Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
Hum Exp Toxicol ; 43: 9603271241266106, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39291655

RESUMO

OBJECTIVE: This study aimed to explore the expression and biological functions of SIRT3 in colorectal cancer cells (HCT-116), the impacts of sulforaphane on the ferroptosis of HCT-116 cells and the involvement of the SIRT3/AMPK/mTOR axis in those effects. METHODS: SIRT3-overexpressing (OE) and SIRT3-knockout (KO) cell lines were treated with different concentrations of sulforaphane, RSL-3, and IKE. Cell viability, intracellular ROS, MDA, iron levels, as well as mRNA and protein expressions of target genes were measured. RESULTS: SIRT3 expression in HCT-116 cells was increased by ferroptosis inducers and decreased by ferroptosis inhibitors. SIRT3 overexpression reduced cell viability and increased intracellular levels of ROS, MDA, and iron, whereas SIRT3 knockdown achieved the opposite effects. SIRT3 overexpression suppressed SLC7A11 expression and promoted the activation of AMPK/mTOR pathway. Restoration of SLC7A11 expression blocked the effects of SIRT3 on ferroptosis induction and cell viability inhibition. SIRT3 effects on cell viability and ferroptosis were antagonized by inhibitors of AMPK or mTOR. Moreover, sulforaphane triggered the ferroptosis of HCT-116 cells by activating the SIRT3/AMPK/mTOR axis. CONCLUSIONS: SIRT3 triggered SLC7A11-mediated ferroptosis in HCT-116 cells, reducing cell viability by activating the AMPK/mTOR pathway, and sulforaphane targets it to inhibit colorectal cancer.


Assuntos
Proteínas Quinases Ativadas por AMP , Neoplasias Colorretais , Ferroptose , Isotiocianatos , Transdução de Sinais , Sirtuína 3 , Sulfóxidos , Serina-Treonina Quinases TOR , Humanos , Isotiocianatos/farmacologia , Sirtuína 3/metabolismo , Sirtuína 3/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Ferroptose/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células HCT116 , Anticarcinógenos/farmacologia , Sobrevivência Celular/efeitos dos fármacos
2.
Drug Dev Res ; 85(5): e22231, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38956926

RESUMO

The close association between inflammation and cancer inspired the synthesis of a series of 1,3,4-oxadiazole derivatives (compounds H4-A-F) of 6-methoxynaphtalene. The chemical structures of the new compounds were validated utilizing Fourier-transform infrared, proton nuclear magnetic resonance, and carbon-13 nuclear magnetic resonance spectroscopic techniques and CHN analysis. Computer-aided drug design methods were used to predict the compounds biological target, ADMET properties, toxicity, and to evaluate the molecular similarities between the design compounds and erlotinib, a standard epidermal growth factor receptor (EGFR) inhibitor. The antiproliferative effects of the new compounds were evaluated by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay, cell cycle analysis, apoptosis detection by microscopy, quantitative reverse transcription-polymerase chain reaction, and immunoblotting, and EGFR enzyme inhibition assay. In silico analysis of the new oxadiazole derivatives indicated that these compounds target EGFR, and that compounds H4-A, H4-B, H4-C, and H4-E show similar molecular properties to erlotinib. Additionally, the results indicated that none of the synthesized compounds are carcinogenic, and that compounds H4-A, H4-C, and H4-F are nontoxic. Compound H4-A showed the best-fit score against EGFR pharmacophore model, however, the in vitro studies indicated that compound H4-C was the most cytotoxic. Compound H4-C caused cytotoxicity in HCT-116 colorectal cancer cells by inducing both apoptosis and necrosis. Furthermore, compounds H4-D, H4-C, and H4-B had potent inhibitory effect on EGFR tyrosine kinase that was comparable to erlotinib. The findings of this inquiry offer a basis for further investigation into the differences between the synthesized compounds and erlotinib. However, additional testing will be needed to assess all of these differences and to identify the most promising compound for further research.


Assuntos
Antineoplásicos , Receptores ErbB , Simulação de Acoplamento Molecular , Naproxeno , Oxidiazóis , Receptores ErbB/antagonistas & inibidores , Humanos , Oxidiazóis/farmacologia , Oxidiazóis/química , Oxidiazóis/síntese química , Naproxeno/farmacologia , Naproxeno/análogos & derivados , Naproxeno/química , Naproxeno/síntese química , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Linhagem Celular Tumoral , Apoptose/efeitos dos fármacos , Cloridrato de Erlotinib/farmacologia , Cloridrato de Erlotinib/química , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/síntese química , Proliferação de Células/efeitos dos fármacos
3.
Chem Biol Interact ; 400: 111163, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39053794

RESUMO

The ineffectiveness of cisplatin therapy in treating colorectal cancer (CRC) is attributed to an increase of resistance. It's necessary to investigate adjunctive agents capable of enhancing drug efficacy. Previous studies have shown that ropivacaine inhibit the growth of various cancer cells, but its impact on cisplatin resistance in tumors is not well understood. This study was to illustrate the impact and mechanism of ropivacaine enhanced cisplatin-sensitivity of CRC. Cisplatin alone treatment resulted in the elevation of reactive oxygen species (ROS) and intracellular Fe2+ levels, as well as a reduction in mitochondrial membrane potential (MMP) in cisplatin-sensitive LOVO cells, while these effects were mitigated in the cisplatin-resistant LOVO/DDP cells. The co-administration of ropivacaine with cisplatin inhibited cell viability and cell migration, decreased MMP, and promoted ROS accumulation and apoptosis in both LOVO cells and LOVO/DDP cells. And they upregulated the levels of ferroptosis makers and downregulated the expression of anti-ferroptosis proteins. However, this effect was reversed by ferroptosis inhibitor ferrostatin-1 or liproxstatin-1. Furthermore, we o demonstrated that the co-administration of ropivacaine and cisplatin resulted in a decrease in SIRT1 expression, and SIRT1 knockdown in LOVO/DDP cells enhanced the ferroptosis and the anti-tumor properties of ropivacaine, while also inhibiting the activation of the Nrf2/Keap1 pathway. The above results suggested that ropivacaine increased the sensitivity of CRC cells to cisplatin by promoting ferroptosis through the inhibition of SIRT1 expression, which proposes a therapeutic approach for overcoming cisplatin resistance in CRC.


Assuntos
Antineoplásicos , Cisplatino , Neoplasias Colorretais , Ferroptose , Fator 2 Relacionado a NF-E2 , Espécies Reativas de Oxigênio , Ropivacaina , Transdução de Sinais , Sirtuína 1 , Humanos , Ropivacaina/farmacologia , Ferroptose/efeitos dos fármacos , Cisplatino/farmacologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Transdução de Sinais/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Sirtuína 1/metabolismo , Sirtuína 1/genética , Linhagem Celular Tumoral , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Apoptose/efeitos dos fármacos
4.
Comput Biol Med ; 178: 108744, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38889631

RESUMO

Cancer alters the structural integrity and morphology of cells. Consequently, the cell function is overshadowed. In this study, the micropipette aspiration process is computationally modeled to predict the mechanical behavior of the colorectal cancer cells. The intended cancer cells are modeled as an incompressible Neo-Hookean visco-hyperelastic material. Also, the micropipette is assumed to be rigid with no deformation. The proposed model is validated with an in-vitro study. To capture the equilibrium and time-dependent behaviors of cells, ramp, and creep tests are respectively performed using the finite element method. Through the simulations, the effects of the micropipette geometry and the aspiration pressure on the colorectal cancer cell lines are investigated. Our findings indicate that, as the inner radius of the micropipette increases, despite the increase in deformation rate and aspirated length, the time to reach the equilibrium state increases. Nevertheless, it is obvious that increasing the tip curvature radius has a small effect on the change of the aspirated length. But, due to the decrease in the stress concentration, it drastically reduces the equilibrium time and increases the deformation rate significantly. Interestingly, our results demonstrate that increasing the aspiration pressure somehow causes the cell stiffening, thereby reducing the upward trend of deformation rate, equilibrium time, and aspirated length. Our findings provide valuable insights for researchers in cell therapy and cancer treatment and can aid in developing more precise microfluidic.


Assuntos
Neoplasias Colorretais , Modelos Biológicos , Humanos , Neoplasias Colorretais/patologia , Linhagem Celular Tumoral , Simulação por Computador , Fenômenos Biomecânicos/fisiologia , Análise de Elementos Finitos , Estresse Mecânico
5.
Cell Mol Biol Lett ; 29(1): 80, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38811901

RESUMO

BACKGROUND: Sodium-glucose transporter 2 (SGLT2) inhibitors (iSGLT2) are approved medications for type 2 diabetes. Recent studies indicate that iSGLT2 inhibit the growth of some cancer cells. However, the mechanism(s) remains to be fully elucidated. METHODS: The SGLT2 levels were determined in normal colon CCD 841 CoN and, HCT 116, HT-29, SW480 and LoVo colorectal cancer (CRC) cell lines by quantitative real-time PCR and western blot. The effect of iSGLT2 canagliflozin on cell proliferation was examined using CCK-8, as its role on CRC cells metabolism and tumorigenesis has been evaluated by XF HS Seahorse Bioanalyzer and flow cytometric analyses. Transient gene silencing experiments and analysis of protein-protein interaction network were conducted to evaluate the SGLT2 molecular targets in CRC cells. RESULTS: Data showed that the treatment with iSGLT2 (50 µM) for 72 h induced cell cycle arrest (p < 0.001), impaired glucose and energetic metabolism (p < 0.001), promoted apoptotic cell death and ER stress flowing into autophagy (p < 0.001) in HCT 116 and HT-29 cells. These cellular events were accompanied by sirtuin 3 (SIRT3) upregulation (p < 0.01), as also supported by SIRT3 transient silencing experiments resulting in the attenuation of the effects of iSGLT2 on the cellular metabolic/energetic alterations and the induction of programmed cell death. The identification and validation of dipeptidyl peptidase 4 (DPP4) as potential common target of SGLT2 and SIRT3 were also assessed. CONCLUSIONS: These results deepened knowledge on the iSGLT2 contribution in limiting CRC tumorigenesis unveiling the SGLT2/SIRT3 axis in the cytotoxic mechanisms.


Assuntos
Apoptose , Proliferação de Células , Neoplasias Colorretais , Estresse do Retículo Endoplasmático , Mitocôndrias , Inibidores do Transportador 2 de Sódio-Glicose , Transportador 2 de Glucose-Sódio , Humanos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transportador 2 de Glucose-Sódio/metabolismo , Transportador 2 de Glucose-Sódio/genética , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Canagliflozina/farmacologia , Células HT29 , Células HCT116 , Sirtuína 3/metabolismo , Sirtuína 3/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Glucose/metabolismo
6.
Front Bioeng Biotechnol ; 12: 1385730, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38803844

RESUMO

With cancer as one of the leading causes of death worldwide, there is a need for the development of accurate, cost-effective, easy-to-use, and fast drug-testing assays. While the NCI 60 cell-line screening as the gold standard is based on a colorimetric assay, monitoring cells electrically constitutes a label-free and non-invasive tool to assess the cytotoxic effects of a chemotherapeutic treatment on cancer cells. For decades, impedance-based cellular assays extensively investigated various cell characteristics affected by drug treatment but lack spatiotemporal resolution. With progress in microelectrode fabrication, high-density Complementary Metal Oxide Semiconductor (CMOS)-based microelectrode arrays (MEAs) with subcellular resolution and time-continuous recording capability emerged as a potent alternative. In this article, we present a new cell adhesion noise (CAN)-based electrical imaging technique to expand CMOS MEA cell-biology applications: CAN spectroscopy enables drug screening quantification with single-cell spatial resolution. The chemotherapeutic agent 5-Fluorouracil exerts a cytotoxic effect on colorectal cancer (CRC) cells hampering cell proliferation and lowering cell viability. For proof-of-concept, we found sufficient accuracy and reproducibility for CAN spectroscopy compared to a commercially available standard colorimetric biological assay. This label-free, non-invasive, and fast electrical imaging technique complements standardized cancer screening methods with significant advances over established impedance-based approaches.

7.
J Enzyme Inhib Med Chem ; 39(1): 2337191, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38634597

RESUMO

Colon cancer remains a clinical challenge in industrialised countries. Its treatment with 5-Flurouracil (5-FU) develops many side effects and resistance. Thus, several strategies have been undertaken so far, including the use of drug cocktails and polypharmacology. Heme oxygenase-1 (HO-1) is an emerging molecular target in the treatment of various cancers. We recently demonstrated that a combination of HO-1 inhibitors with 5-FU and the corresponding hybrids SI1/17, SI1/20, and SI1/22, possessed anticancer activity against prostate and lung cancer cells. In this work, we evaluated these hybrids in a model of colon cancer and found that SI1/22 and the respective combo have greater potency than 5-FU. Particularly, compounds inhibit HO-1 activity in cell lysates, increase ROS and the expression of HO-1, SOD, and Nrf2. Moreover, we observed a decrease of pro-caspase and an increase in cleaved PARP-1 and p62, suggesting apoptotic and autophagic cell death and potential application of these drugs as anticancer agents.


Assuntos
Antineoplásicos , Neoplasias do Colo , Fluoruracila , Humanos , Masculino , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Fluoruracila/farmacologia , Heme Oxigenase-1/antagonistas & inibidores
8.
Cell Oncol (Dordr) ; 47(4): 1459-1474, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38592610

RESUMO

PURPOSE: α2-adrenoceptor agonist dexmedetomidine (DEX) has been reported to promote tumorigenesis. Stem-cell protein Piwil2 is associated with cancer progression. Whether Piwil2 plays a role in tumor-promoting effects of DEX is unknown. METHODS: We examined the expression of Piwil2 in human colorectal cancer (CRC) cell lines with/without DEX treatment. We also studied the roles of Piwil2 in proliferation, invasion, migration, as well as expressions of epithelial-mesenchymal transition (EMT)-related proteins in DEX-treated in vitro and in vivo CRC models. And the experiments with genetic and pharmacological treatments were conducted to investigate the underlying molecular mechanism. RESULTS: RNA-sequencing (RNA-seq) analysis found Piwil2 is one of most upregulated genes upon DEX treatment in CRC cells. Furthermore, Piwil2 protein levels significantly increased in DEX-treated CRC cancer cells, which promoted proliferation, invasion, and migration in both CRC cell lines and human tumor xenografts model. Mechanistically, DEX increased nuclear factor E2-related factor 2 (Nrf2) expression, which enhanced Piwil2 transcription via binding to its promoter. Furthermore, in vitro experiments with Piwil2 knockdown or Siah2 inhibition indicated that DEX promoted EMT process and tumorigenesis through Siah2/PHD3/HIF1α pathway. The experiments with another α2-adrenoceptor agonist Brimonidine and antagonists yohimbine and atipamezole also suggested the role of Piwil2 signaling in tumor-promoting effects via an α2 adrenoceptor-dependent manner. CONCLUSION: DEX promotes CRC progression may via activating α2 adrenoceptor-dependent Nrf2/Piwil2/Siah2 pathway and thus EMT process. Our work provides a novel insight into the mechanism underlying tumor-promoting effects of α2-adrenoceptor agonists.


Assuntos
Proteínas Argonautas , Movimento Celular , Proliferação de Células , Neoplasias Colorretais , Dexmedetomidina , Progressão da Doença , Transição Epitelial-Mesenquimal , Camundongos Nus , Transdução de Sinais , Humanos , Dexmedetomidina/farmacologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/genética , Animais , Transdução de Sinais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Movimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas Argonautas/metabolismo , Camundongos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Invasividade Neoplásica , Camundongos Endogâmicos BALB C , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Pharm Pharmacol ; 76(6): 691-700, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38459835

RESUMO

OBJECTIVES: Improving response rates in colorectal cancer (CRC) is an urgent clinical need. This study aimed to explore the synergistic action of Lebanese rosemary essential oil (REO) and 5-fluorouracil (5-FU) in HCT116 CRC cells. METHODS: We tested the cell viability of monotherapy and combination therapy. The combination index was calculated using CompuSyn software to evaluate drug-drug interactions and the level of synergistic cytotoxicity. We also evaluated cell migration and cytopathology. Furthermore, cell apoptosis-related proteins (i.e. Bax and Bcl-2) were measured by Western blot analysis. KEY FINDINGS: The REO/5-FU combination synergistically reduced cell viability, effectively decreased cell migration, and increased the Bax/Bcl-2 ratio in HCT116 cells. This triggered a proapoptotic morphology and initiated an apoptotic cascade in HCT116 cells, as indicated by a higher Bax/Bcl-2 ratio. CONCLUSIONS: Our results provide evidence of the REO/5-FU combination as a better approach to improve 5-FU anticancer efficacy and allow the use of lower 5-FU doses due to the adjuvant effect of REO.


Assuntos
Apoptose , Sobrevivência Celular , Neoplasias Colorretais , Sinergismo Farmacológico , Fluoruracila , Óleos Voláteis , Proteínas Proto-Oncogênicas c-bcl-2 , Proteína X Associada a bcl-2 , Humanos , Fluoruracila/farmacologia , Óleos Voláteis/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Células HCT116 , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Movimento Celular/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Rosmarinus/química
10.
Artigo em Inglês | MEDLINE | ID: mdl-38555594

RESUMO

5-Fluorouracil (5-FU) is a commonly used anticancer drug for colorectal cancer (CRC). Therefore, it is crucial to elucidate the mechanisms that contribute to 5-FU resistance. We established an acquired 5-FU resistant cell line, HCT116RF10, derived from CRC cells and investigated its energy metabolism as well as the underlying mechanism of 5-FU resistance. We examined the sensitivity to 5-FU and the formation of tumor spheres in parental HCT116 cells and 5-FU-resistant HCT116RF10 cells under 3D culture conditions at high-glucose (HG 25 mM) and low-glucose (LG 5.5 mM) concentrations. These results suggested that the tumor spheres of parental HCT116 cells displayed higher sensitivity to 5-FU under LG conditions than under HG conditions. HCT116RF10 tumor spheres exhibited comparable sensitivity to 5-FU under HG and LG conditions. Furthermore, under HG conditions, there was a marked decrease in extracellular lactate in the HCT116RF10 tumor sphere compared to that in the LG tumor sphere. Similarly, HCT116 tumor spheres showed decreased extracellular lactate levels under LG conditions compared to those grown under HG conditions. Moreover, the evidence reveals that the tumor spheres of HCT116RF10 and HCT116 cells exhibit disparate dependencies on energy metabolism, glycolysis, and mitochondrial respiration under both HG and LG conditions. These results have important clinical implications for overcoming 5-FU resistance and enhancing antitumor treatment strategies.

11.
Chin J Integr Med ; 30(1): 25-33, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37750986

RESUMO

OBJECTIVE: To determine whether monotropein has an anticancer effect and explore its potential mechanisms against colorectal cancer (CRC) through network pharmacology and molecular docking combined with experimental verification. METHODS: Network pharmacology and molecular docking were used to predict potential targets of monotropein against CRC. Cell counting kit assay, plate monoclonal assay and microscopic observation were used to investigate the antiproliferative effects of monotropein on CRC cells HCT116, HT29 and LoVo. Flow cytometry and scratch assay were used to analyze apoptosis and cell cycle, as well as cell migration, respectively in HCT116, HT29, and LoVo cells. Western blotting was used to detect the expression of proteins related to apoptosis, cell cycle, and cell migration, and the expression of proteins key to the Akt pathway. RESULTS: The Gene Ontology and Reactome enrichment analyses indicated that the anticancer potential of monotropein against CRC might be involved in multiple cancer-related signaling pathways. Among these pathways, RAC-beta serine/threonine-protein kinase (Akt1, Akt2), cyclin-dependent kinase 6 (CDK6), matrix metalloproteinase-9 (MMP9), epidermal growth factor receptor (EGFR), cell division control protein 42 homolog (CDC42) were shown as the potential anticancer targets of monotropein against CRC. Molecular docking suggested that monotropein may interact with the 6 targets (Akt1, Akt2, CDK6, MMP9, EGFR, CDC42). Subsequently, cell activity of HCT116, HT29 and LoVo cell lines were significantly suppressed by monotropein (P<0.05). Furthermore, our research revealed that monotropein induced cell apoptosis by inhibiting Bcl-2 and increasing Bax, induced G1-S cycle arrest in colorectal cancer by decreasing the expressions of CyclinD1, CDK4 and CDK6, inhibited cell migration by suppressing the expressions of CDC42 and MMP9 (P<0.05), and might play an anticancer role through Akt signaling pathway. CONCLUSION: Monotropein exerts its antitumor effects primarily by arresting the cell cycle, causing cell apoptosis, and inhibiting cell migration. This indicates a high potential for developing novel medication for treating CRC.


Assuntos
Neoplasias Colorretais , Proteínas Proto-Oncogênicas c-akt , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proliferação de Células , Metaloproteinase 9 da Matriz , Simulação de Acoplamento Molecular , Ciclo Celular , Receptores ErbB , Apoptose , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Linhagem Celular Tumoral
12.
J Chemother ; : 1-18, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38054850

RESUMO

This study investigated the potential of a newly synthesized histone deacetylase (HDAC) inhibitor, MHY446, in inducing cell death in HCT116 colorectal cancer cells and compared its activity with that of suberoylanilide hydroxamic acid (SAHA), a well-known HDAC inhibitor. The results showed that MHY446 increased the acetylation of histones H3 and H4 and decreased the expression and activity of HDAC proteins in HCT116 cells. Additionally, MHY446 was confirmed to bind more strongly to HDAC1 than HDAC2 and inhibit its activity. In vivo experiments using nude mice revealed that MHY446 was as effective as SAHA in inhibiting HCT116 cell-grafted tumor growth. This study also evaluated the biological effects of MHY446 on cell survival and death pathways. The reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) confirmed that ROS play a role in MHY446-induced cell death by reducing poly(ADP-ribose) polymerase cleavage. MHY446 also induced cell death via endoplasmic reticulum (ER) stress by increasing the expression of ER stress-related proteins. NAC treatment decreased the expression of ER stress-related proteins, indicating that ROS mediate ER stress as an upstream signaling pathway and induce cell death. While MHY446 did not exhibit superior HDAC inhibition efficacy compared to SAHA, it is anticipated to provide innovative insights into the future development of therapeutic agents for human CRC by offering novel chemical structure-activity relationship-related information.

13.
J Adv Pharm Technol Res ; 14(4): 317-324, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38107450

RESUMO

This study aimed to evaluate the efficacy of Chemoprevention Curcumin Analog-1.1 (CCA-1.1) and Pentagamavunone-1 (PGV-1) in vivo and in vitro in colorectal cancer model. CCA-1.1 or PGV-1 was administered orally to 1,2-dimethylhydrazine (DMH)-induced rats for 16 weeks. The cytotoxicity of both compounds was tested on Caco-2, CT26, and NIH/3T3 cells using the MTT method. The cell cycle, apoptosis, and reactive oxygen species (ROS) levels were analyzed through flow cytometry. X-gal staining was used to examine the compound's effect on senescence. Oral co-administration of CCA-1.1 or PGV-1 significantly suppressed the carcinogenic characteristics and symptoms of premalignant colon cancer relative to DMH-only and untreated groups. CCA-1.1 and PGV-1 administration did not affect the blood profile. CCA-1.1 and PGV-1 demonstrated great cytotoxicity on Caco-2 and CT26 cells, with 50% inhibition concentration (IC50) values of 4.3 ± 0.2 and 3.1 ± 0.1 µM for CCA-1.1 and 11.2 ± 1.1 and 4.8 ± 0.1 µM for PGV-1, respectively, while not toxic against fibroblast cells. Both compounds instigated G2/M arrest and efficiently induced cell senescence and apoptosis. Moreover, these analogs selectively elevated oxidative stress in colon cancer cells without inducing noticeable changes in fibroblasts. In conclusion, PGV-1 and CCA-1.1 suppressed colorectal tumor formation and induced mitotic arrest.

14.
Curr Pharm Des ; 29(36): 2867-2876, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37957863

RESUMO

BACKGROUND: Curcuminoids, including curcumin, desmethoxycurcumin, and bisdesmethoxycurcumin, are natural polyphenolic compounds that exhibit various biological properties, such as antioxidant, anti-inflammatory, and anticancer activities. Dysregulation of the interleukin (IL)-6-mediated Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signaling pathway is closely associated with the development of colorectal cancer (CRC). METHODS: Here, we have evaluated the modulation of the IL-6/JAK/STAT3 pathway of curcumin, desmethoxycurcumin, and bisdesmethoxycurcumin in LoVo and HT-29 colorectal cancer cells with a single molecular array (Simoa), western blot analysis, real-time polymerase chain reaction (PCR), and pathway analysis system. RESULTS: The study showed that curcuminoids suppressed the amount of IL-6 in LoVo and HT-29 colorectal cancer cells. Meanwhile, curcuminoids inhibited the expression of inflammation regulator-related microRNA (miRNA). We also found that the expression of total STAT3 was downregulated by curcuminoids. Moreover, the pathway analysis system showed that curcuminoids inactivated the JAK/STAT3 signaling pathway. Taken together, we demonstrated that the anti-cancer activities of curcuminoids against colorectal cancer are due to the modulation of the IL-6/JAK/STAT3 cascade. CONCLUSION: Curcuminoids could be a promising anti-cancer agent for the treatment of human colorectal cancer.


Assuntos
Neoplasias Colorretais , Curcumina , Humanos , Janus Quinases , Curcumina/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Interleucina-6/metabolismo , Diarileptanoides , Transdução de Sinais , Neoplasias Colorretais/metabolismo
15.
J Mol Histol ; 54(6): 621-631, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37776473

RESUMO

Endoplasmic reticulum oxidoreductin 1α (ERO1α) is an oxidase that exists in the endoplasmic reticulum and plays an important role in regulating oxidized protein folding and tumor malignant progression. However, the specific role and mechanism of ERO1α in the progression of colorectal cancer (CRC) have not yet been fully elucidated. In this study, 280 specimens of CRC tissues and adjacent noncancerous tissues were collected to detect the expression of ERO1α and analyze the clinical significance. ERO1α was stably knocked-down in RKO and HT29 CRC cells to investigate its function and mechanism in vitro and in vivo. We found that ERO1α was remarkably upregulated in CRC tissues and high ERO1α expression is associated with N stage and poor prognosis of CRC patients. ERO1α knockdown in CRC cells significantly inhibited the proliferation and induced apoptosis while inactivating the PI3K/AKT pathway. Rescue assays revealed that AKT activator 740Y-P could reverse the effects on proliferation and apoptosis of ERO1α knockdown in CRC cells. In vivo tumorigenicity assay also confirmed that ERO1α knockdown suppressed tumor growth. Taken together, our findings demonstrated ERO1α promotes the proliferation and inhibits apoptosis of CRC cells by regulating the PI3K/AKT pathway. High expression of ERO1α is associated with poor prognosis in CRC patients, and ERO1α could be a potential therapeutic target for CRC.


Assuntos
Neoplasias Colorretais , Proteínas Proto-Oncogênicas c-akt , Humanos , Apoptose , Proliferação de Células , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
16.
Chem Biol Drug Des ; 102(5): 1202-1212, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37599210

RESUMO

The present study was to investigate the underlying mechanism of the antitumor effect of curcumin in colorectal cancer cells, focusing on the M2 polarization of tumor-associated macrophages (TAMs). The effect of curcumin on the malignant behavior of colorectal cancer cells was investigated by WST assay for cell growth, and Transwell assay for cell migration/invasion. THP-1 cells were differentiated into macrophages and coculture with colorectal cancer cells to study the influence of curcumin on M2 polarization, presenting as the levels of ARG1 mRNA, IL-10, and CD163-positive cells. GEO database was searched for the shared altered gene of curcumin in colorectal cells and human monocytes. Molecular docking was used to visualize the binding between curcumin and MACC1. Curcumin restricted the proliferation, apoptosis, and migration/invasion of HCT 116 and SW620 cells. Curcumin attenuated levels of the M2 macrophage markers, CD163 + cells, IL-10 secretion, and ARG1 mRNA. MACC1 was a target of curcumin in colorectal cancer cells, relating to macrophage. Rescue experiments showed that MACC1 overexpression can reverse the antitumor effect of curcumin in colorectal cancer cells and M2 polarization of TAMs. Curcumin's antiproliferative and anti-migratory effects in colorectal cancer cells may be mediated by MACC1 and inhibition of M2 polarization of TAMs.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Curcumina , Humanos , Interleucina-10/genética , Interleucina-10/farmacologia , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/patologia , Curcumina/farmacologia , Simulação de Acoplamento Molecular , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/tratamento farmacológico , RNA Mensageiro , Microambiente Tumoral , Transativadores/farmacologia
17.
World J Oncol ; 14(4): 266-276, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37560340

RESUMO

Background: Pogonatherum paniceum (P. paniceum) (Lam.) Hack. plays an important role in detoxification. However, its anticancer activity has not yet been elucidated. The aim of our study was to examine the suppressive proliferation, anti-migration and mutagenic/antimutagenic properties of P. paniceum. Moreover, we set out to determine the cellular mechanism underlying its antiproliferation. Methods: To investigate P. paniceum's anticancer ability, HCT116 and HT29 cell lines were treated with a water extract containing P. paniceum, and then the cell viability was examined using the trypan blue exclusion method which were compared to HEK293 (non-cancerous cells). The anticancer effects were investigated by MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) and colony formation assay. Apoptosis induction, cell cycle distribution, and migration abilities were assessed by cell death detection enzyme-linked immunoassay (ELISA), flow cytometry, and wound healing assay. Finally, the mutagenicity and antimutagenicity were evaluated using the micronucleus assay. Results: Treatment with P. paniceum caused a loss of cell viability in HCT116 and HT29 cells (not found in HEK293), which had an IC50 (half-maximal inhibitory concentration) of 1,156.2 and 1,207.0 µg/mL, respectively. We found that P. paniceum significantly inhibited the proliferative function of HCT116 and HT29 cells. To find the mechanism that exerts a suppressive proliferation effect on P. paniceum, we determined the DNA fragmentation and cell cycle distribution. We also found that P. paniceum treatment increased apoptosis and arrested of the cell cycle at G0/G1 remarkably when compared with the control group. Moreover, P. paniceum could decrease the migration of HCT116 and HT29 cancer cells. Finally, the treatment of P. paniceum did not induce micronucleus formation but did decrease the micronucleus frequency against mutagen-mitomycin C. Conclusions: P. paniceum did not possess any toxicity (cytotoxic and mutagenic) but has the potential for anticancer activity against human colorectal cells by increasing apoptosis, which leads to the suppression of cell proliferation. P. paniceum also inhibits cell migration and exerts antimutagenicity, thereby suggesting that P. paniceum might be useful for colorectal cancer treatment.

18.
Anticancer Agents Med Chem ; 23(17): 1974-1981, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37565553

RESUMO

INTRODUCTION: The marine environment is a rich source of biodiversity, with several of its inhabitants producing unique and physiologically active substances. The use of marine bacterial-derived chemicals over traditional pharmaceuticals is gaining traction due to their larger variety of targets and modes of action. To circumvent the drawbacks of current therapy options, researchers have looked to marine microbes for novel and effective anti-cancer compounds. In this study, we examine one of India's least-examined coastal areas in search of novel bacterial sources of anti-cancer chemicals. METHOD: Soil sediments from the Indian south coast region were collected and microbes were isolated using standard methods. The microorganisms were identified using 16s rRNA sequencing, and cytotoxic extracts were further examined using GC-MS. MTT, clonogenic, and spheroid tests assessed the extract's cytotoxicity and anti-tumor efficacy. RESULTS: Our results indicated that the bacterial isolates with potent cytotoxic activity were Bacillus drentensis and Bacillus haikouensis and had 10 and 12 potent anti-cancer and other bioactive compounds. The extracts had an IC50 of 30.08 and 109.4 µg/ml in the HCT116 cell line, respectively, and strongly inhibited colony formation. The cell cycle analysis indicated that the extract induced cell death as indicated by the subG0 peak. We also showed that these methanolic extracts induced toxicity in a 3D spheroid model indicating a strong anti-tumor activity. Furthermore, we performed molecular docking for the compounds present in the extracts to VEGFR and nucleolin and found that ergostane had favorable binding energy only to VEGFR. CONCLUSION: The results indicate that the ME of B. drentensis and B. haikouensis contains potent anti-cancer compounds to exhibit cytotoxic and anti-tumor activity in colorectal cancer cells.

19.
Antioxidants (Basel) ; 12(8)2023 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-37627542

RESUMO

Quercetin (Qc) inhibits cell proliferation and induces apoptosis in a variety of cancer cells. The molecular mechanism of action has not been fully elucidated; however, interplay with some miRNAs has been reported, specifically with miR-27a, an onco-miRNA overexpressed in several malignancies. Here, we show that Qc reduces cell viability and induces apoptosis in HCT116 and HT-29 colon cancer cells, by upregulating negative modulators of proliferation pathways such as Sprouty2, PTEN and SFRP1. These are targets of miR-27a whose high expression is reduced by Qc. Moreover, miR-23a, and miR-24-2, the two other components of the unique gene cluster, and the pri-miRNA transcript are reduced, evoking a transcriptional regulation of the entire cluster by Sp1. Mechanistically, we show that Qc is rapidly internalized and localizes in the nucleus, where it likely interacts with Sp1, inducing its proteasomal degradation. Sp1 is further repressed by ZBTB10, an Sp1 competitor for DNA binding that is an miR-27a target and whose levels increase following Qc. SP1 mRNA is also reduced, supporting the regulation of its own gene transcription. Finally, Sp1 knockdown elicits the impaired transcription of the entire cluster and the upregulation of the miR-27a targets, phenocopying the effects of Qc. Through this dual mode of action, Qc counteracts the protumoral Sp1-miR-27a axis, opening the way for novel therapies based on its association as neoadjuvant with known anticancer treatments.

20.
Oncol Rep ; 50(3)2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37503758

RESUMO

Oxaliplatin (Oxa) is one of the most effective chemotherapeutic drugs used in the treatment of colorectal cancer (CRC). However, the use of this drug is associated with severe side­effects and patients eventually develop resistance to Oxa. In recent years, copper complexes have been extensively investigated as substitutes for platinum­based drugs. Therefore, a number of copper complexes have also been developed for cancer therapy, such as copper (II) complex of salicylate phenanthroline [Cu(sal)(phen)]. In the present study, the antitumor activity and the related molecular mechanisms of Cu(sal)(phen) were examined in CRC cells. As compared with the chemotherapeutic drug, Oxa, Cu(sal)(phen) was more effective in inducing apoptosis and reactive oxygen species (ROS) production, and in decreasing mitochondrial membrane potential in the CRC cell lines, HCT116 and SW480. In addition, the expression of the apoptosis­related proteins, Bcl­2 and survivin, and those of the upstream regulators, p­JAK2 and p­STAT5, were significantly decreased in the two cell lines following treatment with Cu(sal)(phen). Furthermore, the efficacy of the complex against CRC was found to be excellent in an animal model. The results of immunohistochemical analysis revealed that the expression levels of Bcl­2, survivin and Ki­67 in tumor tissues were decreased following Cu(sal)(phen) treatment. The antitumor mechanisms underlying Cu(Sal)(phen) treatment were the induction of ROS generation, the inhibition of the JAK2/STAT5 signaling pathway and the downregulation of the expression of anti­apoptotic proteins, such as Bcl­2 and survivin. On the whole, the findings of the present study indicated that Cu(sal)(phen) effectively inhibited the viability and proliferation of HCT116 and SW480 CRC cells; in the future, the authors aim to conduct further experiments in future studies to provide more evidence that supports the development of Cu(sal)(phen) as a therapeutic agent for CRC.


Assuntos
Antineoplásicos , Neoplasias Colorretais , Animais , Oxaliplatina/farmacologia , Cobre/farmacologia , Cobre/química , Cobre/metabolismo , Survivina/metabolismo , Fenantrolinas/farmacologia , Fenantrolinas/química , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Salicilatos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Antineoplásicos/farmacologia , Antineoplásicos/química , Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Linhagem Celular Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA