Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Lancet Oncol ; 23(9): 1189-1200, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35952709

RESUMO

BACKGROUND: TGF-ß is an immunosuppressive cytokine that is upregulated in colorectal cancer. TGF-ß blockade improved response to chemoradiotherapy in preclinical models of colorectal adenocarcinoma. We aimed to test the hypothesis that adding the TGF-ß type I receptor kinase inhibitor galunisertib to neoadjuvant chemoradiotherapy would improve pathological complete response rates in patients with locally advanced rectal cancer. METHODS: This was an investigator-initiated, single-arm, phase 2 study done in two medical centres in Portland (OR, USA). Eligible patients had previously untreated, locally advanced, rectal adenocarcinoma, stage IIA-IIIC or IV as per the American Joint Committee on Cancer; Eastern Cooperative Oncology Group status 0-2; and were aged 18 years or older. Participants completed two 14-day courses of oral galunisertib 150 mg twice daily, before and during fluorouracil-based chemoradiotherapy (intravenous fluorouracil 225 mg/m2 over 24 h daily 7 days per week during radiotherapy or oral capecitabine 825 mg/m2 twice per day 5 days per week during radiotherapy; radiotherapy consisted of 50·4-54·0 Gy in 28-30 fractions). 5-9 weeks later, patients underwent response assessment. Patients with a complete response could opt for non-operative management and proceed to modified FOLFOX6 (intravenous leucovorin 400 mg/m2 on day 1, intravenous fluorouracil 400 mg/m2 on day 1 then 2400 mg/m2 over 46 h, and intravenous oxaliplatin 85 mg/m2 on day 1 delivered every 2 weeks for eight cycles) or CAPEOX (intravenous oxaliplatin 130 mg/m2 on day 1 and oral capecitabine 1000 mg/m2 twice daily for 14 days every 3 weeks for four cycles). Patients with less than complete response underwent surgical resection. The primary endpoint was complete response rate, which was a composite of pathological complete response in patients who proceeded to surgery, or clinical complete response maintained at 1 year after last therapy in patients with non-operative management. Safety was a coprimary endpoint. Both endpoints were assessed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT02688712, and is active but not recruiting. FINDINGS: Between Oct 19, 2016, and Aug 31, 2020, 38 participants were enrolled. 25 (71%) of the 35 patients who completed chemoradiotherapy proceeded to total mesorectal excision surgery, five (20%) of whom had pathological complete responses. Ten (29%) patients had non-operative management, three (30%) of whom ultimately chose to have total mesorectal excision. Two (67%) of those three patients had pathological complete responses. Of the remaining seven patients in the non-operative management group, five (71%) had clinical complete responses at 1 year after their last modified FOLFOX6 infusion. In total, 12 (32% [one-sided 95% CI ≥19%]) of 38 patients had a complete response. Common grade 3 adverse events during treatment included diarrhoea in six (16%) of 38 patients, and haematological toxicity in seven (18%) patients. Two (5%) patients had grade 4 adverse events, one related to chemoradiotherapy-induced diarrhoea and dehydration, and the other an intraoperative ischaemic event. No treatment-related deaths occurred. INTERPRETATION: The addition of galunisertib to neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer improved the complete response rate to 32%, was well tolerated, and warrants further assessment in randomised trials. FUNDING: Eli Lilly via ExIST program, The Providence Foundation.


Assuntos
Adenocarcinoma , Segunda Neoplasia Primária , Neoplasias Retais , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Capecitabina , Quimiorradioterapia/efeitos adversos , Diarreia/etiologia , Fluoruracila , Humanos , Terapia Neoadjuvante/efeitos adversos , Estadiamento de Neoplasias , Segunda Neoplasia Primária/patologia , Oxaliplatina , Pirazóis , Quinolinas , Neoplasias Retais/tratamento farmacológico , Neoplasias Retais/patologia , Fator de Crescimento Transformador beta
2.
Saudi Pharm J ; 30(6): 815-824, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35812146

RESUMO

Thiosemicarbazones have received noteworthy attention due to their numerous pharmacological activities. Various thiosemicarbazone derivatives have been reported to play a key role as potential chemotherapeutic agents for the management of cancer. Herein, we aimed to establish the anticancer efficacy of novel thiosemicarbazone derivative C4 against colon cancer in vitro. The MTT viability assay identified C4 as a promising anticancer compound in a panel of cancer cell lines with the most potent activity against colon cancer cells. Further, anticancer potential of C4 was evaluated against HT-29 and SW620 colon cancer cell lines considering the factors like cell adhesion and migration, oxidative stress, cell cycle arrest, and apoptosis. Our results showed that C4 significantly inhibited the migration and adhesion of colon cancer cells. C4 significantly increased the intracellular reactive oxygen species (ROS) and induced apoptotic cell death. Cell cycle analysis revealed that C4 interfered in the cell cycle distribution and arrested the cells at the G2/M phase of the cell cycle. Consistent with these results C4 also down-regulated the Bcl-XL and Bcl-2 and up-regulated the caspase-3 expression. These findings introduced C4 as the potential anticancer agent against colon cancer.

3.
J Enzyme Inhib Med Chem ; 36(1): 1424-1435, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34176414

RESUMO

In the current work, a new set of carbohydrazide linked benzofuran-isatin conjugates (5a-e and 7a-i) was designed and synthesised. The anticancer activity for compounds (5b-d, 7a, 7b, 7d and 7g) was measured against NCI-55 human cancer cell lines. Compound 5d was the most efficient, and thus subjected to the five-dose screen where it showed excellent broad activity against almost all tested cancer subpanels. Furthermore, all conjugates (5a-e and 7a-i) showed a good anti-proliferative activity towards colorectal cancer SW-620 and HT-29 cell lines, with an excellent inhibitory effect for compounds 5a and 5d (IC50 = 8.7 and 9.4 µM (5a), and 6.5 and 9.8 µM for (5d), respectively). Both compounds displayed selective cytotoxicity with good safety profile. In addition, both compounds provoked apoptosis in a dose dependent manner in SW-620 cells. Also, they significantly inhibited the anti-apoptotic Bcl2 protein expression and increased the cleaved PARP level that resulted in SW-620 cells apoptosis.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzofuranos/química , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/patologia , Isatina/química , Antineoplásicos/química , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Desenvolvimento de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
4.
J Enzyme Inhib Med Chem ; 36(1): 319-328, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33345633

RESUMO

Mitochondrial anti-apoptotic Bcl2 and BclxL proteins, are overexpressed in multiple tumour types, and has been involved in the progression and survival of malignant cells. Therefore, inhibition of such proteins has become a validated and attractive target for anticancer drug discovery. In this manner, the present studies developed a series of novel isatin-indole conjugates (7a-j and 9a-e) as potential anticancer Bcl2 and BclxL inhibitors. The progression of the two examined colorectal cancer cell lines was significantly inhibited by all of the prepared compounds with IC50 ranges132-611 nM compared to IC50 = 4.6 µM for 5FU, against HT-29 and IC50 ranges 37-468 nM compared to IC50 = 1.5 µM for 5FU, against SW-620. Thereafter, compounds 7c and 7g were selected for further investigations. Interestingly, both compounds exhibited selective cytotoxicity against both cell lines with high safety to normal fibroblast (HFF-1). In addition, both compounds 7c and 7g induced apoptosis and inhibited Bcl2 and BclxL expression in a dose-dependent manner. Collectively, the high potency and selective cytotoxicity suggested that conjugates 7c and 7g could be a starting point for further optimisation to develop novel pro-apoptotic and antitumor agents towards colon cancer.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Desenvolvimento de Medicamentos , Hidrazinas/farmacologia , Indóis/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Hidrazinas/síntese química , Hidrazinas/química , Indóis/síntese química , Indóis/química , Estrutura Molecular , Relação Estrutura-Atividade , Células Tumorais Cultivadas
5.
Cancer Cell Int ; 20: 126, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32322173

RESUMO

BACKGROUND: Colorectal carcinoma is one of the most deadly cancers that requests effective and safe chemotherapy. Evaluation of natural product-based anticancer drugs as adjuvant treatment with fewer side effects is largely unexplored research fields. Herbal melanin (HM) is an extract of the seed coats of Nigella sativa that modulates an inflammatory response through toll-like receptor 4 (TLR4). This TLR4 receptor is also involved in the modulation of apoptosis. We therefore explored the anticancer potential of HM and specifically its effect on the molecular mechanisms underlying adenocarcinoma and metastatic colorectal cancer (mCRC) cell death in vitro. METHODS: Cell viability was evaluated using the MTT assay. Cellular reactive oxygen species (ROS), glutathione levels, and apoptotic status were assessed using fluorometric and colorimetric detection methods. HM-induced apoptotic and other signaling pathways were investigated using Western blot technology and mitochondrial transition pore assay kit. TLR4 receptor downregulation and blockade were performed using siRNA technology and neutralizing antibody, respectively. RESULTS: Our results showed that HM inhibited the proliferation of the colorectal adenocarcinoma HT29 and mCRC SW620 cell lines. Furthermore, HM enhanced ROS production and decreased glutathione levels. HM-induced apoptosis was associated with mitochondrial outer membrane permeability and cytochrome c release, inhibition of the Bcl2 family proteins, and activation of caspase-3/-7. In addition, HM modulated MAPK pathways by activating the JNK pathway and by inhibiting ERK phosphorylation. TLR4 receptor downregulation enhanced HM-induced apoptosis while TLR4 receptor blockade partially alleviated HM-inhibited ERK phosphorylation. CONCLUSION: Altogether, these findings indicate that HM exerts pro-apoptotic effects and inhibits MAPK pathway through TLR4 in mCRC and colorectal adenocarcinoma cells, suggesting HM as a promising natural-based drug for the treatment of colorectal cancer.

7.
BMC Cancer ; 20(1): 685, 2020 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-32703189

RESUMO

BACKGROUND: Colorectal cancer (CRC) is a heterogeneous tumor having various genetic alterations. The current treatment options had limited impact on disease free survival due to therapeutic resistance. Novel anticancer agents are needed to treat CRC specifically metastatic colorectal cancer. A novel coordination complex of platinum, (salicylaldiminato)Pt(II) complex with dimethylpropylene linkage (PT) exhibited potential anti-cancer activity. In this study, we explored the molecular mechanism of PT-induced cell death in colorectal cancer. METHODS: Colony formation was evaluated using the clonogenic assay. Apoptosis, cell cycle analysis, reactive oxygen species, mitochondrial membrane potential and caspase-3/- 7 were assessed by flow cytometry. Glutathione level was detected by colorimetric assay. PT-induced alteration in pro-apoptotic/ anti-apoptotic proteins and other signaling pathways were investigated using western blotting. P38 downregulation was performed using siRNA. RESULTS: In the present study, we explored the molecular mechanism of PT-mediated inhibition of cell proliferation in colorectal cancer cells. PT significantly inhibited the colony formation in human colorectal cancer cell lines (HT-29, SW480 and SW620) by inducing apoptosis and necrosis. This platinum complex was shown to significantly increase the reactive oxygen species (ROS) generation, depletion of glutathione and reduced mitochondrial membrane potential in colorectal cancer cells. Exposure to PT resulted in the downregulation of anti-apoptotic proteins (Bcl2, BclxL, XIAP) and alteration in Cyclins expression. Furthermore, PT increased cytochrome c release into cytosol and enhanced PARP cleavage leading to activation of intrinsic apoptotic pathway. Moreover, pre-treatment with ROS scavenger N-acetylcysteine (NAC) attenuated apoptosis suggesting that PT-induced apoptosis was driven by oxidative stress. Additionally, we show that PT-induced apoptosis was mediated by activating p38 MAPK and inhibiting AKT pathways. This was demonstrated by using chemical inhibitor and siRNA against p38 kinase which blocked the cytochrome c release and apoptosis in colorectal cancer cells. CONCLUSION: Collectively, our data demonstrates that the platinum complex (PT) exerts its anti-proliferative effect on CRC by ROS-mediated apoptosis and activating p38 MAPK pathway. Thus, our findings reveal a novel mechanism of action for PT on colorectal cancer cells and may have therapeutic implication.


Assuntos
Morte Celular , Neoplasias Colorretais/tratamento farmacológico , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Compostos de Platina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Anexina A5/análise , Apoptose/efeitos dos fármacos , Apoptose/genética , Caspase 3/metabolismo , Caspase 7/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/química , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Ciclinas/metabolismo , Regulação para Baixo , Glutationa/metabolismo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Oxirredução , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ensaio Tumoral de Célula-Tronco , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Proteína bcl-X/metabolismo
8.
Physiol Plant ; 168(2): 289-300, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31209886

RESUMO

The present study evaluated the physiological and biochemical mechanisms through which exogenous sodium hydrosulfide (H2 S donor) mitigates chromium (Cr) stress in cauliflower. The different levels of Cr included 0, 10, 100 and 200 µM. Results reported that Cr exposure reduced growth and biomass, chlorophyll (Chl) contents, gas exchange parameters and enzymatic antioxidants. Chromium stress enhanced the production of electrolyte leakage (EL), hydrogen peroxide (H2 O2 ) and malondialdehyde (MDA) contents and increased Cr content in the roots, stem, leaf and flowers. Exogenous H2 S improved the physiological and biochemical attributes of Cr-stressed cauliflower. Hydrogen sulfide decreased Cr content in different parts of Cr-stressed plants, whereas it increased the Chl contents and gas exchange attributes. H2 S reduced the EL, H2 O2 and MDA concentrations, enhancing the antioxidant enzymes activities in Cr-stressed roots and leaves compared to the Cr treatments alone. Collectively, our results provide an insight into the protective role of H2 S in Cr-stressed cauliflower and suggest H2 S as a potential candidate in reducing Cr toxicity in cauliflower and other crops.


Assuntos
Antioxidantes/metabolismo , Brassica/efeitos dos fármacos , Cromo/toxicidade , Sulfeto de Hidrogênio/farmacologia , Poluentes do Solo/toxicidade , Clorofila , Peróxido de Hidrogênio , Malondialdeído , Estresse Oxidativo
9.
J Enzyme Inhib Med Chem ; 34(1): 672-683, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30821525

RESUMO

Some new 3H-quinazolin-4-one derivatives were synthesised and screened for anticancer, antiphospholipases, antiproteases, and antimetabolic syndrome activities. Compound 15d was more potent in reducing the cell viabilities of HT-29 and SW620 cells lines to 38%, 36.7%, compared to 5-FU which demonstrated cell viabilities of 65.9 and 42.7% respectively. The IC50 values of 15d were ∼20 µg/ml. Assessment of apoptotic activity revealed that 15d decreased the cell viability by down regulating Bcl2 and BclxL. Moreover, compounds, 8j, 8d/15a/15e, 5b, and 8f displayed lowered IC50 values than oleanolic acid against proinflammatory isoforms of hGV, hG-X, NmPLA2, and AmPLA2. In addition, 8d, 8h, 8j, 15a, 15b, 15e, and 15f showed better anti-α-amylase than quercetin, whereas 8g, 8h, and 8i showed higher anti-α-glucosidase activity than allopurinol. Thus, these compounds can be considered as potential antidiabetic agents. Finally, none of the compounds showed higher antiproteases or xanthine oxidase activities than the used reference drugs.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Síndrome Metabólica/tratamento farmacológico , Peptídeo Hidrolases/metabolismo , Fosfolipases/antagonistas & inibidores , Quinazolinonas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Células HT29 , Humanos , Síndrome Metabólica/metabolismo , Estrutura Molecular , Fosfolipases/metabolismo , Quinazolinonas/síntese química , Quinazolinonas/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas
10.
Sensors (Basel) ; 19(23)2019 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-31775385

RESUMO

Speaker diarization systems aim to find 'who spoke when?' in multi-speaker recordings. The dataset usually consists of meetings, TV/talk shows, telephone and multi-party interaction recordings. In this paper, we propose a novel multimodal speaker diarization technique, which finds the active speaker through audio-visual synchronization model for diarization. A pre-trained audio-visual synchronization model is used to find the synchronization between a visible person and the respective audio. For that purpose, short video segments comprised of face-only regions are acquired using a face detection technique and are then fed to the pre-trained model. This model is a two streamed network which matches audio frames with their respective visual input segments. On the basis of high confidence video segments inferred by the model, the respective audio frames are used to train Gaussian mixture model (GMM)-based clusters. This method helps in generating speaker specific clusters with high probability. We tested our approach on a popular subset of AMI meeting corpus consisting of 5.4 h of recordings for audio and 5.8 h of different set of multimodal recordings. A significant improvement is noticed with the proposed method in term of DER when compared to conventional and fully supervised audio based speaker diarization. The results of the proposed technique are very close to the complex state-of-the art multimodal diarization which shows significance of such simple yet effective technique.

11.
Mol Cancer ; 16(1): 33, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28153010

RESUMO

BACKGROUND: Colorectal cancer is third most common malignancy and is the second most common cause of cancer-related death. The MUC1 heterodimeric protein is aberrantly overexpressed in colorectal cancer and has been linked to poor outcomes in this disease. Here, we investigate the effects of the MUC1-C subunit inhibitor (GO-203), which disrupts MUC1-C homo-oligomerization, on human colorectal cancer cells. METHODS: TIGAR mRNA level was determined using qRT-PCR. Western blotting was used to measure TIGAR protein level and AKT-mTOR-S6K1 pathways. Reactive oxygen species and apoptosis were measured by flow cytometry. Effect of MUC1-C peptide, GO-203 was studied on colorectal xenograft tumors. Immunohistochemistry was utilized for TIGAR staining. RESULTS: Treatment of MUC1-overexpressing SKCO-1 and Colo-205 colon cancer cells with GO-203 was associated with downregulation of the TP53-inducible glycolysis and apoptosis regulator (TIGAR) protein. TIGAR promotes the shunting of glycolytic intermediates into the pentose phosphate pathway and thus is of importance for maintaining redox balance. We show that GO-203-induced suppression of TIGAR is mediated by inhibition of AKT and the downstream mTOR pathway. The results also demonstrate that targeting MUC1-C blocks eIF4A cap-dependent translation of TIGAR. In concert with these results, GO-203-induced suppression of TIGAR was associated with decreases in GSH levels. GO-203 treatment also resulted in increases in reactive oxygen species (ROS) and loss of mitochondrial transmembrane potential. Consistent with these results, GO-203 inhibited the growth of colon cancer cells in vitro and as xenografts in nude mice. Inhibition of MUC1-C also downregulated TIGAR expression in xenograft tissues. CONCLUSIONS: These findings indicate that MUC1-C is a potential target for the treatment of colorectal cancer. Colorectal cancer patients who overexpress MUC1-C may be candidates for treatment with the MUC1-C inhibitor alone or in combination therapy with other agents.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mucina-1/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Glutationa/metabolismo , Humanos , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Mucina-1/química , Mucina-1/genética , Oxirredução , Peptídeos/farmacologia , Monoéster Fosfórico Hidrolases , Biossíntese de Proteínas/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
BMC Cancer ; 17(1): 4, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28049506

RESUMO

BACKGROUND: Colorectal cancer (CRC) is the 3rd most common type of cancer worldwide. New anti-cancer agents are needed for treating late stage colorectal cancer as most of the deaths occur due to cancer metastasis. A recently developed compound, 3c has shown to have potent antitumor effect; however the mechanism underlying the antitumor effect remains unknown. METHODS: 3c-induced inhibition of proliferation was measured in the absence and presence NAC using MTT in HT-29 and SW620 cells and xCELLigence RTCA DP instrument. 3c-induced apoptotic studies were performed using flow cytometry. 3c-induced redox alterations were measured by ROS production using fluorescence plate reader and flow cytometry and mitochondrial membrane potential by flow cytometry; NADPH and GSH levels were determined by colorimetric assays. Bcl2 family protein expression and cytochrome c release and PARP activation was done by western blotting. Caspase activation was measured by ELISA. Cell migration assay was done using the real time xCELLigence RTCA DP system in SW620 cells and wound healing assay in HT-29. RESULTS: Many anticancer therapeutics exert their effects by inducing reactive oxygen species (ROS). In this study, we demonstrate that 3c-induced inhibition of cell proliferation is reversed by the antioxidant, N-acetylcysteine, suggesting that 3c acts via increased production of ROS in HT-29 cells. This was confirmed by the direct measurement of ROS in 3c-treated colorectal cancer cells. Additionally, treatment with 3c resulted in decreased NADPH and glutathione levels in HT-29 cells. Further, investigation of the apoptotic pathway showed increased release of cytochrome c resulting in the activation of caspase-9, which in turn activated caspase-3 and -6. 3c also (i) increased p53 and Bax expression, (ii) decreased Bcl2 and BclxL expression and (iii) induced PARP cleavage in human colorectal cancer cells. Confirming our observations, NAC significantly inhibited induction of apoptosis, ROS production, cytochrome c release and PARP cleavage. The results further demonstrate that 3c inhibits cell migration by modulating EMT markers and inhibiting TGFß-induced phosphorylation of Smad2 and Samd3. CONCLUSIONS: Our findings thus demonstrate that 3c disrupts redox balance in colorectal cancer cells and support the notion that this agent may be effective for the treatment of colorectal cancer.


Assuntos
Amidas/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Espécies Reativas de Oxigênio/metabolismo , Ácidos Sulfanílicos/química , Caspase 3/metabolismo , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Citocromos c/metabolismo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Células Tumorais Cultivadas , Cicatrização/efeitos dos fármacos
13.
Sci Technol Adv Mater ; 18(1): 364-373, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28634498

RESUMO

Cerium oxide nanocrystals (CeO2-NCs) exhibit superoxide dismutase and catalase mimetic activities. Based on these catalytic activities, CeO2-NCs have been suggested to have the potential to treat various diseases. The crystalline size of these materials is an important factor that influences the performance of CeO2-NCs. Previous reports have shown that several metal-based nanocrystals, including CeO2-NCs, can induce cytotoxicity in cancer cells. However, the underlying mechanisms have remained unclear. To characterize the anticancer activities of CeO2-NCs, several assays related to the mechanism of cytotoxicity and induction of apoptosis has been performed. Here, we have carried out a systematic study to characterize CeO2-NCs phase purity (X-ray diffraction), morphology (electron microscopy), and optical features (optical absorption, Raman scattering, and photoluminescence) to better establish their potential as anticancer drugs. Our study revealed anticancer effects of CeO2-NCs in HT29 and SW620 colorectal cancer cell lines with half-maximal inhibitory concentration (IC50) values of 2.26 and 121.18 µg ml-1, respectively. Reductions in cell viability indicated the cytotoxic potential of CeO2-NCs in HT29 cells based on inverted and florescence microscopy assessments. The mechanism of cytotoxicity confirmed by estimating possible changes in the expression levels of Bcl2, BclxL, Bax, PARP, cytochrome c, and ß-actin (control) proteins in HT29 cells. Down-regulation of Bcl2 and BclxL and up-regulation of Bax, PARP, and cytochrome c proteins suggested the significant involvement of CeO2-NCs exposure in the induction of apoptosis. Furthermore, biocompatibility assay showed minimum effect of CeO2-NCs on human red blood cells.

14.
Ecotoxicol Environ Saf ; 126: 138-146, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26748375

RESUMO

The present study was aimed to evaluate the morphological, physiological and biochemical responses of Alternanthera Bettzickiana (Regel) G. Nicholson plant subjected to different levels of cadmium (Cd) and lead (Pb) (0, 0.5, 1.0 and 2.0 mM) stress. A. bettzickiana was able to accumulate Cd and Pb in different plant parts and total uptake of both metals was higher in shoots than roots. Plant growth, biomass and photosynthetic pigments increased with increasing metal concentrations, up to 1.0 mM, in soil and then decreased with higher metal levels. The activities of superoxide dismutase (SOD), peroxidase (POD), catalase (CAT) and ascorbate peroxidase (APX) increased under lower metal levels (0.5 and 1.0 mM) while decreased at higher metal levels (2.0 mM). Leaf and root electrolyte leakage (EL), malondialdehyde (MDA) and hydrogen peroxide (H2O2) contents decreased at lower metal levels (≤1.0 mM) while increased at higher levels. The present study clearly signifies the potential of A. bettzickiana plant towards Cd and Pb tolerance and accumulation especially at lower metal levels.


Assuntos
Amaranthaceae/fisiologia , Biodegradação Ambiental , Metais Pesados/metabolismo , Poluentes do Solo/metabolismo , Amaranthaceae/crescimento & desenvolvimento , Análise de Variância , Antioxidantes/metabolismo , Cádmio/metabolismo , Estresse Oxidativo/fisiologia , Fotossíntese/fisiologia , Folhas de Planta/metabolismo , Raízes de Plantas/metabolismo , Solo/química , Poluentes do Solo/análise
15.
J Biol Inorg Chem ; 20(8): 1319-26, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26563952

RESUMO

Cobalt oxide nanoparticles (Co3O4-NPs) were synthesized using simple urea-based thermal decomposition method. Phase purity and particle size of as-synthesized nanoparticles were characterized through X-ray diffraction pattern (XRD) and transmission electron microscopy. Through XRD morphology of the Co3O4-NPs was found to be variable in size with range of 36 nm. In our present study, we explored the potential cytotoxic and antibacterial effects of Co3O4-NPs in human colorectal types of cancerous cells (HT29 and SW620) and also nine Gram-positive and Gram-negative bacteria. Co3O4-NPs showed promising anticancer activity against HT29 and SW620 cells with IC50 value of 2.26 and 394.5 µg/mL, respectively. However, no significant effect of Co3O4-NPs was observed against bacterial strains. Furthermore, a detailed study has been carried out to investigate the possible mechanism of cell death in HT29 cancer cell line through the analysis of expression level of anti-apoptotic Bcl2 and BclxL markers. Western blot analysis results suggested significant role of Co3O4-NPs exposure in cell death due to apoptosis.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos/farmacologia , Bactérias/efeitos dos fármacos , Cobalto/farmacologia , Nanopartículas Metálicas , Óxidos/farmacologia , Antibacterianos/química , Antineoplásicos/química , Bioensaio , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cobalto/química , Relação Dose-Resposta a Droga , Humanos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Transmissão , Óxidos/química
16.
Ecotoxicol Environ Saf ; 120: 310-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26099461

RESUMO

Use of organic acids for promoting heavy metals phytoextraction is gaining worldwide attention. The present study investigated the influence of citric acid (CA) in enhancing copper (Cu) uptake by Brassica napus L. seedlings. 6 Weeks old B. napus seedlings were exposed to different levels of copper (Cu, 0, 50 and 100µM) alone or with CA (2.5mM) in a nutrient medium for 40 days. Exposure to elevated Cu levels (50 and 100µM) significantly reduced the growth, biomass production, chlorophyll content, gas exchange attributes and soluble proteins of B. napus seedlings. In addition, Cu toxicity increased the production of hydrogen peroxide (H2O2), malondialdehyde (MDA) and electrolyte leakage (EL) in leaf and root tissues of B. napus. Activities of antioxidant enzymes such as guaiacol peroxidase (POD), superoxide dismutase (SOD), catalases (CAT), ascorbate peroxidase (APX) in root and shoot tissues of B. napus were increased in response to lower Cu concentration (50µM) but increased under higher Cu concentration (100µM). Addition of CA into nutrient medium significantly alleviated Cu toxicity effects on B. napus seedlings by improving photosynthetic capacity and ultimately plant growth. Increased activities of antioxidant enzymes in CA-treated plants seems to play a role in capturing of stress-induced reactive oxygen species as was evident from lower level of H2O2, MDA and EL in CA-treated plants. Increasing Cu concentration in the nutrient medium significantly increased Cu concentration in in B. napus tissues. Cu uptake was further increased by CA application. These results suggested that CA might be a useful strategy for increasing phytoextraction of Cu from contaminated soils.


Assuntos
Brassica napus/química , Ácido Cítrico/farmacologia , Cobre/farmacocinética , Ascorbato Peroxidases/metabolismo , Biodegradação Ambiental , Catalase/metabolismo , Clorofila/análise , Cobre/química , Cobre/toxicidade , Peróxido de Hidrogênio/metabolismo , Malondialdeído/metabolismo , Peroxidase/metabolismo , Fotossíntese/efeitos dos fármacos , Folhas de Planta/química , Folhas de Planta/efeitos dos fármacos , Raízes de Plantas/química , Raízes de Plantas/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Plântula/efeitos dos fármacos , Superóxido Dismutase/metabolismo
17.
J Biol Chem ; 288(43): 30892-903, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-24043631

RESUMO

Aldehyde dehydrogenase 1A1 (ALDH1A1) activity is used as a marker of breast cancer stem cells; however, little is known about the regulation of ALDH1A1 expression. Mucin 1 (MUC1) is a heterodimeric protein that is aberrantly overexpressed in most human breast cancers. In studies of breast cancer cells stably silenced for MUC1 or overexpressing the oncogenic MUC1-C subunit, we demonstrate that MUC1-C is sufficient for induction of MEK → ERK signaling and that treatment with a MUC1-C inhibitor suppresses ERK activation. In turn, MUC1-C induces ERK-mediated phosphorylation and activation of the CCAAT/enhancer-binding protein ß (C/EBPß) transcription factor. The results further show that MUC1-C and C/EBPß form a complex on the ALDH1A1 gene promoter and activate ALDH1A1 gene transcription. MUC1-C-induced up-regulation of ALDH1A1 expression is associated with increases in ALDH activity and is detectable in stem-like cells when expanded as mammospheres. These findings demonstrate that MUC1-C (i) activates a previously unrecognized ERK→C/EBPß→ALDH1A1 pathway, and (ii) promotes the induction of ALDH activity in breast cancer cells.


Assuntos
Aldeído Desidrogenase/biossíntese , Neoplasias da Mama/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Sistema de Sinalização das MAP Quinases , Mucina-1/metabolismo , Proteínas de Neoplasias/metabolismo , Aldeído Desidrogenase/genética , Família Aldeído Desidrogenase 1 , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteína beta Intensificadora de Ligação a CCAAT/genética , Linhagem Celular Tumoral , Feminino , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Mucina-1/genética , Proteínas de Neoplasias/genética , Subunidades Proteicas , Retinal Desidrogenase , Transcrição Gênica/genética
18.
Nat Cell Biol ; 9(12): 1419-27, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18037881

RESUMO

Nuclear factor-kappaB (NF-kappaB) is constitutively activated in diverse human malignancies by mechanisms that are not understood. The MUC1 oncoprotein is aberrantly overexpressed by most human carcinomas and, similarly to NF-kappaB, blocks apoptosis and induces transformation. This study demonstrates that overexpression of MUC1 in human carcinoma cells is associated with constitutive activation of NF-kappaB p65. We show that MUC1 interacts with the high-molecular-weight IkappaB kinase (IKK) complex in vivo and that the MUC1 cytoplasmic domain binds directly to IKKbeta and IKKgamma. Interaction of MUC1 with both IKKbeta and IKKgamma is necessary for IKKbeta activation, resulting in phosphorylation and degradation of IkappaBalpha. Studies in non-malignant epithelial cells show that MUC1 is recruited to the TNF-R1 complex and interacts with IKKbeta-IKKgamma in response to TNFalpha stimulation. TNFalpha-induced recruitment of MUC1 is dependent on TRADD and TRAF2, but not the death-domain kinase RIP1. In addition, MUC1-mediated activation of IKKbeta is dependent on TAK1 and TAB2. These findings indicate that MUC1 is important for physiological activation of IKKbeta and that overexpression of MUC1, as found in human cancers, confers sustained induction of the IKKbeta-NF-kappaB p65 pathway.


Assuntos
Quinase I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Mucina-1/fisiologia , Fator de Transcrição RelA/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Ativação Enzimática , Humanos , MAP Quinase Quinase Quinases/metabolismo , Fosforilação , Ligação Proteica , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
19.
J Biol Chem ; 287(25): 20866-75, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22544745

RESUMO

The pro-apoptotic BAX protein contains a BH3 domain that is necessary for its dimerization and for activation of the intrinsic apoptotic pathway. The MUC1 (mucin 1) heterodimeric protein is overexpressed in diverse human carcinomas and blocks apoptosis in the response to stress. In this study, we demonstrate that the oncogenic MUC1-C subunit associates with BAX in human cancer cells. MUC1-C·BAX complexes are detectable in the cytoplasm and mitochondria and are induced by genotoxic and oxidative stress. The association between MUC1-C and BAX is supported by the demonstration that the MUC1-C cytoplasmic domain is sufficient for the interaction with BAX. The results further show that the MUC1-C cytoplasmic domain CQC motif binds directly to the BAX BH3 domain at Cys-62. Consistent with binding to the BAX BH3 domain, MUC1-C blocked BAX dimerization in response to (i) truncated BID in vitro and (ii) treatment of cancer cells with DNA-damaging agents. In concert with these results, MUC1-C attenuated localization of BAX to mitochondria and the release of cytochrome c. These findings indicate that the MUC1-C oncoprotein binds directly to the BAX BH3 domain and thereby blocks BAX function in activating the mitochondrial death pathway.


Assuntos
Mucina-1/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína X Associada a bcl-2/metabolismo , Morte Celular/genética , Linhagem Celular Tumoral , Citocromos c/genética , Citocromos c/metabolismo , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mucina-1/genética , Complexos Multiproteicos/genética , Proteínas de Neoplasias/genética , Ligação Proteica , Multimerização Proteica/genética , Estrutura Terciária de Proteína , Proteína X Associada a bcl-2/genética
20.
J Biol Chem ; 287(13): 10703-10713, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22318732

RESUMO

MUC1 is a heterodimeric glycoprotein that is overexpressed in breast cancers. The present studies demonstrate that the oncogenic MUC1 C-terminal subunit (MUC1-C) associates with the TCF7L2 transcription factor. The MUC1-C cytoplasmic domain (MUC1-CD) binds directly to the TCF7L2 C-terminal region. MUC1-C blocks the interaction between TCF7L2 and the C-terminal-binding protein (CtBP), a suppressor of TCF7L2-mediated transcription. TCF7L2 and MUC1-C form a complex on the cyclin D1 gene promoter and MUC1-C promotes TCF7L2-mediated transcription by the recruitment of ß-catenin and p300. Silencing MUC1-C in human breast cancer cells down-regulated activation of the cyclin D1 promoter and decreased cyclin D1 expression. In addition, a MUC1-C inhibitor blocked the interaction with TCF7L2 and suppressed cyclin D1 levels. These findings indicate that the MUC1-C oncoprotein contributes to TCF7L2 activation and thereby promotes cyclin D1 expression in breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Ciclina D1/biossíntese , Regulação Neoplásica da Expressão Gênica , Mucina-1/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Transcrição Gênica , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ciclina D1/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Mucina-1/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , beta Catenina/genética , beta Catenina/metabolismo , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA