Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.550
1.
Life Sci ; 348: 122680, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38697280

AIMS: Hepatocellular carcinoma (HCC) is still a leading cause of cancer-related death worldwide. But its chemotherapeutic options are far from expectation. We here compared H-ras targeted genetic therapy to a commercial docetaxel formulation (DXT) in inhibiting HCC in rats. MAIN METHODS: After the physicochemical characterization of phosphorothioate-antisense oligomer (PS-ASO) against H-ras mutated gene, the PS-ASO-mediated in vitro hemolysis, in vivo hepatic uptake, its pharmacokinetic profile, tissue distribution in some highly perfused organs, its effect in normal rats, antineoplastic efficacy in carcinogen-induced HCC in rats were evaluated and compared against DXT treatment. Mutated H-ras expression by in situ hybridization, hep-par-I, CK-7, CD-15, p53 expression patterns by immunohistochemical methods, scanning electron microscopic evaluation of hepatic architecture, various hepatic marker enzyme levels and caspase-3/9 apoptotic enzyme activities were also carried out in the experimental rats. KEY FINDINGS: PS-ASO showed low in vitro hemolysis (<3 %), and had a sustained PS-ASO blood residence time in vivo compared to DTX, with a time-dependent hepatic uptake. It showed no toxic manifestations in normal rats. PS-ASO distribution was although initially less in the lung than liver and kidney, but at 8 h it accumulated more in lung than kidney. Antineoplastic potential of PS-ASO (treated for 6 weeks) excelled in inhibiting chemically induced tumorigenesis compared to DTX in rats, by inhibiting H-ras gene expression, some immonohistochemical modulations, and inducing caspase-3/9-mediated apoptosis. It prevented HCC-mediated lung metastatic tumor in the experimental rats. SIGNIFICANCE: PS-ASO genetic therapy showed potential to inhibit HCC far more effectively than DXT in rats.


Antineoplastic Agents , Docetaxel , Genetic Therapy , Animals , Docetaxel/pharmacology , Rats , Male , Genetic Therapy/methods , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Apoptosis/drug effects , Rats, Sprague-Dawley , Taxoids/pharmacology
2.
Sci Rep ; 14(1): 8013, 2024 04 05.
Article En | MEDLINE | ID: mdl-38580754

Hepatocellular carcinoma (HCC) seriously threatens human health, mostly developed from liver fibrosis or cirrhosis. Since diethylnitrosamine (DEN) and carbon tetrachloride (CCl4)-induced HCC mouse model almost recapitulates the characteristic of HCC with fibrosis and inflammation, it is taken as an essential tool to investigate the pathogenesis of HCC. However, a comprehensive understanding of the protein expression profile of this model is little. In this study, we performed proteomic analysis of this model to elucidate its proteomic characteristics. Compared with normal liver tissues, 432 differentially expressed proteins (DEPs) were identified in tumor tissues, among which 365 were up-regulated and 67 were down-regulated. Through Gene Ontology (GO) analysis, Ingenuity Pathway Analysis (IPA), protein-protein interaction networks (PPI) analysis and Gene-set enrichment analysis (GSEA) analysis of DEPs, we identified two distinguishing features of DEN and CCl4-induced HCC mouse model in protein expression, the upregulation of actin cytoskeleton and branched-chain amino acids metabolic reprogramming. In addition, matching DEPs from the mouse model to homologous proteins in the human HCC cohort revealed that the DEN and CCl4-induced HCC mouse model was relatively similar to the subtype of HCC with poor prognosis. Finally, combining clinical information from the HCC cohort, we screened seven proteins with prognostic significance, SMAD2, PTPN1, PCNA, MTHFD1L, MBOAT7, FABP5, and AGRN. Overall, we provided proteomic data of the DEN and CCl4-induced HCC mouse model and highlighted the important proteins and pathways in it, contributing to the rational application of this model in HCC research.


Carcinoma, Hepatocellular , Liver Neoplasms, Experimental , Liver Neoplasms , Mice , Animals , Humans , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Proteomics , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Diethylnitrosamine/adverse effects , Liver Cirrhosis/pathology , Disease Models, Animal , Fatty Acid-Binding Proteins
3.
Apoptosis ; 28(7-8): 1184-1197, 2023 08.
Article En | MEDLINE | ID: mdl-37179285

This study was designed to assess the ameliorative effects of eugenol and to propose the possible mechanisms of action of eugenol in diethylnitrosamine (DENA)/acetylaminofluorene (AAF)-caused lung cancer in Wistar rats. To induce lung cancer, DENA at a dose of 150 mg/kg body weight (b.wt) for 2 weeks were intraperitoneally injected once each week and AAF was administered orally at a dose of 20 mg/kg b.wt. four times each week for the next 3 weeks. DENA/AAF-administered rats were orally supplemented with eugenol at a dose of 20 mg/kg b.wt administered once a day until 17 weeks starting from the 1st week of DENA administration. Lung histological lesions, including sheets of tumor cells, micropapillary adenocarcinoma, and apoptotic cells, resulting from the DENA/AAF dosage, were ameliorated by eugenol treatment. However, a significant drop in the levels of LPO in the lungs and a remarkable rise in GSH content and GPx and SOD activities were observed in DENA/AAF-administered rats treated with eugenol compared with those in DENA/AAF-administered controls. Moreover, in DENA/AAF-administered rats, eugenol supplementation significantly reduced TNF-α and IL-1ß levels and mRNA expression levels of NF-κB, NF-κB p65, and MCP-1 but significantly elevated the level of Nrf2. Furthermore, the DENA/AAF-administered rats treated with eugenol exhibited a significant downregulation of Bcl-2 expression levels in addition to a significant upregulation in P53 and Bax expression levels. Otherwise, the administration of DENA/AAF elevated the protein expression level of Ki-67, and this elevation was reversed by eugenol treatment. In conclusion, eugenol has effective antioxidant, anti-inflammatory, proapoptotic, and antiproliferative properties against lung cancer.


Anticarcinogenic Agents , Liver Neoplasms, Experimental , Lung Neoplasms , Rats , Animals , Rats, Wistar , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , 2-Acetylaminofluorene/adverse effects , 2-Acetylaminofluorene/metabolism , Diethylnitrosamine/toxicity , Diethylnitrosamine/metabolism , Eugenol/adverse effects , NF-kappa B/genetics , NF-kappa B/metabolism , Apoptosis , Lung Neoplasms/chemically induced , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology
4.
Int J Mol Sci ; 24(9)2023 May 07.
Article En | MEDLINE | ID: mdl-37176094

This study aimed to analyze the biochemical, histological, and gene expression alterations produced in a hepatocarcinogenesis model induced by the chronic administration of diethylnitrosamine (DEN) and 2-acetylaminofluorene (2-AAF) in Wistar rats. Thirteen rats weighing 180 to 200 g were divided into two groups: control and treated. Rats in the treated group were administered an intraperitoneal (i.p.) injection of DEN (50 mg/kg/week) and an intragastric (i.g.) dose of 2-AAF (25 mg/kg/week) for 18 weeks. The treated group had significant increases in their total cholesterol, HDL-C, AST, ALT, ALKP, and GGT levels. Furthermore, a histological analysis showed the loss of normal liver architecture with nuclear pleomorphism in the hepatocytes, atypical mitosis, and fibrous septa that were distributed between the portal triads and collagen fibers through the hepatic sinusoids. The gene expressions of 24 genes related to fibrosis, inflammation, apoptosis, cell growth, angiogenesis, lipid metabolism, and alpha-fetoprotein (AFP) were analyzed; only TGFß, COL1α1, CYP2E1, CAT, SOD, IL6, TNF-α, and ALB showed significant differences when both groups were compared. Additionally, lung histopathological alterations were found in the treated group, suggesting metastasis. In this model, the chronic administration of DEN+2-AAF induces characteristic alterations of hepatocellular carcinoma in Wistar rats without AFP gene expression changes, highlighting different signatures in hepatocellular carcinoma heterogeneity.


Carcinoma, Hepatocellular , Liver Neoplasms, Experimental , Liver Neoplasms , Rats , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Rats, Wistar , Liver/metabolism , 2-Acetylaminofluorene/toxicity , Diethylnitrosamine/toxicity , alpha-Fetoproteins , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology
5.
Sci Rep ; 12(1): 10548, 2022 06 22.
Article En | MEDLINE | ID: mdl-35732709

Our previous study demonstrated that purple rice bran extract (PRBE) could inhibit diethylnitrosamine (DEN)-induced hepatocarcinogenesis. Protocatechuic acid (PCA) is the major phenolic acid contained in the PRBE. Therefore, this study aimed to determine whether PCA is an anticarcinogenic compound in purple rice extract. Rats were intraperitoneally injected with DEN to induce glutathione S-transferase placental form (GST-P)-positive foci. Rats were fed with PRBE at 500 mg kg-1 body weight or PCA at 4 mg kg-1 body weight for 5 and 15 weeks. PCA administration attenuated DEN-induced hepatic GST-P positive foci to a degree similar to PRBE. The molecular mechanisms of PCA in the initiation stage were correlated with reduced activity of cytochrome P450 reductase and induction of glutathione S-transferase. In addition, PCA also downregulated the expression of TNF-α and IL-1ß genes in rat liver. These genes are associated with the inhibition of inflammation. In the promotion stage, PCA suppressed cell proliferation correlated with the downregulation of Cyclin D1 expression. Moreover, it also induced apoptosis, indicated by increased expression of P53 and Bad genes, and decreased the expression of the anti-apoptotic Bcl-xl in DEN-initiated rats. These findings suggest that PCA is an active compound in the anticarcinogenic action of purple rice bran.


Anticarcinogenic Agents , Liver Neoplasms, Experimental , Oryza , Animals , Anticarcinogenic Agents/pharmacology , Body Weight , Carcinogenesis/metabolism , Diethylnitrosamine/toxicity , Female , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Hydroxybenzoates , Liver/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/prevention & control , Oryza/metabolism , Placenta/metabolism , Plant Extracts/pharmacology , Pregnancy , Rats
6.
Sci Total Environ ; 837: 155685, 2022 Sep 01.
Article En | MEDLINE | ID: mdl-35523338

Developmental arsenic exposure increases cancer risk in later life with the mechanism elusive. Oxidative stress is a dominant determinant in arsenic toxicity. However, the role of Nrf2, a key regulator in antioxidative response, in tumor-augmenting effects by developmental arsenic exposure is unclear. In the present study, wild-type C57BL/6J and Nrf2-konckout (Nrf2-KO) were developmentally exposed to inorganic arsenic via drinking water. For hepatic tumorigenesis analysis, mice were intraperitoneally injected with diethylnitrosamine (DEN) at two weeks of age. Developmental arsenic exposure aggravated tumor multiplicity and burden, and expression of PCNA and AFP in hepatic tumors induced by DEN. Nrf2 activation as indicated by over-expression of Nrf2 and its downstream genes, including Gss, Gsr, p62, Gclc and Gclm, was found in liver tumors, as well as in the livers in developmentally arsenic-exposed pups at weaning. Notably, Nrf2 deficiency attenuated tumor-augmenting effects and over-expression of Nrf2 downstream genes due to developmental arsenic exposure. Furthermore, the levels of urinary DEN metabolite (acetaldehyde) and hepatic DNA damage markers (O6-ethyl-2-deoxyguanosine adducts and γ-histone H2AX) after DEN treatment were elevated by Nrf2 agonist, 2-Cyano-3,12-dioxooleana-1,9-dien-28-imidazolide. Collectively, our data suggest that augmentation of DEN-induced hepatic tumorigenesis by developmental arsenic exposure is dependent on Nrf2 activation, which may be related to the role of Nrf2 in DEN metabolic activation. Our findings reveal, at least in part, the mechanism underlying increased susceptibility to developing cancer due to developmental arsenic exposure.


Arsenic , Liver Neoplasms, Experimental , NF-E2-Related Factor 2 , Animals , Arsenic/toxicity , Carcinogenesis/chemically induced , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Oxidative Stress
7.
Life Sci ; 294: 120369, 2022 Apr 01.
Article En | MEDLINE | ID: mdl-35120919

AIMS: Hepatocellular carcinoma (HCC) is considered one of the main causes of cancer-related death globally. Combination therapy targeting different pathways can improve the efficacy of HCC management. Mitofusin 2 (Mfn2), a mitochondrial fusion protein, and a tissue inhibitor of matrix metalloproteinase 3 (Timp-3) were found to be downregulated in various cancers, including HCC. Our study aimed to evaluate the possible antineoplastic effect of a novel combination in the treatment of HCC through targeting mitochondrial fusion and metastatic proteins. MAIN METHODS: HCC induction was performed using a single intraperitoneal dose of diethylnitrosamine (200 mg/kg), followed by adding phenobarbital sodium (0.05%) to the drinking water for successive 18 weeks. Then, leflunomide (LF, 10 mg/kg) was administered orally for 28 days. Diallyl disulfide (DADS, 50 mg/kg) was also given orally for 28 days, either alone or in combination with LF. KEY FINDINGS: Treatment with LF or DADS could alleviate the HCC- induced histological and biochemical variations, including liver enzyme activities (ALT, AST), alpha-fetoprotein, Bax, cyclin D1, Ki67, malondialdehyde, and reduced glutathione. They could shift the mitochondrial dynamics toward mitochondrial fusion through upregulating the expression of Mfn2 and also exhibited antimetastatic activity through upregulating the expression of Timp-3 and decreasing hepatic MMP9 content. SIGNIFICANCE: the treatment with a combination of LF and DADS displayed a more potent effect than the treatment with each drug alone. Our results suggest that the combined use of LF and a naturally occurring DADS can be used as a promising novel combination in managing HCC.


Allyl Compounds/pharmacology , Carcinoma, Hepatocellular/prevention & control , Disulfides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Leflunomide/pharmacology , Liver Neoplasms, Experimental/prevention & control , Mitochondrial Dynamics/drug effects , Tissue Inhibitor of Metalloproteinase-3/metabolism , Alkylating Agents/toxicity , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Diethylnitrosamine/toxicity , Drug Therapy, Combination , Immunosuppressive Agents/pharmacology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Rats , Rats, Wistar , Tissue Inhibitor of Metalloproteinase-3/genetics
8.
Life Sci ; 289: 120192, 2022 Jan 15.
Article En | MEDLINE | ID: mdl-34871664

AIMS: The number of cancer survivors with cardiovascular disease is increasing. However, the effect of cancer on body fluid regulation remains to be clarified. In this study, we evaluated body osmolyte and water imbalance in rats with hepatocellular carcinoma. MAIN METHODS: Wistar rats were administered diethylnitrosamine, a carcinogenic drug, to establish liver cancer. We analyzed tissue osmolyte and water content, and their associations with aldosterone secretion. KEY FINDINGS: Hepatocellular carcinoma rats had significantly reduced body mass and the amount of total body sodium, potassium, and water. However, these rats had significantly increased relative tissue sodium, potassium, and water content per tissue dry weight. Furthermore, these changes in sodium and water balance in hepatocellular carcinoma rats were significantly associated with increased 24-h urinary aldosterone excretion. Supplementation with 0.25% salt in drinking water improved body weight reduction associated with sodium and water retention in hepatocellular carcinoma rats, which was suppressed by treatment with spironolactone, a mineralocorticoid receptor antagonist. Additionally, the urea-driven water conservation system was activated in hepatocellular carcinoma rats. SIGNIFICANCE: These findings suggest that hepatocellular carcinoma induces body mass loss in parallel with activation of the water conservation system including aldosterone secretion and urea accumulation to retain osmolyte and water. The osmolyte and water retention at the tissue level may be a causative factor for ascites and edema formation in liver failure rats.


Aldosterone/urine , Carcinoma, Hepatocellular/urine , Diethylnitrosamine/toxicity , Liver Neoplasms, Experimental/urine , Water-Electrolyte Balance , Weight Loss , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/drug therapy , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Rats , Rats, Inbred WKY , Receptors, Mineralocorticoid/metabolism , Spironolactone/pharmacology
9.
Mol Med Rep ; 25(2)2022 02.
Article En | MEDLINE | ID: mdl-34913065

Hepatocellular carcinoma is a malignancy with poor clinical prognosis. Hepatic oval cells (HOCs) tend to differentiate into cancerous hepatocellular carcinoma cells (HCCs) in the tumor microenvironment. The purpose of the present study was to explore the role of kangxianruangan granule (KXRG)­containing serum in inhibiting the differentiation of HOCs into HCCs via the Wnt­1/ß­catenin signaling pathway. N­methyl­N'­nitro­N­nitrosoguanidine (MNNG) was applied to induce the transformation of the rat HOC cell line WB­F344 into HCCs. The overexpression plasmid, Wnt­1­up, was utilized to increase Wnt­1 expression. Subsequently, high, medium and low concentrations of KXRG were applied to MNNG­treated WB­F344 cells to assess the inhibitory effect of KXRG on cell differentiation. Flow cytometry was conducted to detect the cell cycle distribution, apoptotic rate and expression of cytokeratin­19 (CK­19) protein in cells. An immunofluorescence double staining protocol was used to detect the expression of Wnt­1 and ß­catenin. ELISAs were performed to detect α fetoprotein in the cell supernatants. Reverse transcription­quantitative PCR and western blotting were conducted to detect the mRNA and protein expression levels of Wnt­1, ß­catenin, Cyclin D1, C­myc, matrix metalloproteinase­7 (MMP­7), Axin2 and epithelial cell adhesion molecule (EpCAM) in cells. Compared with the normal group, the apoptotic rate, proportion of S phase cells, concentration of AFP in the cell supernatant, level of CK­19 protein, and mRNA and protein expression levels of Wnt­1, ß­catenin, Cyclin D1, C­myc, MMP­7, Axin2 and EpCAM were all significantly increased in the model group. Addition of KXRG significantly reduced the aforementioned indicators compared with the model group. Moreover, Wnt­1 overexpression further increased the aforementioned indicators compared with the model group, whereas KXRG significantly inhibited these effects. The results indicated that KXRG inhibited the differentiation of HOCs into HCCs via the Wnt­1/ß­catenin signaling pathway, which suggested the potential clinical application of KXRG for the prevention of hepatocellular carcinoma.


Carcinoma, Hepatocellular/prevention & control , Cell Transformation, Neoplastic/drug effects , Drugs, Chinese Herbal/administration & dosage , Liver Neoplasms, Experimental/prevention & control , Wnt Signaling Pathway/drug effects , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Humans , Liver/cytology , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Methylnitronitrosoguanidine/toxicity , Rats , Tumor Microenvironment/drug effects
10.
Appl Biochem Biotechnol ; 194(1): 368-381, 2022 Jan.
Article En | MEDLINE | ID: mdl-34792748

Biosynthesized silver nanoparticles have a wide range of biological activities and using nanoparticles as one of the novel approaches in cancer therapy. In this present research work, the anti-cancer efficacy of Cucumis melo fruit extract and its silver nanoparticles was explored. Wistar rats were divided into six groups and hepatic cancer was induced with 0.01% DEN (diethylnitrosamine) through drinking water for 16 weeks. Cyclophosphamide was given as the standard drug at the dose of 50 mg/kg body weight. Hematological parameters showed a decrease in the levels of hemoglobin (Hb), packed cell volume (PCV), red blood cells (RBC), mean corpuscular volume (MCV), mean corpuscular Hb (MCH), mean corpuscular Hb concentration (MCHC), and platelets (PLTS) levels except white blood cell (WBC) in DEN-induced cancer animals. Significant alterations in the hematological parameters were observed after treatment which indicate the protective effect of Cucumis melo fruit on the hemopoietic system. The structural integrity of the cells has been damaged in cancer-induced animals, and this results in cytoplasmic leakage of enzyme into the blood stream, leads to the elevated levels of these enzymes in blood with subsequent fall in the tissues. Hence, the levels of liver function markers such as AST ALT, ALP, LDH, GGT, and 5'NT were significantly elevated in serum and the liver of cancer-induced rats. The levels of serum tumor markers, viz., alpha-fetoprotein (AFP) and carcinoembryonic antigen (CEA), elevated in rats induced with DEN, which then were reduced following Cucumis melo fruit treatment, indicating the anti-cancer activity of the drug. Histological evaluation of the liver and kidney was also performed to authenticate the present work. Treatment with crude extract and silver nanoparticles of Cucumis melo fruit indicates that Cucumis melo fruit could have exerted its protective effect.


Carcinoma, Hepatocellular , Cucumis melo/chemistry , Diethylnitrosamine/toxicity , Fruit/chemistry , Liver Neoplasms, Experimental , Metal Nanoparticles , Plant Extracts/chemistry , Silver , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Rats , Rats, Wistar , Silver/chemistry , Silver/pharmacology
12.
Nat Commun ; 12(1): 5725, 2021 09 30.
Article En | MEDLINE | ID: mdl-34593796

Although psycho-social stress is a well-known factor that contributes to the development of cancer, it remains largely unclear whether and how environmental eustress influences malignant diseases and regulates cancer-related therapeutic responses. Using an established eustress model, we demonstrate that mice living in an enriched environment (EE) are protected from carcinogen-induced liver neoplasia and transplantable syngeneic liver tumors, owning to a CD8+ T cell-dependent tumor control. We identify a peripheral Neuro-Endocrine-Immune pathway in eustress, including Sympathetic nervous system (SNS)/ß-adrenergic receptors (ß-ARs)/CCL2 that relieves tumor immunosuppression and overcomes PD-L1 resistance to immunotherapy. Notably, EE activates peripheral SNS and ß-ARs signaling in tumor cells and tumor infiltrated myeloid cells, leading to suppression of CCL2 expression and activation of anti-tumor immunity. Either blockade of CCL2/CCR2 or ß-AR signaling in EE mice lose the tumor protection capability. Our study reveales that environmental eustress via EE stimulates anti-tumor immunity, resulting in more efficient tumor control and a better outcome of immunotherapy.


Drug Resistance, Neoplasm/immunology , Immune Checkpoint Inhibitors/pharmacology , Liver Neoplasms, Experimental/drug therapy , Neuroimmunomodulation , Stress, Psychological/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/metabolism , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Hepatic Stellate Cells , Hepatocytes , Humans , Immune Checkpoint Inhibitors/therapeutic use , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Male , Mice , Organoids , Receptors, Adrenergic, beta/metabolism , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Sympathetic Nervous System/immunology , Tumor Escape , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
13.
J Cancer Res Ther ; 17(4): 1081-1092, 2021.
Article En | MEDLINE | ID: mdl-34528568

BACKGROUND AND AIM: Hepatocellular carcinoma (HCC), a primary liver malignancy, represents a continuous challenge to clinicians as it is a leading cause of death due to cancer widely. Early detection is the only hope to cure patients from this deadly disease or possibly increase life expectancy. Mouse models are most acceptable studies as they have ability to manipulate their genome and transcriptome to evaluate mechanistic changes. In addition, system biology can improvise the understanding of molecular mechanism of HCC and also can reveal the protein hub involved in every stage of HCC. MATERIALS AND METHODS: Herein, diethylnitrosamine and thioacetamide (TAA) were used to develop stage-specific HCC in Wistar rats. Histopathological changes, biochemical parameters, and the oxidative stress were measured in hepatocytes. We have reanalyzed the microarray dataset to identify the complex signaling pathways involved in hepatocarcinogenesis induced by TAA. GSE45050 dataset was downloaded from Gene Expression Omnibus database, and the gene expression profile of nontumor, cirrhosis, and HCC was compared. RESULTS: The study reveals stage-specific development of chronic HCC rat model and promising stage-specific targets (EHMT2, GMPS, and SPRY2) of HCC. CONCLUSIONS: EHMT2, GMPS, and SPRY found as high centrality nodes in protein-protein interaction studies using high-throughput microarray data which tend to be present in signaling pathways and co-occur in a biological state of HCC. These genes can be targeted to understand the possible pathology, molecular changes, and target strategy under cirrhosis and HCC condition.


Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Diethylnitrosamine/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms, Experimental/pathology , Thioacetamide/toxicity , Transcriptome , Alkylating Agents , Animals , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Gene Expression Profiling , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Male , Protein Interaction Maps , Rats , Rats, Wistar
14.
Life Sci ; 285: 119961, 2021 Nov 15.
Article En | MEDLINE | ID: mdl-34536497

AIM: Hepatocellular carcinoma (HCC) is a potentially life-threatening cancer. In the current study, anti-HCC efficacy of amygdalin, or metformin alone or in combination in comparison to doxorubicin was studied. MAIN METHODS: Both in-vitro and in-vivo based models. HepG-2 and Huh-7 cell lines as established in-vitro model for HCC were treated with different concentrations of indicated drugs to evaluate the cytotoxicity and determine IC50 for 24, 48 and 72 h. Moreover, the effect of different treatments on apoptosis and cell cycle using flow cytometric analysis were studied. Hepatocellular carcinoma induced in rats by diethyl-nitrosamine and carbon tetrachloride was established, to further investigate the efficacy of indicated drugs. Aspartate aminotransferase, alanine aminotransferase and alkaline phosphatase were measured by spectrophotometer, alpha-fetoprotein, cytochrome-c, caspase-3 and malondialdehyde were measured by ELISA, and liver biopsies were also evaluated histopathologically. KEY FINDINGS: In-vitro results showed that the combination has a promising effect when compared to amygdalin or metformin alone as it is more cytotoxic and have higher ability for induction of apoptosis and arresting cell cycle. In-vivo doxorubicin has a good effect for treating HCC. Also, the combination showed a promising prognostic effect depending on the cytotoxic activity and tumor marker when compared to amygdalin or metformin alone. SIGNIFICANCE: Based on the current data, it was hypothesized that amygdalin and metformin especially when used in combination will be a promising approach with low side effects for enhancement of HCC.


Amygdalin/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Protocols , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/therapeutic use , Liver Neoplasms/drug therapy , Metformin/therapeutic use , Animals , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/pathology , Diethylnitrosamine , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Male , Prognosis , Rats , Rats, Wistar , alpha-Fetoproteins/analysis
15.
Curr Protoc ; 1(8): e211, 2021 Aug.
Article En | MEDLINE | ID: mdl-34370903

Human hepatocellular carcinoma (HCC) develops most often as a complication of fibrosis or cirrhosis. Although most human studies of HCC provide crucial insights into the molecular signatures of HCC, they seldom address its etiology. Mouse models provide essential tools for investigating the pathogenesis of HCC, but the majority of rodent cancer models do not feature liver fibrosis. Detailed here is a protocol for an experimental mouse model of HCC that arises in association with advanced liver fibrosis. The disease model is induced by a single injection of N-nitrosodiethylamine (DEN) at 2 weeks of age followed by repeated administration of carbon tetrachloride (CCl4 ) from 8 weeks of age for up to 14 consecutive weeks. A dramatic potentiation of liver tumor incidence is observed following administration of DEN and CCl4 , with 100% of mice developing liver tumors at 5 months of age. This model has been employed for studying the molecular mechanisms of fibrogenesis and HCC development, as well as for cancer hazard/chemotherapy testing of drug candidates. © 2021 Wiley Periodicals LLC. Basic Protocol: The DEN and CCl4 -induced mouse model of fibrosis and inflammation-associated hepatocellular carcinoma.


Carcinoma, Hepatocellular , Liver Neoplasms, Experimental , Liver Neoplasms , Animals , Humans , Inflammation , Liver Cirrhosis/chemically induced , Liver Neoplasms, Experimental/chemically induced , Mice
16.
Nucleic Acids Res ; 49(17): 9836-9850, 2021 09 27.
Article En | MEDLINE | ID: mdl-34428289

Multiple pathways mediate the repair of DNA double-strand breaks (DSBs), with numerous mechanisms responsible for driving choice between the pathways. Previously, we reported that mutating five putative phosphorylation sites on the non-homologous end joining (NHEJ) factor, Ku70, results in sustained retention of human Ku70/80 at DSB ends and attenuation of DSB repair via homologous recombination (HR). In this study, we generated a knock-in mouse, in which the three conserved putative phosphorylation sites of Ku70 were mutated to alanine to ablate potential phosphorylation (Ku703A/3A), in order to examine if disrupting DSB repair pathway choice by modulating Ku70/80 dynamics at DSB ends results in enhanced genomic instability and tumorigenesis. The Ku703A/3A mice developed spontaneous and have accelerated chemical-induced hepatocellular carcinoma (HCC) compared to wild-type (Ku70+/+) littermates. The HCC tumors from the Ku703A/3A mice have increased γH2AX and 8-oxo-G staining, suggesting decreased DNA repair. Spontaneous transformed cell lines from Ku703A/3A mice are more radiosensitive, have a significant decrease in DNA end resection, and are more sensitive to the DNA cross-linking agent mitomycin C compared to cells from Ku70+/+ littermates. Collectively, these findings demonstrate that mutating the putative Ku70 phosphorylation sites results in defective DNA damage repair and disruption of this process drives genomic instability and accelerated development of HCC.


Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Liver Neoplasms, Experimental/genetics , Recombinational DNA Repair , Animals , Cells, Cultured , Female , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Mutation , Phosphorylation , Radiation Tolerance
17.
Arch Toxicol ; 95(9): 3117-3131, 2021 09.
Article En | MEDLINE | ID: mdl-34269859

1,3-Dichloro-2-propanol (1,3-DCP), a food contaminant, exerts carcinogenic effects in multiple organs, including the liver and kidneys, in rats. However, the underlying mechanisms of 1,3-DCP-induced carcinogenesis remain unclear. Here, the in vivo mutagenicity and tumor-promoting activity of 1,3-DCP in the liver and kidneys were evaluated using medium-term gpt delta rat models previously established in our laboratory (GPG and GNP models). Six-week-old male F344 gpt delta rats were treated with 0 or 50 mg/kg body weight/day 1,3-DCP by gavage for 4 weeks. After 2 weeks of cessation, partial hepatectomy or unilateral nephrectomy was performed to collect samples for in vivo mutation assays, followed by single administration of diethylnitrosamine (DEN) for tumor initiation. One week after DEN injection, 1,3-DCP treatment was resumed, and tumor-promoting activity was evaluated in the residual liver or kidneys by histopathological analysis of preneoplastic lesions. gpt mutant frequencies increased in excised liver and kidney tissues following 1,3-DCP treatment. 1,3-DCP did not affect the development of glutathione S-transferase placental form-positive foci in residual liver tissues, but enhanced atypical tubule hyperplasia in residual kidney tissues. Detailed histopathological analyses revealed glomerular injury and increased cell proliferation of renal tubular cells in residual kidney tissues of rats treated with 1,3-DCP. These results suggested possible involvement of genotoxic mechanisms in 1,3-DCP-induced carcinogenesis in the liver and kidneys. In addition, we found that 1,3-DCP exhibited limited tumor-promoting activity in the liver, but enhanced clonal expansion in renal carcinogenesis via proliferation of renal tubular cells following glomerular injury.


Kidney/drug effects , Liver/drug effects , Mutagens/toxicity , alpha-Chlorohydrin/analogs & derivatives , Animals , Carcinogenesis/drug effects , Carcinogens/toxicity , Cell Proliferation/drug effects , Escherichia coli Proteins/genetics , Kidney/pathology , Kidney Neoplasms/chemically induced , Kidney Neoplasms/pathology , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Mutagenesis/drug effects , Mutagenicity Tests , Pentosyltransferases/genetics , Rats , Rats, Inbred F344 , Rats, Transgenic , alpha-Chlorohydrin/toxicity
18.
J Basic Clin Physiol Pharmacol ; 32(4): 505-510, 2021 Jun 25.
Article En | MEDLINE | ID: mdl-34214328

OBJECTIVES: For designing early treatment for liver cancer, it is important to prepare an animal model to evaluate cancer prevention treatment by using inflammation disease. The hepatocarcinogenic N-Nitrosodiethylamine (NDEA) has been reportedly able to produce free radicals that cause liver inflammation leading to liver carcinoma. This study aimed to evaluate the inflammation disease model of mice induced with hepatocarcinogenic NDEA for five weeks induction. METHODS: The BALB-c mice were induced with NDEA 25 mg/kg of body weight once a week for five weeks intraperitonially and it was then evaluated for the body weight during study periods. The mice were then sacrificed and excised for evaluating their organs including physical and morphological appearances and histopathology evaluations. RESULTS: The results showed a significant decrease of body weight of mice after five times induction of 25 mg NDEA/kgBW per week intraperitonially. Different morphological appearances and weight of mice organs specifically for liver and spleen had also been observed. The histopathology examination showed that there were hepatic lipidosis and steatohepatitis observed in liver and spleen, respectively that might indicate the hepatocellular injury. CONCLUSIONS: It can be concluded that inducing mice with NDEA intraperitonially resulted in fatty liver disease leading to progress of cancer disease.


Diethylnitrosamine , Liver Neoplasms, Experimental , Animals , Body Weight , Diethylnitrosamine/toxicity , Inflammation/chemically induced , Liver , Liver Neoplasms, Experimental/chemically induced , Mice , Mice, Inbred BALB C
19.
Hepatology ; 74(6): 3037-3055, 2021 12.
Article En | MEDLINE | ID: mdl-34292642

BACKGROUND AND AIMS: Insulin receptor (IR) transduces cell surface signal through phosphoinositide 3-kinase (PI3K)-AKT pathways or translocates to the nucleus and binds to the promoters to regulate genes associated with insulin actions, including de novo lipogenesis (DNL). Chronic activation of IR signaling drives malignant transformation, but the underlying mechanisms remain poorly defined. Down-regulation of fructose-1,6-bisphosphate aldolase (ALDO) B in hepatocellular carcinoma (HCC) is correlated with poor prognosis. We aim to study whether and how ALDOB is involved in IR signaling in HCC. APPROACH AND RESULTS: Global or liver-specific ALDOB knockout (L-ALDOB-/- ) mice were used in N-diethylnitrosamine (DEN)-induced HCC models, whereas restoration of ALDOB expression was achieved in L-ALDOB-/- mice by adeno-associated virus (AAV). 13 C6 -glucose was employed in metabolic flux analysis to track the de novo fatty acid synthesis from glucose, and nontargeted lipidomics and targeted fatty acid analysis using mass spectrometry were performed. We found that ALDOB physically interacts with IR and attenuates IR signaling through down-regulating PI3K-AKT pathways and suppressing IR nuclear translocation. ALDOB depletion or disruption of IR/ALDOB interaction in ALDOB mutants promotes DNL and tumorigenesis, which is significantly attenuated with ALDOB restoration in L-ALDOB-/- mice. Notably, attenuated IR/ALDOB interaction in ALDOB-R46A mutant exhibits more significant tumorigenesis than releasing ALDOB/AKT interaction in ALDOB-R43A, whereas knockdown IR sufficiently diminishes tumor-promoting effects in both mutants. Furthermore, inhibiting phosphorylated AKT or fatty acid synthase significantly attenuates HCC in L-ALDOB-/- mice. Consistently, ALDOB down-regulation is correlated with up-regulation of IR signaling and DNL in human HCC tumor tissues. CONCLUSIONS: Our study reports a mechanism by which loss of ALDOB activates IR signaling primarily through releasing IR/ALDOB interaction to promote DNL and HCC, highlighting a potential therapeutic strategy in HCC.


Carcinogenesis/genetics , Fructose-Bisphosphate Aldolase/metabolism , Lipogenesis/genetics , Liver Neoplasms, Experimental/genetics , Receptor, Insulin/metabolism , Animals , Carcinogenesis/chemically induced , Carcinogenesis/pathology , Cell Line, Tumor , Diethylnitrosamine/administration & dosage , Down-Regulation , Fatty Acids/biosynthesis , Fructose-Bisphosphate Aldolase/genetics , Gene Expression Regulation, Neoplastic , Lipidomics , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice, Knockout , Phosphorylation
20.
Molecules ; 26(14)2021 Jul 12.
Article En | MEDLINE | ID: mdl-34299510

Cratoxylum formosum ssp. pruniflorum (Kurz) Gogelein (CP) is an indigenous plant found mainly in southeast Asia. Several in vitro studies have confirmed its activity against hepatocellular carcinoma; however, in vivo studies of the effect of CP on liver cancer are needed. This study investigated the effect of CP on early-stage hepatocarcinogenesis in rat liver when using diethylnitrosamine (DEN) as a carcinogen. Immunohistochemistry was used to detect (a) upregulation of glutathione S-transferase placental (GST-P) positive foci, (b) the proliferating cell nuclear antigen PCNA, and (c) apoptotic cells in the liver as indicators of early-stage carcinogenesis. Immunohistochemical parameters were observed in rats given CP orally following DEN injection. Rats given DEN presented overexpression of GST-P positive foci, PCNA, and apoptotic cells, indicating the formation of cancerous tissues, and these effects were diminished by CP treatment. CP thus inhibited hepatocarcinogenic effects in an animal model. These results could help plan further in vivo studies and support the use of CP to prevent processes that promote the pathogenesis of hepatocellular carcinoma in humans.


Anticarcinogenic Agents/pharmacology , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/drug therapy , Clusiaceae/chemistry , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms/drug therapy , Liver/drug effects , Animals , Carcinogens/pharmacology , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Diethylnitrosamine/pharmacology , Glutathione Transferase/metabolism , Liver/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Male , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Wistar
...