Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.881
Filter
1.
J Biochem Mol Toxicol ; 38(8): e23771, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39015057

ABSTRACT

Colon cancer contributes to high mortality rates internationally that has seriously endangered human health. Aurora kinase A (AURKA) served as a key molecule in colon cancer. However, its role of AURKA on regulating ferroptosis in colon cancer and their possible interactions with miRNAs and circRNAs remain still elusive. Comprehensive bioinformatics analysis after RNA-sequencing was conducted to determine the differentially expressed genes (DEGs), ferroptosis-related DEGs and hub genes. The direct relationship between miR-506-3p and hsa_circRNA_007630 or AURKA was predicted, then verified by dual luciferase reporter and quantitative real-time polymerase chain reaction. The rescue experiments were conducted by cotransfection with si-hsa_circRNA_007630, miR-506-3p inhibitor or pcDNA-AURKA in HT29 cells. Erastin was used to induce ferroptosis in HT29 cells and validated by detecting levels of intracellular Fe2+, lipid reactive oxygen species, glutathione, malondialdehyde and ferroptosis markers expression. We screened a total of 331 DEGs, 26 ferroptosis-related genes, among which 3 hub genes were identified through PPI network analysis. Therein, AURKA expression was elevated in colon cancer cells. Moreover, AURKA was targeted by miR-506-3p, and hsa_circRNA_007630 operated as miR-506-3p sponge. The effect of hsa_circRNA_007630 depletion on the inhibiting malignant phenotypes of HT29 cells was rescued by inhibition of miR-506-3p or AURKA overexpression. Additionally, AURKA reduced erastin-induced ferroptosis in HT29 cells. Depletion of circRNA_007630 exerts as a suppressive role in colon cancer through a novel miR-506-3p/AURKA pathway related to ferroptosis, and might become a novel marker for colon cancer.


Subject(s)
Aurora Kinase A , Colonic Neoplasms , Ferroptosis , MicroRNAs , RNA, Circular , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Ferroptosis/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , HT29 Cells , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Disease Progression , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
2.
Biomed Res Int ; 2024: 6231095, 2024.
Article in English | MEDLINE | ID: mdl-39015603

ABSTRACT

Background: Studies have concentrated on the therapeutic potential of thymoquinone (TQ), a natural polyphenol, in diverse malignancies, such as colorectal cancer. Nevertheless, the precise mechanisms of TQ-mediated anticancer properties are not yet fully elucidated. Objective: The present study has been designed to scrutinize the impact of TQ on 5-fluorouracil (5-FU)-mediated apoptosis in SW-480 cells. Materials and Methods: SW-480 cells were treated with TQ, 5-FU, and a combination of TQ + 5-FU. MTT assay was employed to assess cell viability. Quantitative real-time polymerase chain reaction (qRT-PCR) was applied to evaluate apoptotic markers comprising Bcl-2, Bax, and caspase-9 expression levels. The γ-H2AX protein expression was assessed by western blotting, and Annexin V flow cytometry was implemented to determine the apoptosis rate. Results: 5-FU significantly reversed the cell proliferation in a dose-dependent circumstance. The concurrent administration of TQ and 5-FU led to a substantial inhibition of cell growth in comparison to single treatments (p < 0.05). TQ also facilitated apoptosis via upregulating Bax and caspase-9 proapoptotic markers and suppressing antiapoptotic mediators, like Bcl-2. In addition, TQ augmented 5-FU-induced apoptosis in SW-480 cells. 5-FU, combined with TQ, increased the protein expression of γ-H2AX in SW-480 cells compared with groups treated with TQ and 5-FU alone. Conclusion: The present study's findings unveil the significance of TQ as a potential therapeutic substance in colorectal cancer, particularly through enhancing 5-FU-induced apoptosis.


Subject(s)
Apoptosis , Benzoquinones , Cell Proliferation , Colonic Neoplasms , Fluorouracil , Humans , Fluorouracil/pharmacology , Benzoquinones/pharmacology , Cell Line, Tumor , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cell Proliferation/drug effects , bcl-2-Associated X Protein/metabolism , Cell Survival/drug effects , Caspase 9/metabolism , Caspase 9/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism
3.
BMC Gastroenterol ; 24(1): 222, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992586

ABSTRACT

BACKGROUND: CFAP65 (cilia and flagella associated protein 65) is a fundamental protein in the development and formation of ciliated flagella, but few studies have focused on its role in cancer. This study aimed to investigate the prognostic significance of CFAP65 in colon cancer. METHODS: The functionally enriched genes related to CFAP65 were analyzed through the Gene Ontology (GO) database. Subsequently, CFAP65 expression levels in colon cancer were evaluated by reverse transcription and quantitative polymerase chain reaction (RT-qPCR) and immunoblotting in 20 pairs of frozen samples, including tumors and their matched paratumor tissue. Furthermore, protein expression of CFAP65 in 189 colon cancer patients were assessed via immunohistochemical staining. The correlations between CFAP65 expression and clinical features as well as long-term survival were statistically analyzed. RESULTS: CFAP65-related genes are significantly enriched on cellular processes of cell motility, ion channels, and GTPase-associated signaling. The expression of CFAP65 was significantly higher in colon cancer tissue compared to paratumor tissue. The proportion of high expression and low expression of CFAP65 in the clinical samples of colon cancer were 61.9% and 38.1%, respectively, and its expression level was not associated with the clinical parameters including gender, age, tumor location, histological differentiation, tumor stage, vascular invasion and mismatch repair deficiency. The five-year disease-free survival rate of the patients with CFAP65 low expression tumors was significantly lower than that those with high expression tumors (56.9% vs. 72.6%, P = 0.03), but the overall survival rate has no significant difference (69% vs. 78.6%, P = 0.171). The cox hazard regression analysis model showed that CFAP65 expression, tumor stage and tumor location were independent prognostic factors. CONCLUSIONS: In conclusion, we demonstrate CFAP65 is a potential predictive marker for tumor progression in colon cancer.


Subject(s)
Biomarkers, Tumor , Colonic Neoplasms , Humans , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Male , Female , Middle Aged , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Aged , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Clinical Relevance , Membrane Proteins , Neoplasm Proteins
4.
J Photochem Photobiol B ; 257: 112966, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38970968

ABSTRACT

BACKGROUND/AIM: Although photobiomodulation therapy (PBMt) is available to alleviate post-operative side effects of malignant diseases, its application is still controversial due to some potential of cancer recurrence and occurrence of a secondary malignancy. We investigated effect of PBMt on mitochondrial function in HT29 colon cancer cells. METHODS: HT29 cell proliferation was determined with MTT assay after PBMt. Immunofluorescent staining was performed to determine mitochondrial biogenesis and reactive oxygen species (ROS). Mitochondrial membrane potential was measured with Mitotracker. Western blotting was executed to determine expression of fission, fusion, UCP2, and cyclin B1 and D1 proteins. In vivo study was performed by subcutaneously inoculating cancer cells into nude mice and immunohistochemistry was done to determine expression of FIS1, MFN2, UCP2, and p-AKT. RESULTS: The proliferation and migration of HT29 cells reached maximum with PBMt (670 nm, light emitting diode, LED) at 2.0 J/cm2 compared to control (P < 0.05) with more expression of cyclin B1 and cyclin D1 (P < 0.05). Immunofluorescent staining showed that ROS and mitochondrial membrane potential were enhanced after PBMt compared to control. ATP synthesis of mitochondria was also higher in the PBMt group than in the control (P < 0.05). Expression levels of fission and fusion proteins were significantly increased in the PBMt group than in the control (P < 0.05). Electron microscopy revealed that the percentage of mitochondria showing fission was not significantly different between the two groups. Oncometabolites including D-2-hydoxyglutamate in the supernatant of cell culture were higher in the PBMt group than in the control with increased UCP2 expression (P < 0.05). Both tumor size and weight of xenograft in nude mice model were bigger and heavier in the PBMt group than in the control (P < 0.05). Immunohistologically, mitochondrial biogenesis proteins UCP2 and p-AKT in xenograft of nude mice were expressed more in the PBMt group than in the control (P < 0.05). CONCLUSIONS: Treatment with PBM using red light LED may induce proliferation and progression of HT29 cancer cells by increasing mitochondrial activity and fission.


Subject(s)
Cell Proliferation , Colonic Neoplasms , Membrane Potential, Mitochondrial , Mice, Nude , Mitochondria , Reactive Oxygen Species , Humans , HT29 Cells , Mitochondria/metabolism , Mitochondria/radiation effects , Animals , Cell Proliferation/radiation effects , Mice , Reactive Oxygen Species/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/metabolism , Membrane Potential, Mitochondrial/radiation effects , Low-Level Light Therapy , Cell Movement/radiation effects , Cyclin B1/metabolism , Mitochondrial Dynamics/radiation effects , Cyclin D1/metabolism , Proto-Oncogene Proteins c-akt/metabolism
5.
Molecules ; 29(13)2024 Jun 23.
Article in English | MEDLINE | ID: mdl-38998936

ABSTRACT

Metabolic alterations are increasingly recognized as important aspects of colorectal cancer (CRC), offering potential avenues for identifying therapeutic targets. Previous studies have demonstrated the cytotoxic potential of bamboo leaf extract obtained from Guadua incana (BLEGI) against HCT-116 colon cancer cells. However, the altered metabolic pathways in these tumor cells remain unknown. Therefore, this study aimed to employ an untargeted metabolomic approach to reveal the metabolic alterations of the endometabolome and exometabolome of HCT-116 cells upon exposure to BLEGI treatment. First, a chemical characterization of the BLEGI was conducted through liquid chromatography coupled with mass spectrometry (LC-MS). Next, we assessed cell viability via MTT and morphological analysis using an immunofluorescence assay against colon cancer cells, and anti-inflammatory activity using an LPS-stimulated macrophage model. Subsequently, we employed LC-MS and proton nuclear magnetic resonance (1H-NMR) to investigate intra- and extracellular changes. Chemical characterization primarily revealed the presence of compounds with a flavone glycoside scaffold. Immunofluorescence analysis showed condensed chromatin and subsequent formation of apoptotic bodies, suggesting cell death by apoptosis. The results of the metabolomic analysis showed 98 differential metabolites, involved in glutathione, tricarboxylic acid cycle, and lipoic acid metabolism, among others. Additionally, BLEGI demonstrated significant nitric oxide (NO) inhibitory capacity in macrophage cells. This study enhances our understanding of BLEGI's possible mechanism of action and provides fresh insights into therapeutic targets for treating this disease.


Subject(s)
Colonic Neoplasms , Plant Extracts , Plant Leaves , Humans , Plant Extracts/pharmacology , Plant Extracts/chemistry , Plant Leaves/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Metabolomics/methods , Metabolome/drug effects , Cell Survival/drug effects , Apoptosis/drug effects , Animals , RAW 264.7 Cells , Mice , Chromatography, Liquid
6.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000160

ABSTRACT

222 nm far-ultraviolet (F-UV) light has a bactericidal effect similar to deep-ultraviolet (D-UV) light of about a 260 nm wavelength. The cytotoxic effect of 222 nm F-UV has not been fully investigated. DLD-1 cells were cultured in a monolayer and irradiated with 222 nm F-UV or 254 nm D-UV. The cytotoxicity of the two different wavelengths of UV light was compared. Changes in cell morphology after F-UV irradiation were observed by time-lapse imaging. Differences in the staining images of DNA-binding agents Syto9 and propidium iodide (PI) and the amount of cyclobutane pyrimidine dimer (CPD) were examined after UV irradiation. F-UV was cytotoxic to the monolayer culture of DLD-1 cells in a radiant energy-dependent manner. When radiant energy was set to 30 mJ/cm2, F-UV and D-UV showed comparable cytotoxicity. DLD-1 cells began to expand immediately after 222 nm F-UV light irradiation, and many cells incorporated PI; in contrast, PI uptake was at a low level after D-UV irradiation. The amount of CPD, an indicator of DNA damage, was higher in cells irradiated with D-UV than in cells irradiated with F-UV. This study proved that D-UV induced apoptosis from DNA damage, whereas F-UV affected membrane integrity in monolayer cells.


Subject(s)
Apoptosis , Cell Membrane , Colonic Neoplasms , DNA Damage , Ultraviolet Rays , Humans , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/radiation effects , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Apoptosis/radiation effects , Pyrimidine Dimers/metabolism
7.
Proc Natl Acad Sci U S A ; 121(29): e2313851121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38976734

ABSTRACT

Mass spectrometry-based omics technologies are increasingly used in perturbation studies to map drug effects to biological pathways by identifying significant molecular events. Significance is influenced by fold change and variation of each molecular parameter, but also by multiple testing corrections. While the fold change is largely determined by the biological system, the variation is determined by experimental workflows. Here, it is shown that memory effects of prior subculture can influence the variation of perturbation profiles using the two colon carcinoma cell lines SW480 and HCT116. These memory effects are largely driven by differences in growth states that persist into the perturbation experiment. In SW480 cells, memory effects combined with moderate treatment effects amplify the variation in multiple omics levels, including eicosadomics, proteomics, and phosphoproteomics. With stronger treatment effects, the memory effect was less pronounced, as demonstrated in HCT116 cells. Subculture homogeneity was controlled by real-time monitoring of cell growth. Controlled homogeneous subculture resulted in a perturbation network of 321 causal conjectures based on combined proteomic and phosphoproteomic data, compared to only 58 causal conjectures without controlling subculture homogeneity in SW480 cells. Some cellular responses and regulatory events were identified that extend the mode of action of arsenic trioxide (ATO) only when accounting for these memory effects. Controlled prior subculture led to the finding of a synergistic combination treatment of ATO with the thioredoxin reductase 1 inhibitor auranofin, which may prove useful in the management of NRF2-mediated resistance mechanisms.


Subject(s)
Proteomics , Humans , Proteomics/methods , Cell Line, Tumor , HCT116 Cells , Cell Culture Techniques/methods , Colonic Neoplasms/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Arsenic Trioxide/pharmacology , Auranofin/pharmacology , Cell Proliferation/drug effects , Mass Spectrometry/methods
8.
Bull Exp Biol Med ; 177(1): 162-168, 2024 May.
Article in English | MEDLINE | ID: mdl-38960963

ABSTRACT

In adult male C57BL/6 mice with high (HR) and low (LR) resistance to hypoxia, morphological features of colon tumors and blood parameters were evaluated 70 days after intraperitoneal injection of azoxymethane and subsequent consumption of 3 cycles of dextran sulfate sodium. On macroscopic analysis, tumors were found in the distal colon in 35% (7 of 20 animals) of HR and 31% (4 of 13 animals) of LR animals. Microscopic analysis of the distal colon revealed tumors in 75% (15 of 20 animals) of HR and 69% (9 of 13 animals) of LR mice. The tumors were presented by areas of glandular intraepithelial neoplasia and adenocarcinomas; the incidence and the area of the tumors did not differ in groups of HR and LR mice. The number of neuroendocrine and goblet cells in the distal colon mucosa in the areas of tumors was similar in the compared groups. However, in both HR and LR mice of the experimental groups, the content of goblet cells in tumors was lower and the content of endocrine cells was higher than in the corresponding control groups. In the peripheral blood, the erythrocyte count and hemoglobin content decreased in HR and LR mice of the experimental groups; the relative number of monocytes increased only in HR mice and the absolute number of lymphocytes and monocytes decreased in LR mice. Thus, 70 days after azoxymethane administration and dextran sulfate sodium consumption, the tumors in mice were presented by glandular intraepithelial neoplasia and adenocarcinomas, and their incidence and area did not differ between animals with different tolerance to hypoxia.


Subject(s)
Adenocarcinoma , Azoxymethane , Colonic Neoplasms , Dextran Sulfate , Mice, Inbred C57BL , Animals , Mice , Colonic Neoplasms/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Male , Dextran Sulfate/toxicity , Azoxymethane/toxicity , Adenocarcinoma/pathology , Adenocarcinoma/chemically induced , Adenocarcinoma/metabolism , Hypoxia/pathology , Colon/pathology , Goblet Cells/pathology , Goblet Cells/metabolism , Intestinal Mucosa/pathology , Hemoglobins/metabolism , Monocytes/pathology , Monocytes/metabolism , Erythrocyte Count
9.
Biomed Mater ; 19(5)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38953496

ABSTRACT

Colon cancer (CC) is one of the most prevalent cancers in the world, and chemotherapy is widely applied to combat it. However, chemotherapy drugs have severe side effects and emergence of multi drug resistance (MDR) is common. This bottleneck can be overcome by niosome nanocarriers that minimize drug dose/toxicity meanwhile allow co-loading of incompatible drugs for combination therapy. In this research, silibinin (Sil) as a hydrophobic drug was loaded into the lipophilic part, and methotrexate (MTX) into the hydrophilic part of niosome by the thin film hydration (TFH) method to form Nio@MS NPs for CT26 colon cancer therapyin vitro. Our results indicated synthesis of ideal niosome nanoparticles (NPs) with spherical morphology, size of ∼100 nm, and a zeta potential of -10 mV. The IC50value for Nio@MS was determined ∼2.6 µg ml-1, which was significantly lower than MTX-Sil (∼6.86 µg ml-1), Sil (18.46 µg ml-1), and MTX (9.8 µg ml-1). Further, Nio@MS significantly reduced cell adhesion density, promoted apoptosis and increased gene expression level of caspase 3 and BAX while promoted significant downregulation of BCL2. In conclusion, the design and application of niosome to co-administer Sil and MTX can increase the drugs cytotoxicity, reduce their dose and improve anti-cancer potential by combating MDR.


Subject(s)
Apoptosis , Colonic Neoplasms , Methotrexate , Silybin , Methotrexate/chemistry , Methotrexate/pharmacology , Silybin/pharmacology , Silybin/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cell Line, Tumor , Apoptosis/drug effects , Nickel/chemistry , Liposomes/chemistry , Humans , Animals , Nanoparticles/chemistry , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice , Drug Carriers/chemistry
10.
Commun Biol ; 7(1): 785, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951178

ABSTRACT

Accurate, rapid and non-invasive cancer cell phenotyping is a pressing concern across the life sciences, as standard immuno-chemical imaging and omics require extended sample manipulation. Here we combine Raman micro-spectroscopy and phase tomography to achieve label-free morpho-molecular profiling of human colon cancer cells, following the adenoma, carcinoma, and metastasis disease progression, in living and unperturbed conditions. We describe how to decode and interpret quantitative chemical and co-registered morphological cell traits from Raman fingerprint spectra and refractive index tomograms. Our multimodal imaging strategy rapidly distinguishes cancer phenotypes, limiting observations to a low number of pristine cells in culture. This synergistic dataset allows us to study independent or correlated information in spectral and tomographic maps, and how it benefits cell type inference. This method is a valuable asset in biomedical research, particularly when biological material is in short supply, and it holds the potential for non-invasive monitoring of cancer progression in living organisms.


Subject(s)
Phenotype , Spectrum Analysis, Raman , Humans , Spectrum Analysis, Raman/methods , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/metabolism , Cell Line, Tumor
11.
J Gene Med ; 26(7): e3709, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949077

ABSTRACT

BACKGROUND: Colorectal cancer is the third most common malignancy worldwide and is one of the leading causes of cancer-related mortality. Ubiquitin-specific peptidase 18 (USP18) protein has been reported to exert different tumor-related effects in distinct tumor types. Here, we initially investigated the expression and signaling pathways of USP18 in colon adenocarcinoma (COAD). METHODS: A quantitative real-time PCR was conducted to evaluate the mRNA level of USP18 in cultured cells. Immunohistochemical staining was used to explore the protein expression of USP18 in clinical COAD samples. Specific knockdown was achieved by transient transfection of small interfering RNAs into SW480 and HT29 cells using Lipo3000. Cell conting kit-8 assay, transwell assay and matrigel-transwell assays were conducted to evaluate proliferation, migration and invasion capacities, respectively. Western blotting was performed to analyze downstream signaling pathways. A chi-squared test and univariate and multivariate analyses were used to evaluate the clinical data. Xenografts from mice model were assessed to validate the in vitro findings. RESULTS: Higher USP18 level was identified in COAD tissues and was positively correlated with advanced tumor stage. High USP18 protein expression indicated poorer prognosis of COAD patients. Silencing USP18 suppressed COAD cell proliferation and invasion via destabilizing extracellular signal-regulated kinase (ERK) protein and suppressing ERK downstream pathways. Simultaneously silencing interferon-stimulated gene 15 (ISG15) with USP18 can partially rescue the tumor cell viability, indicating its involvement in USP18 signaling. The oncogenic effects of USP18 were also confirmed in mice models. CONCLUSIONS: USP18 plays oncogenic effects in colon adenocarcinoma via ISG15-ERK pathways. High USP18 expression indicates poor clinical outcomes for colon adenocarcinoma patients.


Subject(s)
Adenocarcinoma , Cell Movement , Cell Proliferation , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Signal Transduction , Ubiquitin Thiolesterase , Humans , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Animals , Mice , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Male , Cell Movement/genetics , Female , Cell Line, Tumor , Disease Progression , Middle Aged , Prognosis , MAP Kinase Signaling System , Extracellular Signal-Regulated MAP Kinases/metabolism , HT29 Cells , Mice, Nude
12.
Neoplasia ; 54: 101009, 2024 08.
Article in English | MEDLINE | ID: mdl-38850836

ABSTRACT

BACKGROUND: Lipid metabolism and regulated cell death (RCD) play a role in the remodeling of tumor immune microenvironment and regulation of cancer progression. Since the underlying immune mechanisms of colon cancer remain elusive, this study aims to identify potential therapeutic target genes. METHODS: Differential genes related to lipid metabolism and RCD in COAD patients were identified using R language and online tools. Based on the expression of genes, two groups were classified using consensus clustering. CIBERSORT and ssGSEA were used to detect immune infiltration in both groups. Prognostic signature genes for colon cancer were screened using machine learning algorithms. KEGG, GO and GSEA for gene pathway enrichment. In addition, interacting genes in the immune module were obtained using a weighted gene co-expression network (WGCNA). Finally, expression and mutation of key in colon cancer genes were detected using TIMER, HPR, cBioPortal website and qPCR. RESULTS: The consensus clustering analysis revealed that 231 relevant differential genes were highly associated with immune infiltration. A series of machine learning and website analyses identified AGT as a hub gene linked to lipid metabolism and regulated cell death, which is overexpressed in colon cancer. CONCLUSION: AGT, as a signature gene of lipid metabolism and regulated cell death, plays a critical role in the development of COAD and is associated with tumor immune infiltration.


Subject(s)
Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Lipid Metabolism , Tumor Microenvironment , Humans , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Lipid Metabolism/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Prognosis , Cell Death/genetics , Biomarkers, Tumor/genetics , Computational Biology/methods , Gene Expression Profiling , Gene Regulatory Networks , Transcriptome
13.
Sci Adv ; 10(23): eadk9996, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38838152

ABSTRACT

Immunotoxicity remains a major hindrance to chemotherapy in cancer therapy. Nanocarriers may alleviate the immunotoxicity, but the optimal design remains unclear. Here, we created two variants of maytansine (DM1)-loaded synthetic high-density lipoproteins (D-sHDL) with either physically entrapped (ED-sHDL) or chemically conjugated (CD-sHDL) DM1. We found that CD-sHDL showed less accumulation in the tumor draining lymph nodes (DLNs) and femur, resulting in a lower toxicity against myeloid cells than ED-sHDL via avoiding scavenger receptor class B type 1 (SR-B1)-mediated DM1 transportation into the granulocyte-monocyte progenitors and dendritic cells. Therefore, higher densities of lymphocytes in the tumors, DLNs, and blood were recorded in mice receiving CD-sHDL, leading to a better efficacy and immune memory of CD-sHDL against colon cancer. Furthermore, liposomes with conjugated DM1 (CD-Lipo) showed lower immunotoxicity than those with entrapped drug (ED-Lipo) through the same mechanism after apolipoprotein opsonization. Our findings highlight the critical role of drug loading patterns in dictating the biological fate and activity of nanomedicine.


Subject(s)
Nanoparticles , Animals , Nanoparticles/chemistry , Mice , Cell Line, Tumor , Humans , Scavenger Receptors, Class B/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Lipoproteins, HDL/metabolism , Drug Carriers/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Liposomes/chemistry , Lipids/chemistry
14.
Mol Biol Rep ; 51(1): 732, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872006

ABSTRACT

BACKGROUND: The present study aimed to elucidate the potential anticancer activity and mechanism of P. harmala's alkaloid extract, harmine (HAR), and harmaline (HAL) in HCT-116 colorectal cancer cells. METHODS AND RESULTS: P. harmala's alkaloid was extracted from harmala seeds. HCT-116 cells were treated with P. harmala's alkaloid extract, HAR and HAL. Cytotoxicity was determined by MTT assay, apoptotic activity detected via flow cytometry and acridine orange (AO)/ethidium bromide (EB) dual staining, and cell cycle distribution analyzed with flow cytometry. The mRNA expression of Bcl-2-associated X protein (Bax) and glycogen synthase kinase-3 beta (GSK3ß) was measured by real-time PCR. Furthermore, the expression of Bax, Bcl-2, GSK3ß and p53 proteins, were determined by western blotting. The findings indicated that, P. harmala's alkaloids extract, HAR and HAL were significantly cytotoxic toward HCT116 cells after 24 and 48 h of treatment. We showed that P. harmala's alkaloid extract induce apoptosis and cell cycle arrest at G2 phase in the HCT116 cell line. Downregulation of GSK3ß and Bcl-2 and upregulation of Bax and p53 were observed. CONCLUSION: The findings of this study indicate that the P. harmala's alkaloid extract has anticancer activity and may be further investigated to develop future anticancer chemotherapeutic agents.


Subject(s)
Apoptosis , Colonic Neoplasms , Glycogen Synthase Kinase 3 beta , Harmine , Peganum , Seeds , Humans , Peganum/chemistry , HCT116 Cells , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Seeds/chemistry , Harmine/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/genetics , Plant Extracts/pharmacology , Plant Extracts/chemistry , Alkaloids/pharmacology , Harmaline/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Cell Proliferation/drug effects
15.
PLoS One ; 19(6): e0304701, 2024.
Article in English | MEDLINE | ID: mdl-38870120

ABSTRACT

This paper presents the initial exploration of the free radical scavenging capabilities of peptides derived from protein hydrolysates (PPH) obtained from Zingiber cassumunar rhizomes (Phlai). To replicate the conditions of gastrointestinal digestion, a combination of pepsin and pancreatin proteolysis was employed to generate these hydrolysates. Subsequently, the hydrolysate underwent fractionation using molecular weight cut-off membranes at 10, 5, 3, and 0.65 kDa. The fraction with a molecular weight less than 0.65 kDa exhibited the highest levels ABTS, DPPH, FRAP, and NO radical scavenging activity. Following this, RP-HPLC was used to further separate the fraction with a molecular weight less than 0.65 kDa into three sub-fractions. Among these, the F5 sub-fraction displayed the most prominent radical-scavenging properties. De novo peptide sequencing via quadrupole-time-of-flight-electron spin induction-mass spectrometry identified a pair of novel peptides: Asp-Gly-Ile-Phe-Val-Leu-Asn-Tyr (DGIFVLNY or DY-8) and Ile-Pro-Thr-Asp-Glu-Lys (IPTDEK or IK-6). Database analysis confirmed various properties, including biological activity, toxicity, hydrophilicity, solubility, and potential allergy concerns. Furthermore, when tested on the human adenocarcinoma colon (Caco-2) cell line, two synthetic peptides demonstrated cellular antioxidant activity in a concentration-dependent manner. These peptides were also assessed using the FITC Annexin V apoptosis detection kit with PI, confirming the induction of apoptosis. Notably, the DY-8 peptide induced apoptosis, upregulated mRNA levels of caspase-3, -8, and -9, and downregulated Bcl-2, as confirmed by real-time quantitative polymerase chain reaction (RT-qPCR). Western blot analysis indicated increased pro-apoptotic Bax expression and decreased anti-apoptotic Bcl-2 expression in Caco-2 cells exposed to the DY-8 peptide. Molecular docking analysis revealed that the DY-8 peptide exhibited binding affinity with Bcl-2, Bcl-xL, and Mcl-1, suggesting potential utility in combating colon cancer as functional food ingredients.


Subject(s)
Apoptosis , Colonic Neoplasms , Peptides , Rhizome , Signal Transduction , Humans , Apoptosis/drug effects , Rhizome/chemistry , Caco-2 Cells , Signal Transduction/drug effects , Peptides/pharmacology , Peptides/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Zingiberaceae/chemistry , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Free Radical Scavengers/pharmacology , Free Radical Scavengers/chemistry
16.
Pharmacol Res Perspect ; 12(4): e1226, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38886975

ABSTRACT

Although classically recognized as a neurotransmitter, gamma aminobutyric acid (GABA) has also been identified in colonic tumors. Moreover, the gut microbiome represents another potential source of GABA. Both GABAA and GABAB receptors have been implicated in contributing to the effects of GABA in colorectal cancer, with both pro- and anti-tumorigenic functions identified. However, their subunit composition is often overlooked. Studies to date have not addressed whether the GABA-producing potential of the microbiome changes over the course of colon tumor development or whether receptor subunit expression patterns are altered in colon cancer. Therefore, we investigated the clusters of orthologous group frequencies of glutamate decarboxylase (GAD) in feces from two murine models of colon cancer and found that the frequency of microbial GAD was significantly decreased early in the tumorigenic process. We also determined that microbial-derived GABA inhibited proliferation of colon cancer cells in vitro and that this effect of GABA on SW480 cells involved both GABAA and GABAB receptors. GABA also inhibited prostaglandin E2 (PGE2)-induced proliferation and interleukin-6 (IL-6) expression in these cells. Gene expression correlations were assessed using the "Cancer Exploration" suite of the TIMER2.0 web tool and identified that GABA receptor subunits were differentially expressed in human colon cancer. Moreover, GABAA receptor subunits were predominantly positively associated with PGE2 synthase, cyclooxygenase-2 and IL-6. Collectively, these data demonstrate decreased potential of the microbiome to produce GABA during tumorigenesis, a novel anti-tumorigenic pathway for GABA, and that GABA receptor subunit expression adds a further layer of complexity to GABAergic signaling in colon cancer.


Subject(s)
Cell Proliferation , Colonic Neoplasms , Gastrointestinal Microbiome , Receptors, GABA-A , Receptors, GABA-B , Signal Transduction , gamma-Aminobutyric Acid , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , gamma-Aminobutyric Acid/metabolism , Humans , Mice , Cell Line, Tumor , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-B/metabolism , Dinoprostone/metabolism , Glutamate Decarboxylase/metabolism , Interleukin-6/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Carcinogenesis , Feces/microbiology , Receptors, GABA/metabolism , Receptors, GABA/genetics , Male , Mice, Inbred C57BL , Female
17.
Phytomedicine ; 131: 155775, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838401

ABSTRACT

BACKGROUND: The cyclin-dependent kinase 4 (CDK4) interacts with its canonical and non-canonical substrates modulating the cell cycle in tumor cells. However, the potential substrates and the beyond-cell-cycle-regulated functions of CDK4 in colon cancer (CC) are still unknown. Hernandezine (HER) is previously verified to induce G0/G1 phase arrest and autophagic cell death in human cancer cells, which implies that HER might target G0/G1 phase-related proteins, including CDK4. PURPOSE: The present study tried to investigate the glycolytic metabolism and oxidative stress functions of CDK4 in colon cancer. Furthermore, the inhibitory effects and potential binding sites of HER on CDK4, as well as its anti-tumor activity were investigated in CC cells. METHODS: The mass spectrometry assay was performed to identify potential endogenous substrates of CDK4 and the correlation between glycolytic metabolic rate and CDK4 level in COAD patient tissues. Meanwhile, after inhibiting the activity or the expression of CDK4, the binding capacity of CDK4 to PKM2 and NRF2 and the latter two protein distributions in cytoplasm and nucleus were detected in CC cells. In vitro, the regulatory effects of the CDK4-PKM2-NRF2 axis on glycolysis and oxidative stress were performed by ECAR, OCR, and ROS assay. The inhibitory effect of HER on CDK4 activity was explored in CC cells and the potential binding sites were predicted and testified in vitro. Furthermore, tumor growth inhibition of HER by suppressing the CDK4-PKM2-NRF2 axis was also investigated in vitro and in vivo. RESULTS: PKM2 and NRF2 were identified as endogenous substrates of CDK4 and, high-expressed CDK4 was associated with low-level glycolysis in COAD. In vitro, inactivated CDK4 facilitated CDK4-PKM2-NRF2 complex formation which resulted in 1) inhibited PKM2 activity and retarded the glycolytic rate; 2) cytoplasm-detained NRF2 failed to transcript anti-oxidative gene expressions and induced oxidant stress. Additionally, as a CDK4 inhibitor, HER developed triple anti-tumor effects including induced G0/G1 phase arrest, suppressed glycolysis, and disrupted the anti-oxidative capacity of CC cells. CONCLUSION: The results first time revealed that CDK4 modulated glycolytic and anti-oxidative capacity of CC cells via bound to its endogenous substrates, PKM2 and NRF2. Additionally, 140Asp145Asn amino acid sites of CDK4 were potential targets of HER. HER exerts anti-tumor activity by inhibited the activity of CDK4, promoted the CDK4-PKM2-NRF2 complex formation in the CC cells.


Subject(s)
Carrier Proteins , Colonic Neoplasms , Cyclin-Dependent Kinase 4 , Membrane Proteins , NF-E2-Related Factor 2 , Thyroid Hormone-Binding Proteins , Thyroid Hormones , NF-E2-Related Factor 2/metabolism , Humans , Cyclin-Dependent Kinase 4/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Animals , Membrane Proteins/metabolism , Thyroid Hormones/metabolism , Cell Line, Tumor , Carrier Proteins/metabolism , Glycolysis/drug effects , Mice , Oxidative Stress/drug effects , Mice, Nude , Mice, Inbred BALB C , Female
18.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 206-210, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836659

ABSTRACT

We aimed to explore the role of regulating Smac expression levels in the occurrence and development of colon cancer through in vitro and in vivo experiments. Colon cancer cells HT-29 were cultured and transfected into different groups. qRT-PCR was used to detect the expression level of Smac in cells; Flow cytometry was used to detect the apoptotic ability of each group of cells; Western blot was used to detect the protein expression of Smac and apoptosis-related factors Survivin and Caspase-3; The nude mouse tumorigenesis experiment was conducted to detect the regulatory effect of regulating Smac expression levels on the growth of colon cancer transplanted tumors in vivo. In comparison to the FHC group, the HT-29 group exhibited a decrease in Smac expression. The si-Smac group, when compared with the si-NC group, showed significant reductions in Smac mRNA and protein levels, weaker cell apoptosis, increased Survivin, and decreased Caspase-3 expression. Contrarily, the oe-Smac group, against the oe-NC group, displayed increased Smac mRNA and protein levels, enhanced apoptosis, reduced Survivin, and elevated Caspase-3 expression. In nude mice tumor transplantation experiments, the LV-sh-Smac group, as opposed to the LV-sh-NC group, had tumors with greater volume and weight, reduced Smac and Caspase-3, and increased Survivin expression. In contrast, the LV-oe-Smac group, compared with the LV-oe-NC group, showed tumors with decreased volume and mass, increased expressions of Smac and Caspase-3, and decreased Survivin. Smac is lowly expressed in colon cancer. Upregulation of Smac expression can inhibit the occurrence and development of colon cancer, possibly by inhibiting Survivin expression and promoting Caspase-3 expression, thereby enhancing the pro-apoptotic function.


Subject(s)
Apoptosis Regulatory Proteins , Apoptosis , Caspase 3 , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins , Mice, Nude , Mitochondrial Proteins , Survivin , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Humans , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Survivin/metabolism , Survivin/genetics , Caspase 3/metabolism , Caspase 3/genetics , HT29 Cells , Mice , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice, Inbred BALB C , Cell Proliferation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Sci Rep ; 14(1): 13042, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38844625

ABSTRACT

Colon cancer (CC) is a highly malignant tumor with a high incidence and poor prognosis. This study aimed to explore the function and molecular mechanisms of activating transcription factor 4 (ATF4) in CC. The expression levels of ATF4, GCN2, and ASNS in CC tissues were measured using immunohistochemistry (IHC) and reverse transcription quantitative PCR (RT-qPCR). Cell counting kit-8 (CCK-8), clone formation, transwell, and flow cytometry assays were conducted to assess cell viability, clonogenicity, migration, invasion, cell cycle, and apoptosis, respectively, in the ATF4 knockdown and overexpression SW480 cell lines. The effect of ATF4 on the expression of GCN2 and ASNS was detected using RT-qPCR, Chip-qPCR, and western blotting. ATF4, GCN2, and ASNS were expressed at low levels in CC tissues, and all had a significant negative correlation with tumor diameter. ATF4 knockdown promoted cell proliferation, invasion, and S-phase cell cycle and inhibited apoptosis in SW480 cells. In contrast, ATF4 overexpression had the opposite effect. Furthermore, ATF4 overexpression enhanced ATF4 binding to the ASNS promoter region. ATF4 knockdown significantly inhibited the expression of p-GCN2 and ASNS, whereas ATF4 overexpression significantly upregulated their expression. ATF4 inhibited CC cell viability, clone formation ability, migration, and invasion and promoted apoptosis, possibly by regulating the expression of p-GCN2 and ASNS. Our study provides a novel potential therapeutic target for the treatment of CC.


Subject(s)
Activating Transcription Factor 4 , Apoptosis , Cell Movement , Cell Proliferation , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases , Up-Regulation , Humans , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Apoptosis/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cell Movement/genetics , Male , Female , Middle Aged
20.
Sci Rep ; 14(1): 13906, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886545

ABSTRACT

Colon adenocarcinoma (COAD) is the second leading cause of cancer death, and there is still a lack of diagnostic biomarkers and therapeutic targets. In this study, bioinformatics analysis of the TCGA database was used to obtain RUNX1, a gene with prognostic value in COAD. RUNX1 plays an important role in many malignancies, and its molecular regulatory mechanisms in COAD remain to be fully understood. To explore the physiological role of RUNX1, we performed functional analyses, such as CCK-8, colony formation and migration assays. In addition, we investigated the underlying mechanisms using transcriptome sequencing and chromatin immunoprecipitation assays. RUNX1 is highly expressed in COAD patients and significantly correlates with survival. Silencing of RUNX1 significantly slowed down the proliferation and migratory capacity of COAD cells. Furthermore, we demonstrate that CDC20 and MCM2 may be target genes of RUNX1, and that RUNX1 may be physically linked to the deubiquitinating enzyme USP31, which mediates the upregulation of RUNX1 protein to promote transcriptional function. Our results may provide new insights into the mechanism of action of RUNX1 in COAD and reveal potential therapeutic targets for this disease.


Subject(s)
Cdc20 Proteins , Core Binding Factor Alpha 2 Subunit , Gene Expression Regulation, Neoplastic , Minichromosome Maintenance Complex Component 2 , Ubiquitination , Humans , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Cdc20 Proteins/metabolism , Cdc20 Proteins/genetics , Minichromosome Maintenance Complex Component 2/metabolism , Minichromosome Maintenance Complex Component 2/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cell Proliferation/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics , Disease Progression , Cell Movement/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...