Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (95)

Search Parameters:
Keywords = SW837 human colorectal cancer cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 4414 KB  
Article
Investigating the Molecular Mechanisms of the Anticancer Effects of Eugenol and Cinnamaldehyde Against Colorectal Cancer (CRC) Cells In Vitro
by Alberto Bernacchi, Maria Chiara Valerii, Renato Spigarelli, Nikolas Kostantine Dussias, Fernando Rizzello and Enzo Spisni
Int. J. Mol. Sci. 2026, 27(2), 649; https://doi.org/10.3390/ijms27020649 - 8 Jan 2026
Viewed by 172
Abstract
Colorectal cancer is one of the leading causes of cancer-associated mortality, and multifactorial resistance remains one of the main challenges in its treatment. Essential oils and their main compounds show interesting anticancer properties, but their mechanism of action is yet to be defined. [...] Read more.
Colorectal cancer is one of the leading causes of cancer-associated mortality, and multifactorial resistance remains one of the main challenges in its treatment. Essential oils and their main compounds show interesting anticancer properties, but their mechanism of action is yet to be defined. This study aims to assess the cytotoxic effects of eugenol (EU) and cinnamaldehyde (CN) on colorectal cancer (CRC) cells, highlighting possible mechanisms of action. These compounds were tested on normal immortalized colonocytes (NCM-460) and two CRC cell lines: Caco-2, a human colon epithelial adenocarcinoma cell line, and SW-620, colon cancer cells derived from a lymph node metastatic site. The efficacy of EU and CN was evaluated through CellTiter-Glo® and clonogenic assays and by determining proinflammatory cytokine secretion. Transcriptome analysis was used to identify possible pathways affected by EU and CN treatments. The results confirmed that EU and CN were selectively cytotoxic and pro-apoptotic against CRC cells, with different putative mechanisms. While EU drove cytotoxicity through robust transcriptional remodeling, CN yielded a stronger anti-inflammatory action. We confirmed that EU and CN are promising natural candidates in CRC prevention and treatment, even in association with chemotherapeutic drugs. Full article
Show Figures

Figure 1

25 pages, 5307 KB  
Article
Fibrillarin Contributes to the Oncogenic Characteristics of Colorectal Cancer Cells and Reduces Sensitivity to 5-Fluorouracil
by Ting Wu, Mounira Chalabi-Dchar, Wei Xiong, Lucie Arnould, Eliezer Aimontche, Sabine Beaumel, Charles Dumontet, Virginie Marcel, Tanguy Fenouil, Jean-Jacques Diaz, Marie Alexandra Albaret and Hichem Claude Mertani
Cancers 2025, 17(24), 3900; https://doi.org/10.3390/cancers17243900 - 5 Dec 2025
Viewed by 402
Abstract
Background/Objectives: Fibrillarin (FBL) is a key nucleolar methyltransferase involved in ribosome biogenesis through 2′-O-ribose methylation of rRNA. While its oncogenic role has been reported in several cancer types, its expression and function in human colorectal cancer (CRC) have remained largely unexplored. This study [...] Read more.
Background/Objectives: Fibrillarin (FBL) is a key nucleolar methyltransferase involved in ribosome biogenesis through 2′-O-ribose methylation of rRNA. While its oncogenic role has been reported in several cancer types, its expression and function in human colorectal cancer (CRC) have remained largely unexplored. This study aims to investigate the expression of FBL in human CRC tissues and cell lines and to determine its functional role in tumor progression and metastasis. Methods: We examined FBL expression in paired human CRC primary tumors and liver metastases using immunohistochemistry. Functional studies were performed using SW-480 (primary tumor) and SW-620 (lymph node metastasis) CRC cell lines derived from the same patient. Cell migration, invasion, and 3D spheroid growth were analyzed following FBL downregulation. In vivo tumor growth was assessed in SCID mice xenografted with FBL-deficient cells. Molecular changes were explored through phosphorylation arrays and Western blotting. Results: FBL expression was significantly higher in human metastatic lesions than in primary tumors. FBL downregulation impaired migration, invasion, and spheroid growth in SW-480 and SW-620 cells and reduced tumor growth in vivo. Mechanistically, FBL inhibition decreased activation of MAPK/ERK, PI3K/AKT, and JNK/p38 pathways and reduced phosphorylation of the transcription factor CREB. Conclusions: Our study identifies FBL as a potential contributor to colorectal cancer progression, with elevated expression associated particularly with metastatic disease. By demonstrating that FBL expression is elevated in patient-derived metastatic tissues and functionally promotes migration, invasion, and tumor growth, our findings expand the role of ribosome biogenesis factors beyond protein synthesis. The observed suppression of key oncogenic pathways and CREB phosphorylation upon FBL inhibition suggests that FBL integrates ribosomal regulation with cancer cell signaling. These insights open new avenues for targeting nucleolar activity in advanced CRC and highlight FBL as a potential biomarker and therapeutic target in metastatic disease. Full article
(This article belongs to the Special Issue Colorectal Cancer Liver Metastases)
Show Figures

Figure 1

17 pages, 5442 KB  
Article
Solid-State Fermentation of Riceberry Rice with Mushroom Mycelium for Enhanced Beta-Glucan Production and Health Applications
by Jutamat Nacha, Hongyu Chen, Amorn Owatworakit, Kittirat Saharat, Anupong Makeudom and Sunita Chamyuang
Molecules 2025, 30(19), 3879; https://doi.org/10.3390/molecules30193879 - 25 Sep 2025
Viewed by 1573
Abstract
Beta-glucans (β-glucans), polysaccharides found in cereals and fungi, are recognized for their prebiotic and potential anti-cancer activities, particularly in the colorectal area. This study aims to optimize the production of β-glucan through the solid-state fermentation of germinated Riceberry rice with Pleurotus ostreatus and [...] Read more.
Beta-glucans (β-glucans), polysaccharides found in cereals and fungi, are recognized for their prebiotic and potential anti-cancer activities, particularly in the colorectal area. This study aims to optimize the production of β-glucan through the solid-state fermentation of germinated Riceberry rice with Pleurotus ostreatus and evaluate the bioactivities of the resulting extract. The crude β-glucan extract, obtained with a recovery rate of 54.95% and 79.98% purity, demonstrated an effective extraction process, as confirmed by thermogravimetric analysis (TGA). Fourier-transform infrared spectroscopy (FTIR) analysis verified the presence of β-1,3/1,6-glycosidic linkages, characteristic of the bioactive β-glucans found in yeast and mushrooms. The biological assessment demonstrated the extract’s functional properties. At a concentration of 1 mg/mL, the crude β-glucan extract significantly promoted the growth of probiotics Lacticaseibacillus rhamnosus and Bacillus coagulans, exhibiting high Prebiotic Index (PI) values of 6.36 ± 0.72 and 115.70 ± 10.19, respectively, with PI values indicating strong prebiotic potential. For comparison, the standard prebiotic inulin yielded PI values of 0.41 ± 0.09 and 90.53 ± 2.28 for the same respective bacteria, highlighting the superior performance of the fungal-fermented β-glucan. Furthermore, the extract displayed efficacy in inhibiting colon cancer cells in preliminary in vitro tests. It reduced the viability of the SW480 colorectal cancer cell line by 66.23% and induced cell death in 27.94 ± 0.93% of the cells after 48 h of treatment, performing comparably to a commercial yeast β-glucan standard. Crucially, the extract showed no significant cytotoxicity toward the normal human colon cell line, CCD-841 CoN. These findings highlight the promising method of fungal solid-state fermentation on germinated Riceberry rice in the production of high-purity, bioactive β-glucans for use in functional foods. Full article
(This article belongs to the Special Issue New Development in Fermented Products—Third Edition)
Show Figures

Graphical abstract

25 pages, 2959 KB  
Article
Synthesis, Characterization, HSA/DNA Binding, and Cytotoxic Activity of [RuCl26-p-cymene)(bph-κN)] Complex
by Stefan Perendija, Dušan Dimić, Thomas Eichhorn, Aleksandra Rakić, Luciano Saso, Đura Nakarada, Dragoslava Đikić, Teodora Dragojević, Jasmina Dimitrić Marković and Goran N. Kaluđerović
Molecules 2025, 30(15), 3088; https://doi.org/10.3390/molecules30153088 - 23 Jul 2025
Viewed by 973
Abstract
A novel ruthenium(II) complex, [RuCl26-p-cymene)(bph-κN)] (1), was synthesized and structurally characterized using FTIR and NMR spectroscopy. Density functional theory (DFT) calculations supported the proposed geometry and allowed for comparative analysis of experimental and [...] Read more.
A novel ruthenium(II) complex, [RuCl26-p-cymene)(bph-κN)] (1), was synthesized and structurally characterized using FTIR and NMR spectroscopy. Density functional theory (DFT) calculations supported the proposed geometry and allowed for comparative analysis of experimental and theoretical spectroscopic data. The interaction of complex 1 with human serum albumin (HSA) and calf thymus DNA was investigated through fluorescence quenching experiments, revealing spontaneous binding driven primarily by hydrophobic interactions. The thermodynamic parameters indicated mixed quenching mechanisms in both protein and DNA systems. Ethidium bromide displacement assays and molecular docking simulations confirmed DNA intercalation as the dominant binding mode, with a Gibbs free binding energy of −34.1 kJ mol−1. Antioxidant activity, assessed by EPR spectroscopy, demonstrated effective scavenging of hydroxyl and ascorbyl radicals. In vitro cytotoxicity assays against A375, MDA-MB-231, MIA PaCa-2, and SW480 cancer cell lines revealed selective activity, with pancreatic and colorectal cells showing the highest sensitivity. QTAIM analysis provided insight into metal–ligand bonding characteristics and intramolecular stabilization. These findings highlight the potential of 1 as a promising candidate for further development as an anticancer agent, particularly against multidrug-resistant tumors. Full article
(This article belongs to the Special Issue Transition Metal Complexes with Bioactive Ligands)
Show Figures

Figure 1

17 pages, 44923 KB  
Article
Inhibition of PCSK9 Attenuates Liver Endothelial Cell Activation Induced by Colorectal Cancer Stem Cells During Liver Metastasis
by Ander Martin, Daniela Gerovska, Marcos J. Arauzo-Bravo, Maitane Duarte García-Escudero, Helena García García, Iratxe Bañares, Naroa Fontal, Geraldine Siegfried, Serge Evrad, Simon Pernot, Abdel-Majid Khatib and Iker Badiola
Cancers 2025, 17(12), 1977; https://doi.org/10.3390/cancers17121977 - 13 Jun 2025
Viewed by 1682
Abstract
Background: Colorectal cancer (CRC) is among the most prevalent and lethal cancers globally, with liver metastasis representing the leading cause of CRC-related mortality. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has recently gained attention due to its overexpression in colorectal tumor tissues and its [...] Read more.
Background: Colorectal cancer (CRC) is among the most prevalent and lethal cancers globally, with liver metastasis representing the leading cause of CRC-related mortality. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has recently gained attention due to its overexpression in colorectal tumor tissues and its potential role in driving metastatic progression. This aims to investigate the involvement of PCSK9 in the liver metastatic niche, focusing on its effects on liver sinusoidal endothelial cells (LSECs), key components of the liver microenvironment. Methods: LSECs were stimulated with conditioned media derived from differentiated colorectal cancer cells and cancer stem cells (CSCs), the latter generated by reprogramming SW620 and CT26 cell lines. RNA sequencing was used to profile gene expression in LSECs. PCSK9 mRNA and protein levels were quantified by qPCR and Western blotting, respectively. PCSK9 expression in CRC liver metastases was evaluated by immunofluorescent staining. Results: PCSK9 was detected in both human and murine LSECs and significantly upregulated following exposure to CSC-conditioned media. Immunofluorescent staining confirmed PCSK9 expression in LSECs within CRC liver metastases. Total RNA sequencing revealed that a pre-treatment of LSECs with the PCSK9 inhibitor PF-06446864 prior to CSC stimulation seems to reduce the expression of microRNAs linked to cell migration and proliferation. Functional assays demonstrated that CSC-conditioned media enhanced LSEC proliferation and migration, effects reversed by PCSK9 inhibition. Conclusions: PCSK9 promotes the activation of LSECs in response to colorectal CSCs, contributing to a pro-metastatic phenotype. These findings highlight PCSK9 as a potential therapeutic target in colorectal liver metastasis. Full article
Show Figures

Figure 1

22 pages, 9088 KB  
Article
Inhibitory Effect and Mechanism of the Down-Regulation of TRIM32 in Colorectal Cancer
by Jiayu Ning, Xiaohua Cai, Yintong Su, Xingxing Fan and Mei Shen
Int. J. Mol. Sci. 2025, 26(11), 5047; https://doi.org/10.3390/ijms26115047 - 23 May 2025
Viewed by 1305
Abstract
TRIM32 protein represents a crucial member of TRIM family that is highly expressed in numerous human cancers, and is associated with a poor prognosis. However, the mechanism of TRIM32 in colorectal cancer (CRC) is unclear. The expression of TRIM32 and its prognostic value [...] Read more.
TRIM32 protein represents a crucial member of TRIM family that is highly expressed in numerous human cancers, and is associated with a poor prognosis. However, the mechanism of TRIM32 in colorectal cancer (CRC) is unclear. The expression of TRIM32 and its prognostic value in CRC were analyzed using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. Real-time quantitative PCR, immunohistochemistry (IHC), and cell proliferation assays were used to explore the effects of down-regulation of TRIM32 expression on the proliferation, migration, and apoptosis of cultured CRC cells (HCT116 and SW480 cells) and in xenogeneic tumorigenic animals. Bioinformatics analysis showed that TRIM32 is up-regulated in many types of cancers, and exhibits significant prognostic value in CRC. Western blotting results showed that after knocking down TRIM32, the expression level of IκBα increased, and the expression levels of TRIM32, p-p65, Bcl-2, and IKKβ decreased. The inhibitory effect of TRIM32 on CRC in vivo was evaluated by measuring tumor volume and weight, Hematoxylin and eosin (H&E) staining, and Ki67 IHC staining in heterotopic tumor-forming mice with CRC. Down-regulation of TRIM32 can inhibit the activation of the NF-κB signaling pathway and the occurrence of CRC. Our research provides a new insight into the pathogenesis of CRC, and a therapeutic target for the treatment of CRC. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

17 pages, 1625 KB  
Article
Phenolic Composition, Antioxidant, and Anti-Proliferative Activities Against Human Colorectal Cancer Cells of Amazonian Fruits Copoazú (Theobroma grandiflorum) and Buriti (Mauritia flexuosa)
by Sebastián Saldarriaga, Carlos Andrés Rodríguez-Salazar, Delia Piedad Recalde-Reyes, Gloria Magally Paladines Beltrán, Liceth N. Cuéllar Álvarez and Yudy Lorena Silva Ortíz
Molecules 2025, 30(6), 1250; https://doi.org/10.3390/molecules30061250 - 11 Mar 2025
Cited by 8 | Viewed by 3953
Abstract
Amazonian fruits are a source of bioactive compounds, among which phenolic compounds, flavonoids, and carotenes stand out. These compounds play a crucial role in restoring oxidative balance, consequently reducing the proliferation of cancer cells. However, the content of these metabolites and their biological [...] Read more.
Amazonian fruits are a source of bioactive compounds, among which phenolic compounds, flavonoids, and carotenes stand out. These compounds play a crucial role in restoring oxidative balance, consequently reducing the proliferation of cancer cells. However, the content of these metabolites and their biological properties may vary significantly depending on the geographical location and the environmental conditions where plants grow. This research assessed the content of metabolites, free radical scavenging capacity, and hemolytic and antiproliferative effects of the hydro-methanolic extracts of the Amazonian fruits Theobroma grandiflorum and Mauritia flexuosa. The results revealed that the extracts derived from the seeds of Theobroma grandiflorum sourced from the Balcanes experimental farm and the pulp of Mauritia flexuosa harvested in Florencia exhibited higher contents compared to other analyzed sites: Total phenolic content (TPC) (619.41 ± 12.05 and 285.75 ± 10.06 mg GAE/100 g FW), Total flavonoid content (TFC) (569.09 ± 4.51 and 223.21 ± 3.92 mg CAT/100 g FW), and Total carotenoid content (TCC) (25.12 ± 0.16 and 48.00 ± 0.28 mg eq β-carotene/100 g FW), respectively. Also, these samples demonstrated superior scavenging capacities for the ABTS and DPPH radicals, while the peel of Mauritia flexuosa exhibited the highest scavenging capacity for the oxygen radical (526.23 ± 2.08 µmol Trolox.g−1). The hemolytic effect shows dose-dependent responses with IC50 values of 27.73 μg/mL for the Balcanes seeds and 1.27 μg/mL for the Florencia pulp. Furthermore, it was observed that treatment with the fruit-derived extracts effectively reduced the number of viable human colorectal cancer cells, using SW480 ATCC cell line, demonstrating a non-dose-dependent behavior compared to the control cells. Full article
Show Figures

Graphical abstract

16 pages, 4948 KB  
Article
Identification of Lipid Species Signatures in FOLFOXIRI-Resistant Colorectal Cancer Cells
by George M. Ramzy, Isabel Meister, Serge Rudaz, Julien Boccard and Patrycja Nowak-Sliwinska
Int. J. Mol. Sci. 2025, 26(3), 1169; https://doi.org/10.3390/ijms26031169 - 29 Jan 2025
Cited by 2 | Viewed by 2377
Abstract
Chronic drug treatment can alter the lipidome of cancer cells, potentially leading to significant biological changes, such as drug resistance or increased tumor aggressiveness. This study examines the lipidome profiles of four human colorectal cancer (CRC) cell lines, comparing treatment-naïve cells with the [...] Read more.
Chronic drug treatment can alter the lipidome of cancer cells, potentially leading to significant biological changes, such as drug resistance or increased tumor aggressiveness. This study examines the lipidome profiles of four human colorectal cancer (CRC) cell lines, comparing treatment-naïve cells with the same cells after chronic exposure to a clinically used combination therapy (FOLFOXIRI: folinic acid, 5-fluorouracil, oxaliplatin, and irinotecan). Lipidomic profiling was obtained with untargeted liquid chromatography coupled with high-resolution mass spectrometry (LC-HRMS). For data deconvolution and to interpret the multifactorial dataset generated, Analysis of Variance Multiblock Orthogonal Partial Least Squares (AMOPLS) was used. Our results indicate specific shifts in triglycerides (TGs), sphingolipids, and phospholipids in CRC cells resistant to FOLFOXIRI. The overall shift in TGs, phosphatidylcholine, and cholesteryl ester species was notably linked to FOLFOXIRI resistance (-R) in SW620 cells, whereas an increased abundance of phospholipids, mainly hexosylceramide and sphingomyelin, was present in the signatures of HCT116-R, LS174T-R, and DLD1-R cells. These altered lipid species may serve as potential prognostic markers in CRC following chemotherapy. Furthermore, lipid-targeting therapies aimed at reprogramming the lipid profiles of drug-resistant cells could play a crucial role in restoring drug sensitivity and improving patient survival. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

23 pages, 9051 KB  
Article
Synthetic CB1 Cannabinoids Promote Tunneling Nanotube Communication, Cellular Migration, and Epithelial–Mesenchymal Transition in Pancreatic PANC-1 and Colorectal SW-620 Cancer Cell Lines
by David A. Bunsick, Leili Baghaie, Yunfan Li, Abdulrahman M. Yaish, Emilyn B. Aucoin, Elizabeth Skapinker, Rashelle Aldbai and Myron R. Szewczuk
Cells 2025, 14(2), 71; https://doi.org/10.3390/cells14020071 - 7 Jan 2025
Cited by 2 | Viewed by 2262
Abstract
Metastasizing cancer cells surreptitiously can adapt to metabolic activity during their invasion. By initiating their communications for invasion, cancer cells can reprogram their cellular activities to initiate their proliferation and migration and uniquely counteract metabolic stress during their progression. During this reprogramming process, [...] Read more.
Metastasizing cancer cells surreptitiously can adapt to metabolic activity during their invasion. By initiating their communications for invasion, cancer cells can reprogram their cellular activities to initiate their proliferation and migration and uniquely counteract metabolic stress during their progression. During this reprogramming process, cancer cells’ metabolism and other cellular activities are integrated and mutually regulated by tunneling nanotube communications to alter their specific metabolic functional drivers of tumor growth and progression. Here, we investigated the in vitro effects of the synthetic CB1 cannabinoids AM-404, arvanil, and olvanil on human pancreatic PANC-1 and colorectal SW-620 cancer cell lines to understand further cellular behaviors and the potential risks of their use in cancer therapy. For the first time, the synthetic CB1 cannabinoids AM-404, arvanil, and olvanil significantly altered cancer cells in forming missile-like shapes to induce tunneling nanotube (TNT) communications in PANC-1 cells. Oseltamivir phosphate (OP) significantly prevented TNT formation. To assess the key survival pathways critical for pancreatic cancer progression, we used the AlamarBlue assay to determine synthetic CB1 cannabinoids to induce the cell’s metabolic viability drivers to stage migratory intercellular communication. The synthetic CB1 cannabinoids significantly increased cell viability compared to the untreated control for PANC-1 and SW-620 cells, and this response was significantly reduced with the NMBR inhibitor BIM-23127, neuraminidase-1 inhibitor OP, and MMP-9 inhibitor (MMP-9i). CB1 cannabinoids also significantly increased N-cadherin and decreased E-cadherin EMT markers compared to the untreated controls, inducing the process of metastatic phenotype for invasion. BIM-23127, MMP9i, and OP significantly inhibited CB1 agonist-induced NFκB-dependent secretory alkaline phosphatase (SEAP) activity. To confirm this concept, we investigated the migratory invasiveness of PANC-1 and SW-620 cancer cells treated with the synthetic CB1 cannabinoids AM-404, arvanil, and olvanil in a scratch wound assay. CB1 cannabinoids significantly induced the rate of migration and invasiveness of PANC-1 cancer cells, whereas they had minimal effect on the rate of migration of already metastatic SW-620 cancer cells. Interestingly, olvanil-treated SW-620 cells significantly enhanced the migration rate and invasiveness of these cells. The data support the cellular and molecular mechanisms of the synthetic CB1 cannabinoids, orchestrating intercellular conduits to enhance metabolic drivers to stage migratory intercellular communication in pancreatic cancer cells. Full article
(This article belongs to the Special Issue Cellular and Molecular Mechanisms of Cancer Invasion and Metastasis)
Show Figures

Figure 1

13 pages, 3547 KB  
Article
Protective Effects of Astaxanthin against Oxidative Stress: Attenuation of TNF-α-Induced Oxidative Damage in SW480 Cells and Azoxymethane/Dextran Sulfate Sodium-Induced Colitis-Associated Cancer in C57BL/6 Mice
by Haifeng Zhang, Min Wang, Yu Zhou, Shaojie Bao, Feng Wang and Chunmei Li
Mar. Drugs 2024, 22(10), 469; https://doi.org/10.3390/md22100469 - 12 Oct 2024
Cited by 8 | Viewed by 3578
Abstract
In this study, we investigated the protective effects of astaxanthin (AST) against oxidative stress induced by the combination of azoxymethane (AOM) and dextran sulfate sodium (DSS) in colitis-associated cancer (CAC) and TNF-α-induced human colorectal cancer cells (SW480), as well as the underlying mechanism. [...] Read more.
In this study, we investigated the protective effects of astaxanthin (AST) against oxidative stress induced by the combination of azoxymethane (AOM) and dextran sulfate sodium (DSS) in colitis-associated cancer (CAC) and TNF-α-induced human colorectal cancer cells (SW480), as well as the underlying mechanism. In vitro experiments revealed that astaxanthin reduced reactive oxygen species (ROS) generation and inhibited the expression of Phosphorylated JNK (P-JNK), Phosphorylated ERK (P-ERK), Phosphorylated p65 (P-p65), and the NF-κB downstream protein cyclooxygenase-2 (COX-2). In vivo experiments showed that astaxanthin ameliorated AOM/DSS-induced weight loss, shortened the colon length, and caused histomorphological changes. In addition, astaxanthin suppressed cellular inflammation by modulating the MAPK and NF-κB pathways and inhibiting the expression of the proinflammatory cytokines IL-6, IL-1β, and TNF-α. In conclusion, astaxanthin attenuates cellular inflammation and CAC through its antioxidant effects. Full article
(This article belongs to the Special Issue Application of Marine Nature Products to Reduce Oxidative Stress)
Show Figures

Graphical abstract

13 pages, 4738 KB  
Article
A Benzimidazole-Based N-Heterocyclic Carbene Derivative Exhibits Potent Antiproliferative and Apoptotic Effects against Colorectal Cancer
by Sarah Al-Nasser, Maha Hamadien Abdulla, Noura Alhassan, Mansoor-Ali Vaali-Mohammed, Suliman Al-Omar, Naceur Hamdi, Yasser Elnakady, Sabine Matou-Nasri and Lamjed Mansour
Medicina 2024, 60(9), 1379; https://doi.org/10.3390/medicina60091379 - 23 Aug 2024
Cited by 4 | Viewed by 2028
Abstract
Background and Objectives: Colorectal cancer (CRC) remains a major global health issue. Although chemotherapy is the first-line treatment, its effectiveness is limited due to drug resistance developed in CRC. To overcome resistance and improve the prognosis of CRC patients, investigating new therapeutic [...] Read more.
Background and Objectives: Colorectal cancer (CRC) remains a major global health issue. Although chemotherapy is the first-line treatment, its effectiveness is limited due to drug resistance developed in CRC. To overcome resistance and improve the prognosis of CRC patients, investigating new therapeutic approaches is necessary. Materials and Methods: Using human colorectal adenocarcinoma (HT29) and metastatic CRC (SW620) cell lines, the potential anticancer properties of a newly synthesized compound 1-(Isobutyl)-3-(4-methylbenzyl) benzimidazolium chloride (IMBZC) were evaluated by performing MTT cytotoxicity, cell migration, and colony formation assays, as well as by monitoring apoptosis-related protein and gene expression using Western blot and reverse transcription–quantitative polymerase chain reaction technologies. Results: Tested at various concentrations, the half-maximal inhibitory concentrations (IC50) of IMBZC on HT29 and SW620 cell growth were determined to be 22.13 µM (6.97 μg/mL) and 15.53 µM (4.89 μg/mL), respectively. IMBZC did not alter the cell growth of normal HEK293 cell lines. In addition, IMBZC inhibited cell migration and significantly decreased colony formation, suggesting its promising role in suppressing cancer metastasis. Mechanistic analyses revealed that IMBZC treatment increased the expression of pro-apoptotic proteins p53 and Bax, while decreasing the expression of anti-apoptotic proteins Bcl-2 and Bcl-xL, thus indicating the induction of apoptosis in IMBZC-treated CRC cells, compared to untreated cells. Additionally, the addition of IMBZC to conventional chemotherapeutic drugs (i.e., 5-fluorouracil, irinotecan, and oxaliplatin) resulted in an increase in the cytotoxic potential of the drugs. Conclusions: This study suggests that IMBZC has substantial anticancer effects against CRC cells through its ability to induce apoptosis, inhibit cancer cell migration and colony formation, and enhance the cytotoxic effects of conventional chemotherapeutic drugs. These findings indicate that IMBZC could be a promising chemotherapeutic drug for the treatment of CRC. Further research should be conducted using in vivo models to confirm the anti-CRC activities of IMBZC. Full article
(This article belongs to the Section Gastroenterology & Hepatology)
Show Figures

Figure 1

24 pages, 5703 KB  
Article
Quantitative Analysis of Isopimpinellin from Ammi majus L. Fruits and Evaluation of Its Biological Effect on Selected Human Tumor Cells
by Magdalena Bartnik, Adrianna Sławińska-Brych, Magdalena Mizerska-Kowalska, Anna Karolina Kania and Barbara Zdzisińska
Molecules 2024, 29(12), 2874; https://doi.org/10.3390/molecules29122874 - 17 Jun 2024
Cited by 5 | Viewed by 2971
Abstract
Ammi majus L. (Apiaceae) is a medicinal plant with a well-documented history in phytotherapy. The aim of the present work was to isolate isopimpinellin (5,8-methoxypsoralen; IsoP) from the fruit of this plant and evaluate its biological activity against selected tumor cell lines. The [...] Read more.
Ammi majus L. (Apiaceae) is a medicinal plant with a well-documented history in phytotherapy. The aim of the present work was to isolate isopimpinellin (5,8-methoxypsoralen; IsoP) from the fruit of this plant and evaluate its biological activity against selected tumor cell lines. The methanol extract obtained with the use of an accelerated solvent extraction (ASE) method was the most suitable for the quantitative analysis of coumarins in the A. majus fruit matrix. The coumarin content was estimated by RP-HPLC/DAD, and the amount of IsoP was found to be 404.14 mg/100 g dry wt., constituting 24.56% of the total coumarin fraction (1.65 g/100 g). This, along with the presence of xanthotoxin (368.04 mg/100 g, 22.36%) and bergapten (253.05 mg/100 g, 15.38%), confirmed A. majus fruits as an excellent source of these compounds. IsoP was isolated (99.8% purity) by combined liquid chromatography/centrifugal partition chromatography (LC/CPC) and tested for the first time on its antiproliferative activity against human colorectal adenocarcinoma (HT29, SW620), osteosarcoma (Saos-2, HOS), and multiple myeloma (RPMI8226, U266) cell lines. MTT assay results (96 h incubation) demonstrated a dose- and cell line-dependent decrease in cell proliferation/viability, with the strongest effect of IsoP against the Saos-2 cell line (IC50; 42.59 µM), medium effect against U266, HT-29, and RPMI8226 (IC50 = 84.14, 95.53, and 105.0 µM, respectively), and very weak activity against invasive HOS (IC50; 321.6 µM) and SW620 (IC50; 711.30 µM) cells, as well as normal human skin fibroblasts (HSFs), with IC50; 410.7 µM. The mechanistic study on the Saos-2 cell line showed that IsoP was able to reduce DNA synthesis and trigger apoptosis via caspase-3 activation. In general, IsoP was found to have more potency towards cancerous cells (except for HOS and SW620) than against healthy cells. The Selective Index (SI) was determined, underlining the higher selectivity of IsoP towards cancer cells compared to healthy cells (SI = 9.62 against Saos-2). All these results suggest that IsoP might be a promising molecule in the chemo-prevention and treatment of primary osteosarcoma. Full article
Show Figures

Graphical abstract

22 pages, 3368 KB  
Article
General Direct Anticancer Effects of Deer Growing Antler Extract in Several Tumour Cell Lines, and Immune System-Mediated Effects in Xenograft Glioblastoma
by Alessandra Rossetti, Louis Chonco, Nicolas Alegría, Veronica Zelli, Andrés J. García, Carmen Ramírez-Castillejo, Alessandra Tessitore, Carlos de Cabo, Tomás Landete-Castillejos and Claudio Festuccia
Pharmaceutics 2024, 16(5), 610; https://doi.org/10.3390/pharmaceutics16050610 - 30 Apr 2024
Cited by 2 | Viewed by 6032
Abstract
Deer antlers are the fastest growing tissue. Because they are based on proto-oncogenes, to avoid the risk of cancer, antlers evolved strong anticancer mechanisms, and thus their extract (DVA) is effective also against the few human tumours studied so far. We assessed whether [...] Read more.
Deer antlers are the fastest growing tissue. Because they are based on proto-oncogenes, to avoid the risk of cancer, antlers evolved strong anticancer mechanisms, and thus their extract (DVA) is effective also against the few human tumours studied so far. We assessed whether DVA is a general anticancer compound by testing the direct effects in cells of different tumours: glioblastoma (GBM; lines U87MG and U251), colorectal (CRC; lines DLD-1, HT-29, SW480, and SW620), breast cancer (BRCA; lines MCF7, SKBR3, and PA00), and leukaemia (THP-1). DVA reduced the viability of tumours but not healthy cells (NHC; lines 293T and HaCaT). Mobility decreased at least for the longest test (72 h). Intraperitoneal/oral 200 mg DVA/kg administration in GBM xenograft mice for 28 d reduced tumour weight by 66.3% and 61.4% respectively, and it also reduced spleen weight (43.8%). In addition, tumours treated with DVA showed symptoms of liquefactive necrosis. Serum cytokines showed DVA up-regulated factors related to tumour fighting and down-regulated those related to inducing immune tolerance to the tumour. DVA shows general anticancer effects in the lines tested and, in GBM mice, also strong indirect effects apparently mediated by the immune system. DVA may contain a future anticancer medicine without secondary effects. Full article
Show Figures

Figure 1

10 pages, 1451 KB  
Brief Report
Rescue of Methionine Dependence by Cobalamin in a Human Colorectal Cancer Cell Line
by Sarita Garg and Isabelle R. Miousse
Nutrients 2024, 16(7), 997; https://doi.org/10.3390/nu16070997 - 28 Mar 2024
Cited by 6 | Viewed by 2535
Abstract
Methionine dependence is a characteristic of most cancer cells where they are unable to proliferate when the essential amino acid methionine is replaced with its precursor homocysteine in the growing media. Normal cells, on the other hand, thrive under these conditions and are [...] Read more.
Methionine dependence is a characteristic of most cancer cells where they are unable to proliferate when the essential amino acid methionine is replaced with its precursor homocysteine in the growing media. Normal cells, on the other hand, thrive under these conditions and are referred to as methionine-independent. The reaction that adds a methyl group from 5-methyltetrahydrofolate to homocysteine to regenerate methionine is catalyzed by the enzyme methionine synthase with the cofactor cobalamin (vitamin B12). However, decades of research have shown that methionine dependence in cancer is not due to a defect in the activity of methionine synthase. Cobalamin metabolism has been tied to the dependent phenotype in rare cell lines. We have identified a human colorectal cancer cell line in which the cells regain the ability to proliferation in methionine-free, L-homocystine-supplemented media when cyanocobalamin is supplemented at a level of 1 µg/mL. In human SW48 cells, methionine replacement with L-homocystine does not induce any measurable increase in apoptosis or reactive oxygen species production in this cell line. Rather, proliferation is halted, then restored in the presence of cyanocobalamin. Our data show that supplementation with cyanocobalamin prevents the activation of the integrated stress response (ISR) in methionine-deprived media in this cell line. The ISR-associated cell cycle arrest, characteristic of methionine-dependence in cancer, is also prevented, leading to the continuation of proliferation in methionine-deprived SW48 cells with cobalamin. Our results highlight differences between cancer cell lines in the response to cobalamin supplementation in the context of methionine dependence. Full article
(This article belongs to the Special Issue Methyl Nutrients and One-Carbon Metabolism in Chronic Diseases)
Show Figures

Figure 1

27 pages, 6411 KB  
Article
Modes of Interactions with DNA/HSA Biomolecules and Comparative Cytotoxic Studies of Newly Synthesized Mononuclear Zinc(II) and Heteronuclear Platinum(II)/Zinc(II) Complexes toward Colorectal Cancer Cells
by Samir Vučelj, Rušid Hasić, Darko Ašanin, Biljana Šmit, Angelina Caković, Jovana Bogojeski, Marina Ćendić Serafinović, Bojana Simović Marković, Bojan Stojanović, Sladjana Pavlović, Isidora Stanisavljević, Irfan Ćorović, Milica Dimitrijević Stojanović, Ivan Jovanović, Tanja V. Soldatović and Bojana Stojanović
Int. J. Mol. Sci. 2024, 25(5), 3027; https://doi.org/10.3390/ijms25053027 - 6 Mar 2024
Cited by 5 | Viewed by 2450
Abstract
A series of mono- and heteronuclear platinum(II) and zinc(II) complexes with 4,4′,4″-tri-tert-butyl-2,2′:6′,2″-terpyridine ligand were synthesized and characterized. The DNA and protein binding properties of [ZnCl2(terpytBu)] (C1), [{cis-PtCl(NH3)2(μ [...] Read more.
A series of mono- and heteronuclear platinum(II) and zinc(II) complexes with 4,4′,4″-tri-tert-butyl-2,2′:6′,2″-terpyridine ligand were synthesized and characterized. The DNA and protein binding properties of [ZnCl2(terpytBu)] (C1), [{cis-PtCl(NH3)2(μ-pyrazine)ZnCl(terpytBu)}](ClO4)2 (C2), [{trans-PtCl(NH3)2(μ-pyrazine)ZnCl(terpytBu)}](ClO4)2 (C3), [{cis-PtCl(NH3)2(μ-4,4′-bipyridyl)ZnCl(terpytBu)}](CIO4)2 (C4) and [{trans-PtCl(NH3)2(μ-4,4′-bipyridyl)ZnCl(terpytBu)}](CIO4)2 (C5) (where terpytBu = 4,4′,4″-tri-tert-butyl-2,2′:6′,2″-terpyridine), were investigated by electronic absorption, fluorescence spectroscopic, and molecular docking methods. Complexes featuring transplatin exhibited lower Kb and Ksv constant values compared to cisplatin analogs. The lowest Ksv value belonged to complex C1, while C4 exhibited the highest. Molecular docking studies reveal that the binding of complex C1 to DNA is due to van der Waals forces, while that of C2C5 is due to conventional hydrogen bonds and van der Waals forces. The tested complexes exhibited variable cytotoxicity toward mouse colorectal carcinoma (CT26), human colorectal carcinoma (HCT116 and SW480), and non-cancerous mouse mesenchymal stem cells (mMSC). Particularly, the mononuclear C1 complex showed pronounced selectivity toward cancer cells over non-cancerous mMSC. The C1 complex notably induced apoptosis in CT26 cells, effectively arrested the cell cycle in the G0/G1 phase, and selectively down-regulated Cyclin D. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Graphical abstract

Back to TopTop