactive oxygen species (ROS) levels. TEF treatment causes elevation of IRE1-α and activates calcium ions (Ca2+) with concomitant increase in JNK levels. Elevated Ca2+ ion translocates from ER to mitochondria which leads to ROS release and oxidative stress. TEF treatment further elevated levels of pro-apoptotic factors and down-regulated the level of Bcl2. TEF led to activation of mito-JNK (mitochondrial JNK), which plays a crucial role in activation of oxidative stress and caspase mediated apoptotic cell death. Moreover, JNK inhibition shown to suppress TEF induced apoptosis in HCT-116 colon cancer cells. Therefore, this study reveals the apoptotic role of TEF against HCT-116 cell line via IRE1-α and mito-JNK pathway.


Assuntos
Apoptose/fisiologia , Neoplasias do Colo/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Mitocôndrias/metabolismo , Quercetina/análogos & derivados , Quercetina/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Relação Dose-Resposta a Droga , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células HCT116 , Humanos , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Silanos/química , Silanos/farmacologia
14.
Cancer Res ; 76(5): 1146-1157, 2016 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-26676750

RESUMO

Ornithine decarboxylase (ODC) is a rate-limiting enzyme in the first step of polyamine biosynthesis that is associated with cell growth and tumor formation. Existing catalytic inhibitors of ODC have lacked efficacy in clinical testing or displayed unacceptable toxicity. In this study, we report the identification of an effective and nontoxic allosteric inhibitor of ODC. Using computer docking simulation and an in vitro ODC enzyme assay, we identified herbacetin, a natural compound found in flax and other plants, as a novel ODC inhibitor. Mechanistic investigations defined aspartate 44 in ODC as critical for binding. Herbacetin exhibited potent anticancer activity in colon cancer cell lines expressing high levels of ODC. Intraperitoneal or oral administration of herbacetin effectively suppressed HCT116 xenograft tumor growth and also reduced the number and size of polyps in a mouse model of APC-driven colon cancer (ApcMin/+). Unlike the well-established ODC inhibitor DFMO, herbacetin treatment was not associated with hearing loss. Taken together, our findings defined the natural product herbacetin as an allosteric inhibitor of ODC with chemopreventive and antitumor activity in preclinical models of colon cancer, prompting its further investigation in clinical trials.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Flavonoides/farmacologia , Inibidores da Ornitina Descarboxilase/farmacologia , Regulação Alostérica , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anticarcinógenos/farmacologia , Eflornitina/farmacologia , Feminino , Flavonoides/toxicidade , Células HCT116 , Perda Auditiva/induzido quimicamente , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ornitina Descarboxilase/química , Ornitina Descarboxilase/metabolismo
15.
Free Radic Biol Med ; 89: 1218-30, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26561775

RESUMO

Aflatoxin B1 (AFB1) is the most toxic aflatoxin species and has been shown to be associated with specific as well as non-specific immune responses. In the present study, using murine macrophage Raw 264.7 cells as a model, we report that short exposure (6h) to AFB1 caused an increase in the cellular calcium pool in mitochondria, which in turn elevated reactive oxygen species (ROS)-mediated oxidative stress and led to loss of mitochondrial membrane potential and ultimately c-Jun N-terminal kinases (JNK)-mediated caspase-dependent cell death. On the contrary, longer exposure (12h) to AFB1 reduced JNK phosphorylation and cell death in macrophages. Measurement of autophagic flux demonstrated that autophagy induction through the canonical pathway was responsible for suppressing AFB1-induced apoptosis after 12h. As a detailed molecular mechanism, we found that the unfolded protein response (UPR) machinery was active at 12h post-exposure to AFB1 and induced cytoprotective autophagy as confirmed by determination of major autophagic markers. Inhibition of autophagy by Beclin-1 siRNA also resulted in JNK-mediated cell death. We further established that glutathione S transferase omega1-1 (GSTO1-1), a specific class of GST, was the responsible factor between apoptosis and autophagy crosstalk. Targeting of GSTO1-1 increased JNK-mediated apoptosis by 2-fold compared to the control, whereas autophagy rate was reduced. Thus, increased expression of GSTO1-1 was associated with increased protein glutathionylation, an important protein modification in response to cellular redox status.


Assuntos
Aflatoxina B1/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Glutationa Transferase/metabolismo , Macrófagos/efeitos dos fármacos , Venenos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Glutationa/metabolismo , Imunoprecipitação , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional , RNA Mensageiro/genética , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Life Sci ; 142: 36-48, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26475763

RESUMO

AIMS: Cells possess multiple methods for counteracting the deleterious consequences of stress induced by physical and chemical stimuli. Heat stress causes variations in the cellular environment, leading to cellular morbidity or mortality. Natural compounds that contain phenolic antioxidants, offer various therapeutic and biological activities. Vitexin, a natural flavonoid, has been reported to treat various pathologies due to its multifaceted effects. Herein, we investigated the therapeutic efficacy of vitexin and its underlying mechanism against heat stress in human lung epithelial cells. MAIN METHODS: Effect of vitexin on the expression of molecular chaperones, antioxidant enzymes, mitogen activated protein kinases (MAPKs), endoplasmic reticulum (ER)-stress and autophagy was measured by immunoblotting. qRT-PCR and EMSA were performed for Hsp90 expression and HSF-1 binding affinity. Cell viability was assessed by MTT and LDH assays. Detection of autophagy was confirmed by acridine orange staining. Role of Hsp90 inhibition on signaling pathways was elucidated by using specific chemical inhibitor, radicicol. KEY FINDINGS: Whereas hyperthermia reduced cell viability, result of MTT and LDH assays showed that vitexin pre-treatment enhanced cell viability after heat stress. EMSA analysis shows DNA binding affinity of HSF-1 during heat stress. Vitexin upregulated Hsp90 expression, subsequently activating ER-stress induced autophagy. Modulation of MAPKs expression and fluorescence image analysis showed vacuole accumulation, indicating autophagic flux in cells. Hsp90 inhibition reversed the effect of vitexin and activates the apoptosis pathway. SIGNIFICANCE: Our data suggest that vitexin can protect against hyperthermic cellular injury by induction of Hsp90 expression, antioxidant activity and MAPKs via ER stress-induced autophagy.


Assuntos
Apigenina/farmacologia , Autofagia/efeitos dos fármacos , Citotoxinas/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Resposta ao Choque Térmico/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Transcrição de Choque Térmico , Humanos , Fatores de Transcrição/metabolismo
17.
Saudi J Biol Sci ; 22(6): 706-13, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26586997

RESUMO

In this study, the effect of purified quercetin-3-O-ß-d-glucopyranosyl-(1 â†’ 6)-ß-d-glucopyranosid (QCGG) on melanogenesis was investigated. QCGG was isolated from the calyx of a traditional Korean medicinal herb, Persimmon (Diospyros kaki). The hypopigmentation effects of QCGG were determined by examination of cellular melanin contents, tyrosinase activity assay, cAMP assay, and Western blotting of α-MSH-stimulated B16F10 mouse melanoma cells. Our results showed that QCGG inhibited both melanin synthesis and tyrosinase activity in a concentration-dependent manner as well as significantly reduced the expression of melanogenic proteins such as microphthalmia-associated transcription factor (MITF), tyrosinase-related protein-1, tyrosinase-related protein-2, and tyrosinase. Moreover, QCGG inhibited intracellular cAMP levels, cAMP response element-binding protein (CREB), and p38 MAPK expression in α-MSH-stimulated B16F10 cells. Taken together, the suppressive effects of QCGG on melanogenesis may involve down-regulation of MITF and its downstream signaling pathway via phosphorylation of p38 MAPK and CREB along with reduced cAMP levels. These results indicate that QCGG reduced melanin synthesis by reducing expression of tyrosine and tyrosine-related proteins via extracellular signal-related protein kinase (ERK) activation, followed by down-regulation of CREB, p38, and MITF.

18.
Int Immunopharmacol ; 18(2): 340-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24316253

RESUMO

The potent role of thymol, a natural compound, in modulation of macrophage activity was evaluated by determining all the sequential steps involved during phagocytosis. We found a significant increase in the proliferation of splenocytes in the presence of thymol and it proved to be a good mitogen. Uptake capacity of macrophages was enhanced due to increased membrane fluidity after treatment with thymol and it also increases lysosomal activity of macrophages. Data of superoxide anion generation revealed the involvement of thymol in the generation of respiratory burst as it potentiated this property of macrophages at a concentration of 150 µM. In the case of TNF-α, IL-1ß and PGE(2) a decreased level of secretion was observed 154 ρg/ml, 736.1 ρg/ml, and 151 ρg/ml respectively when compared with lipopolysaccharide treated cells, where the level of these cytokines was significantly high. We also determined the anti-complementary activity of thymol which showed to be more effective than rosmarinic acid. Thus, the results obtained from the study suggest the potential role of thymol as a natural immunostimulatory drug which can be used in the treatment of various immunological disorders.


Assuntos
Adjuvantes Imunológicos/farmacologia , Macrófagos/efeitos dos fármacos , Mitógenos/farmacologia , Fagocitose/efeitos dos fármacos , Timol/farmacologia , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Interleucina-1beta/metabolismo , Lisossomos/metabolismo , Macrófagos/fisiologia , Fluidez de Membrana , Camundongos , Baço/citologia , Fator de Necrose Tumoral alfa/metabolismo
19.
Int Immunopharmacol ; 22(2): 356-65, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25068824

RESUMO

Morin, a natural flavonoid that is the primary bioactive constituent of the family Moraceae, has been found to be associated with many therapeutic properties. In this study, we evaluated the immunomodulatory activities of increasing concentration of morin hydrate in vitro. Three different concentrations of morin hydrate (5, 10, and 15µM) were used to evaluate their effect on splenocyte proliferation, phagocytic activity of macrophages, cytokine secretion and complement inhibition. We also evaluated the role of morin hydrate on lipopolysaccharide (LPS) induced autophagy. Our study demonstrated that morin hydrate elicited a significant increase in splenocyte proliferation, phagocytic capacity and suppressed the production of cytokines and nitric oxide in activated macrophages. Humoral immunity measured by anti-complement activity showed an increase in inhibition of the complement system after the addition of morin hydrate, where morin hydrate at 15µM concentration induced a significant inhibition. Depending on our results, we can also conclude that morin hydrate protects macrophages from LPS induced autophagic cell death. Our findings suggest that morin hydrate represents a structurally diverse class of flavonoid and this structural variability can profoundly affect its cell-type specificity and its biological activities. Supplementation of immune cells with morin hydrate has an upregulating and immunoprotective effect that shows potential as a countermeasure to the immune dysfunction and suggests an interesting use in inflammation related diseases.


Assuntos
Flavonoides/farmacologia , Fatores Imunológicos/farmacologia , Macrófagos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Dinoprostona/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos , Macrófagos/fisiologia , Camundongos , Óxido Nítrico/metabolismo , Explosão Respiratória/efeitos dos fármacos , Baço/citologia , Fator de Necrose Tumoral alfa/metabolismo
20.
J Photochem Photobiol B ; 131: 96-103, 2014 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-24514458

RESUMO

DNA protection is one of the most important strategies in cancer therapy. Since quercetin and its derivatives are found to be potent antioxidant agents, they are able to scavenge radicals significantly. Therefore, we focused on the DNA protection activity of 3,5,7,3',4'-pentamethoxyflavone (PMF), a quercetin derivative isolated from Kaemperia parviflora. Although, PMF was found to be a very poor antioxidant compound, still it could remarkably protect DNA from oxidative damage. DNA binding assay showed that PMF bound to the minor groove of DNA, which suggests a possible mechanism for its DNA protective effects. Cellular toxicity assay on RAW 264.7 macrophages showed this compound is very safe for therapeutic applications.


Assuntos
Dano ao DNA/efeitos dos fármacos , DNA/metabolismo , Flavonas/farmacologia , Animais , Antioxidantes/farmacologia , Benzotiazóis/química , Compostos de Bifenilo/química , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ensaio Cometa , Relação Dose-Resposta a Droga , Sequestradores de Radicais Livres/farmacologia , Camundongos , Óxido Nítrico/metabolismo , Picratos/química , Substâncias Protetoras/farmacologia , Espectrofotometria Ultravioleta , Ácidos Sulfônicos/química , Testes de Toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA