Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.478
Filter
1.
FASEB J ; 38(4): e23480, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38354025

ABSTRACT

Accumulating evidence suggests that dysregulation of FOXO3a plays a significant role in the progression of various malignancies, including hepatocellular carcinoma (HCC). FOXO3a inactivation, driven by oncogenic stimuli, can lead to abnormal cell growth, suppression of apoptosis, and resistance to anticancer drugs. Therefore, FOXO3a emerges as a potential molecular target for the development of innovative treatments in the era of oncology. Linagliptin (LNGTN), a DPP-4 inhibitor known for its safe profile, has exhibited noteworthy anti-inflammatory and anti-oxidative properties in previous in vivo studies. Several potential molecular mechanisms have been proposed to explain these effects. However, the capacity of LNGTN to activate FOXO3a through AMPK activation has not been investigated. In our investigation, we examined the potential repurposing of LNGTN as a hepatoprotective agent against diethylnitrosamine (DENA) intoxication. Additionally, we assessed LNGTN's impact on apoptosis and autophagy. Following a 10-week administration of DENA, the liver underwent damage marked by inflammation and early neoplastic alterations. Our study presents the first experimental evidence demonstrating that LNGTN can reinstate the aberrantly regulated FOXO3a activity by elevating the nuclear fraction of FOXO3a in comparison to the cytosolic fraction, subsequent to AMPK activation. Moreover, noteworthy inactivation of NFκB induced by LNGTN was observed. These effects culminated in the initiation of apoptosis, the activation of autophagy, and the manifestation of anti-inflammatory, antiproliferative, and antiangiogenic outcomes. These effects were concomitant with improved liver function and microstructure. In conclusion, our findings open new avenues for the development of novel therapeutic strategies targeting the AMPK/FOXO3a signaling pathway in the management of chronic liver damage.


Subject(s)
Carcinoma, Hepatocellular , Dipeptidyl-Peptidase IV Inhibitors , Liver Neoplasms , Animals , Rats , Linagliptin/pharmacology , AMP-Activated Protein Kinases , Diethylnitrosamine/toxicity , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/chemically induced , Liver Neoplasms/drug therapy , Hypoglycemic Agents , Protease Inhibitors , Antiviral Agents , Anti-Inflammatory Agents
2.
Cell ; 140(2): 197-208, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20141834

ABSTRACT

Epidemiological studies indicate that overweight and obesity are associated with increased cancer risk. To study how obesity augments cancer risk and development, we focused on hepatocellular carcinoma (HCC), the common form of liver cancer whose occurrence and progression are the most strongly affected by obesity among all cancers. We now demonstrate that either dietary or genetic obesity is a potent bona fide liver tumor promoter in mice. Obesity-promoted HCC development was dependent on enhanced production of the tumor-promoting cytokines IL-6 and TNF, which cause hepatic inflammation and activation of the oncogenic transcription factor STAT3. The chronic inflammatory response caused by obesity and enhanced production of IL-6 and TNF may also increase the risk of other cancers.


Subject(s)
Carcinoma, Hepatocellular/immunology , Interleukin-6/immunology , Liver Neoplasms/immunology , Obesity/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/etiology , Cell Proliferation , Diethylnitrosamine , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hepatitis/etiology , Hepatitis/immunology , Liver Neoplasms/chemically induced , Liver Neoplasms/etiology , Male , Mice , Obesity/complications , STAT3 Transcription Factor/metabolism
3.
Nature ; 568(7753): 557-560, 2019 04.
Article in English | MEDLINE | ID: mdl-30971822

ABSTRACT

The cell cycle is a tightly regulated process that is controlled by the conserved cyclin-dependent kinase (CDK)-cyclin protein complex1. However, control of the G0-to-G1 transition is not completely understood. Here we demonstrate that p38 MAPK gamma (p38γ) acts as a CDK-like kinase and thus cooperates with CDKs, regulating entry into the cell cycle. p38γ shares high sequence homology, inhibition sensitivity and substrate specificity with CDK family members. In mouse hepatocytes, p38γ induces proliferation after partial hepatectomy by promoting the phosphorylation of retinoblastoma tumour suppressor protein at known CDK target residues. Lack of p38γ or treatment with the p38γ inhibitor pirfenidone protects against the chemically induced formation of liver tumours. Furthermore, biopsies of human hepatocellular carcinoma show high expression of p38γ, suggesting that p38γ could be a therapeutic target in the treatment of this disease.


Subject(s)
Carcinogenesis/pathology , Cell Cycle , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Liver/enzymology , Liver/pathology , Mitogen-Activated Protein Kinase 12/metabolism , Aged , Animals , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Female , Hepatocytes/cytology , Hepatocytes/pathology , Humans , Liver/surgery , Liver Neoplasms/chemically induced , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase 12/antagonists & inhibitors , Phosphorylation , Pyridones/pharmacology , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Sequence Homology , Substrate Specificity
4.
J Biol Chem ; 299(11): 105301, 2023 11.
Article in English | MEDLINE | ID: mdl-37777158

ABSTRACT

Wilm's tumor 1-associating protein (WTAP), a regulatory protein of the m6A methyltransferase complex, has been found to play a role in regulating various physiological and pathological processes. However, the in vivo role of WTAP in the pathogenesis of hepatocellular carcinoma (HCC) is unknown. In this study, we have elucidated the crucial role of WTAP in HCC progression and shown that hepatic deletion of Wtap promotes HCC pathogenesis through activation of multiple signaling pathways. A single dose of diethylnitrosamine injection causes more and larger HCCs in hepatocyte-specific Wtap knockout (Wtap-HKO) mice than Wtapflox/flox mice fed with either normal chow diet or a high-fat diet. Elevated CD36, IGFBP1 (insulin-like growth factor-binding protein 1), and chemokine (C-C motif) ligand 2 (CCL2) expression leads to steatosis and inflammation in the Wtap-HKO livers. The hepatocyte proliferation is dramatically increased in Wtap-HKO mice, which is due to higher activation of extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription-3 signaling pathways. Hepatic deletion of Wtap activates the ERK signaling pathway by increasing the protein stability of GRB2 and ERK1/2, which is due to the decreased expression of proteasome-related genes. Restoring PSMB4 or PSMB6 (two key components of the proteasome) leads to the downregulation of GRB2 and ERK1/2 in Wtap-HKO hepatocytes. Mechanistically, WTAP interacts with RNA polymerase II and H3K9ac to maintain expression of proteasome-related genes. These results demonstrate that hepatic deletion of Wtap promotes HCC progression through activating GRB2-ERK1/2-mediated signaling pathway depending on the downregulation of proteasome-related genes especially Psmb4 and Psmb6.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocytes/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice, Knockout , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Mice, Inbred C57BL
5.
J Hepatol ; 80(1): 20-30, 2024 01.
Article in English | MEDLINE | ID: mdl-37734683

ABSTRACT

BACKGROUND & AIMS: Recent studies reported that moderate HBV DNA levels are significantly associated with hepatocellular carcinoma (HCC) risk in hepatitis B e antigen (HBeAg)-positive, non-cirrhotic patients with chronic hepatitis B (CHB). We aimed to develop and validate a new risk score to predict HCC development using baseline moderate HBV DNA levels in patients entering into HBeAg-positive CHB from chronic infection. METHODS: This multicenter cohort study recruited 3,585 HBeAg-positive, non-cirrhotic patients who started antiviral treatment with entecavir or tenofovir disoproxil fumarate at phase change into CHB from chronic infection in 23 tertiary university-affiliated hospitals of South Korea (2012-2020). A new HCC risk score (PAGED-B) was developed (training cohort, n = 2,367) based on multivariable Cox models. Internal validation using bootstrap sampling and external validation (validation cohort, n = 1,218) were performed. RESULTS: Sixty (1.7%) patients developed HCC (median follow-up, 5.4 years). In the training cohort, age, gender, platelets, diabetes and moderate HBV DNA levels (5.00-7.99 log10 IU/ml) were independently associated with HCC development; the PAGED-B score (based on these five predictors) showed a time-dependent AUROC of 0.81 for the prediction of HCC development at 5 years. In the validation cohort, the AUROC of PAGED-B was 0.85, significantly higher than for other risk scores (PAGE-B, mPAGE-B, CAMD, and REAL-B). When stratified by the PAGED-B score, the HCC risk was significantly higher in high-risk patients than in low-risk patients (sub-distribution hazard ratio = 8.43 in the training and 11.59 in the validation cohorts, all p <0.001). CONCLUSIONS: The newly established PAGED-B score may enable risk stratification for HCC at the time of transition into HBeAg-positive CHB. IMPACT AND IMPLICATIONS: In this study, we developed and validated a new risk score to predict hepatocellular carcinoma (HCC) development in patients entering into hepatitis B e antigen (HBeAg)-positive chronic hepatitis B (CHB) from chronic infection. The newly established PAGED-B score, which included baseline moderate HBV DNA levels (5-8 log10 IU/ml), improved on the predictive performance of prior risk scores. Based on a patient's age, gender, diabetic status, platelet count, and moderate DNA levels (5-8 log10 IU/ml) at the phase change into CHB from chronic infection, the PAGED-B score represents a reliable and easily available risk score to predict HCC development during the first 5 years of antiviral treatment in HBeAg-positive patients entering into CHB. With a scoring range from 0 to 12 points, the PAGED-B score significantly differentiated the 5-year HCC risk: low <7 points and high ≥7 points.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B, Chronic , Liver Neoplasms , Humans , Child, Preschool , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/chemically induced , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/drug therapy , Hepatitis B e Antigens , DNA, Viral , Liver Neoplasms/etiology , Liver Neoplasms/chemically induced , Cohort Studies , Persistent Infection , Antiviral Agents/therapeutic use , Risk Factors , Hepatitis B virus/genetics
6.
Mol Med ; 30(1): 79, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38844847

ABSTRACT

BACKGROUND: Increased level of serum cholic acid (CA) is often accompanied with decreased CYP2E1 expression in hepatocellular carcinoma (HCC) patients. However, the roles of CA and CYP2E1 in hepatocarcinogenesis have not been elucidated. This study aimed to investigate the roles and the underlying mechanisms of CYP2E1 and CA in HCC cell growth. METHODS: The proteomic analysis of liver tumors from DEN-induced male SD rats with CA administration was used to reveal the changes of protein expression in the CA treated group. The growth of CA-treated HCC cells was examined by colony formation assays. Autophagic flux was assessed with immunofluorescence and confocal microscopy. Western blot analysis was used to examine the expression of CYP2E1, mTOR, AKT, p62, and LC3II/I. A xenograft tumor model in nude mice was used to examine the role of CYP2E1 in CA-induced hepatocellular carcinogenesis. The samples from HCC patients were used to evaluate the clinical value of CYP2E1 expression. RESULTS: CA treatment significantly increased the growth of HCC cells and promoted xenograft tumors accompanied by a decrease of CYP2E1 expression. Further studies revealed that both in vitro and in vivo, upregulated CYP2E1 expression inhibited the growth of HCC cells, blocked autophagic flux, decreased AKT phosphorylation, and increased mTOR phosphorylation. CYP2E1 was involved in CA-activated autophagy through the AKT/mTOR signaling. Finally, decreased CYP2E1 expression was observed in the tumor tissues of HCC patients and its expression level in tumors was negatively correlated with the serum level of total bile acids (TBA) and gamma-glutamyltransferase (GGT). CONCLUSIONS: CYP2E1 downregulation contributes to CA-induced HCC development presumably through autophagy regulation. Thus, CYP2E1 may serve as a potential target for HCC drug development.


Subject(s)
Autophagy , Carcinoma, Hepatocellular , Cell Proliferation , Cholic Acid , Cytochrome P-450 CYP2E1 , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/chemically induced , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/chemically induced , Humans , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP2E1/genetics , Male , Autophagy/drug effects , Cell Line, Tumor , Rats , Cell Proliferation/drug effects , Mice , Rats, Sprague-Dawley , Signal Transduction , Proteomics/methods , Disease Models, Animal , Mice, Nude
7.
J Gene Med ; 26(9): e3723, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39228142

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) remains a formidable challenge in oncology, with its pathogenesis and progression influenced by myriad factors. Among them, the pervasive organic synthetic compound, bisphenol A (BPA), previously linked with various adverse health effects, has been speculated to play a role. This study endeavors to elucidate the complex interplay between BPA, the immune microenvironment of HCC, and the broader molecular landscape of this malignancy. METHODS: A comprehensive analysis was undertaken using data procured from both The Cancer Genome Atlas and the Comparative Toxicogenomics Database. Rigorous differential expression analyses were executed, supplemented by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. In addition, single-sample gene set enrichment analysis, gene set enrichment analysis and gene set variation analysis were employed to reveal potential molecular links and insights. Immune infiltration patterns were delineated, and a series of in vitro experiments on HCC cells were conducted to directly assess the impact of BPA exposure. RESULTS: Our findings unveiled a diverse array of active immune cells and functions within HCC. Distinct correlations emerged between high-immune-related scores, established markers of the tumor microenvironment and the expression of immune checkpoint genes. A significant discovery was the identification of key genes simultaneously associated with immune-related pathways and BPA exposure. Leveraging these genes, a prognostic model was crafted, offering predictive insights into HCC patient outcomes. Intriguingly, in vitro studies suggested that BPA exposure could promote proliferation in HCC cells. CONCLUSION: This research underscores the multifaceted nature of HCC's immune microenvironment and sheds light on BPA's potential modulatory effects therein. The constructed prognostic model, if validated further, could serve as a robust tool for risk stratification in HCC, potentially guiding therapeutic strategies. Furthermore, the implications of the findings for immunotherapy are profound, suggesting new avenues for enhancing treatment efficacy. As the battle against HCC continues, understanding of environmental modulators like BPA becomes increasingly pivotal.


Subject(s)
Benzhydryl Compounds , Carcinoma, Hepatocellular , Disease Progression , Gene Expression Regulation, Neoplastic , Liver Neoplasms , Phenols , Tumor Microenvironment , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Benzhydryl Compounds/adverse effects , Liver Neoplasms/immunology , Liver Neoplasms/genetics , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Phenols/adverse effects , Phenols/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , Gene Expression Profiling , Biomarkers, Tumor/genetics
8.
Histochem Cell Biol ; 161(4): 337-343, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38296878

ABSTRACT

The third most prevalent malignancy to cause mortality is hepatocellular carcinoma (HCC). The Hedgehog (Hh) signaling pathway is activated by binding to the transmembrane receptor Patched-1 (PTCH-1), which depresses the transmembrane G protein-coupled receptor Smoothened (SMO). This study was performed to examine the preventative and therapeutic effects of cannabidiol in adult rats exposed to diethyl nitrosamine (DENA)-induced HCC.A total of 50 male rats were divided into five groups of 10 rats each. Group I was the control group. Group II received intraperitoneal (IP) injections of DENA for 14 weeks. Group III included rats that received cannabidiol (CBD) orally (3-30 mg/kg) for 2 weeks and DENA injections for 14 weeks. Group IV rats received oral CBD for 2 weeks before 14 weeks of DENA injections. Group V included rats that received CBD orally for 2 weeks after their last injection of DENA. Measurements were made for alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma glutamyl transferase (GGT), superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA), and alpha fetoprotein (AFP). Following total RNA extraction, Smo, Hhip, Ptch-1, and Gli-1 expressions were measured using quantitative real-time polymerase chain reaction (qRT-PCR). A histopathological analysis of liver tissues was performed.The liver enzymes, oxidant-antioxidant state, morphological, and molecular parameters of the adult male rat model of DENA-induced HCC showed a beneficial improvement after CBD administration. In conclusion, by focusing on the Hh signaling system, administration of CBD showed a beneficial improvement in the liver enzymes, oxidant-antioxidant status, morphological, and molecular parameters in the DENA-induced HCC in adult male rats.


Subject(s)
Cannabidiol , Carcinoma, Hepatocellular , Liver Neoplasms , Rats , Male , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Hedgehog Proteins/genetics , Liver Neoplasms/chemically induced , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Cannabidiol/adverse effects , Antioxidants , Diethylnitrosamine/adverse effects , Signal Transduction , Oxidants/adverse effects , Gene Expression
9.
J Biochem Mol Toxicol ; 38(10): e23850, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39275950

ABSTRACT

Hepatocellular carcinoma (HCC) is caused by various factors including toxic substances and xenobiotics. Numerous treatment strategies are used to address toxicity to the liver and HCC, yet their adverse effects are drawbacks. This study aimed to assess the effect of DEN/CCl4 on morphological changes in the liver, body weight, tumor incidence, and hematological tumor incidence, hematological parameters, hepatic markers, and histopathological analysis in mice following a preventive measure by using ß-caryophyllene (BCP). Adult Balb/c mice were administered a single dose of DEN 1-mg/kg body weight and 0.2-mL CCl4/kg body weight intraperitoneal twice a week (i.p.) for 22 weeks. BCP was treated in one group of mice at 30-mg/kg body weight, intraperitoneal, for 7 weeks. BCP alone was treated in one group of mice at 300-mg/kg body weight intraperitoneal for 22 weeks. DEN/CCl4 caused a reduction in mice's body weight, which was significantly attenuated by BCP administration. BCP supplementation attenuated the tumor incidence DEN/CCl4 (100%) to about 25%. DEN/CCl4 caused alterations in the hematological parameters, serum total protein albumin globulin, A/G ratio, liver function markers (AST, ALT, ALP, GGT, ACP, and bilirubin), and lipid profile markers that were significantly reinstated by BCP administration. Oxidative stress markers (MDA, SOD, CAT, NO, LDH, and GST) were reduced by DEN/CCl4, which were significantly increased in BCP-treated groups. The liver histopathology alterations caused by DEN/CCl4 were amended considerably by BCP treatment. Immunohistochemical studies suggest that AFP, caspase-3, and COX-2 were chronically overexpressed in DEN/CCl4-exposed mice, notably attenuated by BCP administration. BCP suppressed tumor incidence by downregulating inflammation and inducing caspase-3-mediated apoptosis. Conclusively, BCP appears to be a potent natural supplement capable of repressing liver inflammation and carcinoma through the mitigation of oxidative stress and inflammation pathways.


Subject(s)
Carcinoma, Hepatocellular , Inflammation , Mice, Inbred BALB C , Oxidative Stress , Polycyclic Sesquiterpenes , Animals , Polycyclic Sesquiterpenes/pharmacology , Oxidative Stress/drug effects , Mice , Inflammation/metabolism , Inflammation/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/prevention & control , Carcinoma, Hepatocellular/chemically induced , Male , Down-Regulation/drug effects , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/prevention & control , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/drug therapy , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Liver Neoplasms/drug therapy , Liver/metabolism , Liver/pathology , Liver/drug effects , Carbon Tetrachloride/toxicity
10.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33468664

ABSTRACT

We have shown previously that phosphorylation of Mdm2 by ATM and c-Abl regulates Mdm2-p53 signaling and alters the effects of DNA damage in mice, including bone marrow failure and tumorigenesis induced by ionizing radiation. Here, we examine the physiological effects of Mdm2 phosphorylation by Akt, another DNA damage effector kinase. Surprisingly, Akt phosphorylation of Mdm2 does not alter the p53-mediated effects of ionizing radiation in cells or mice but regulates the p53 response to oxidative stress. Akt phosphorylation of Mdm2 serine residue 183 increases nuclear Mdm2 stability, decreases p53 levels, and prevents senescence in primary cells exposed to reactive oxidative species (ROS). Using multiple mouse models of ROS-induced cancer, we show that Mdm2 phosphorylation by Akt reduces senescence to promote KrasG12D-driven lung cancers and carcinogen-induced papilloma and hepatocellular carcinomas. Collectively, we document a unique physiologic role for Akt-Mdm2-p53 signaling in regulating cell growth and tumorigenesis in response to oxidative stress.


Subject(s)
Neoplasms, Radiation-Induced/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/radiation effects , Carcinogens/toxicity , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Cell Proliferation/genetics , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , DNA Damage/genetics , DNA Damage/radiation effects , Disease Models, Animal , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasms, Radiation-Induced/pathology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Papillomaviridae/pathogenicity , Phosphorylation/drug effects , Phosphorylation/radiation effects , Radiation, Ionizing , Reactive Oxygen Species/metabolism
11.
Ecotoxicol Environ Saf ; 271: 115952, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38218109

ABSTRACT

Cigarette smoking is one of the most impactful behavior-related risk factors for multiple cancers including hepatocellular carcinoma (HCC). Nicotine, as the principal component of tobacco, is not only responsible for smoking addiction but also a carcinogen; nevertheless, the underlying mechanisms remain unclear. Here we report that nicotine enhances HCC cancer stemness and malignant progression by upregulating the expression of GC-rich binding factor 2 (GCF2), a gene that was revealed to be upregulated in HCC and whose upregulation predicts poor prognosis, and subsequently activating the Wnt/ꞵ-catenin/SOX2 signaling pathway. We found that nicotine significantly increased GCF2 expression and that silencing of GCF2 reduced nicotine-induced cancer stemness and progression. Mechanistically, nicotine could stabilize the protein level of GCF2, and then GCF2 could robustly activate its downstream Wnt/ß-catenin signaling pathway. Taken together, our results thus suggest that GCF2 is a potential target for a therapeutic strategy against nicotine-promoted HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Nicotine/toxicity , Cell Line, Tumor , Wnt Signaling Pathway/genetics , Gene Expression Regulation, Neoplastic , Cell Proliferation
12.
Environ Toxicol ; 39(6): 3666-3678, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38506534

ABSTRACT

Liver malignancy is well recognized as a prominent health concern, with numerous treatment options available. Natural products are considered a renewable source, providing inspiring chemical moieties that could be used for cancer treatment. Suaeda vermiculata Forssk has traditionally been employed for management of hepatic conditions, including liver inflammation, and liver cirrhosis, as well as to improve general liver function. The findings of our earlier study demonstrated encouraging in vivo hepatoprotective benefits against liver injury generated by paracetamol and carbon tetrachloride. Additionally, Suaeda vermiculata Forssk exhibited cytotoxic activities in vitro against Hep-G2 cell lines and cell lines resistant to doxorubicin. The present investigation aimed to examine the potential in vivo hepatoprotective efficacy of Suaeda vermiculata Forssk extract (SVE) against hepatocellular carcinoma induced by diethylnitrosamine (DENA) in rats. The potential involvement of the PI3K/AKT/mTOR/NF-κB pathway was addressed. Sixty adult male albino rats were allocated into five groups randomly (n = 10). First group received a buffer, whereas second group received SVE only, third group received DENA only, and fourth and fifth groups received high and low doses of SVE, respectively, in the presence of DENA. Liver toxicity and tumor markers (HGFR, p-AKT, PI3K, mTOR, NF-κB, FOXO3a), apoptosis markers, and histopathological changes were analyzed. The current results demonstrated that SVE inhibited PI3K/AKT/mTOR/NF-κB pathway as well as increased expression of apoptotic parameters and FOXO3a levels, which were deteriorated by DENA treatment. Furthermore, SVE improved liver toxicity markers and histopathological changes induced by DENA administration. This study provided evidence for the conventional hepatoprotective properties attributed to SV and investigated the underlying mechanism by which its extract, SVE, could potentially serve as a novel option for hepatocellular carcinoma (HCC) treatment derived from a natural source.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Signal Transduction , Animals , Male , Rats , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Chenopodiaceae/chemistry , Diethylnitrosamine/toxicity , Forkhead Box Protein O3/metabolism , Liver/drug effects , Liver/pathology , Liver/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
13.
Drug Dev Res ; 85(4): e22198, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38764200

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the deadliest cancers. The prevention and therapy for this deadly disease remain a global medical challenge. In this study, we investigated the effect of pantoprazole (PPZ) on the carcinogenesis and growth of HCC. Both diethylnitrosamine (DEN) plus CCl4-induced and DEN plus high fat diet (HFD)-induced HCC models in mice were established. Cytokines and cell proliferation-associated gene in the liver tissues of mice and HCC cells were analyzed. Cellular glycolysis and Na+/H+ exchange activity were measured. The preventive administration of pantoprazole (PPZ) at a clinically relevant low dose markedly suppressed HCC carcinogenesis in both DEN plus CCl4-induced and HFD-induced murine HCC models, whereas the therapeutic administration of PPZ at the dose suppressed the growth of HCC. In the liver tissues of PPZ-treated mice, inflammatory cytokines, IL1, CXCL1, CXCL5, CXCL9, CXCL10, CCL2, CCL5, CCL6, CCL7, CCL20, and CCL22, were reduced. The administration of CXCL1, CXCL5, CCL2, or CCL20 all reversed PPZ-suppressed DEN plus CCL4-induced HCC carcinogenesis in mice. PPZ inhibited the expressions of CCNA2, CCNB2, CCNE2, CDC25C, CDCA5, CDK1, CDK2, TOP2A, TTK, AURKA, and BIRC5 in HCC cells. Further results showed that PPZ reduced the production of these inflammatory cytokines and the expression of these cell proliferation-associated genes through the inhibition of glycolysis and Na+/H+ exchange. In conclusion, PPZ suppresses the carcinogenesis and growth of HCC, which is related to inhibiting the production of inflammatory cytokines and the expression of cell proliferation-associated genes in the liver through the inhibition of glycolysis and Na+/H+ exchange.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Glycolysis , Liver Neoplasms , Pantoprazole , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Glycolysis/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Mice , Pantoprazole/pharmacology , Male , Cell Proliferation/drug effects , Humans , Mice, Inbred C57BL , Carcinogenesis/drug effects , Diethylnitrosamine/toxicity , Cytokines/metabolism , Cell Line, Tumor , Diet, High-Fat/adverse effects
14.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38673992

ABSTRACT

Lipopolysaccharides (LPSs) have been reported to be important factors in promoting the progression of hepatocellular carcinoma (HCC), but the corresponding molecular mechanisms remain to be elucidated. We hypothesize that epiregulin (EREG), an epidermal growth factor (EGF) family member derived from hepatic stellate cells (HSCs) and activated by LPS stimulation, is a crucial mediator of HCC progression with epidermal growth factor receptor (EGFR) expression in the tumor microenvironment. We used a mouse xenograft model of Huh7 cells mixed with half the number of LX-2 cells, with/without intraperitoneal LPS injection, to elucidate the role of EREG in LPS-induced HCC. In the mouse model, LPS administration significantly enlarged the size of xenografted tumors and elevated the expression of EREG in tumor tissues compared with those in negative controls. Moreover, CD34 immunostaining and the gene expressions of angiogenic markers by a reverse transcription polymerase chain reaction revealed higher vascularization, with increased interleukin-8 (IL-8) expression in the tumors of the mice group treated with LPS compared to those without LPS. Our data collectively suggested that EREG plays an important role in the cancer microenvironment under the influence of LPS to increase not only the tumor cell growth and migration/invasion of EGFR-positive HCC cells but also tumor neovascularization via IL-8 signaling.


Subject(s)
Carcinoma, Hepatocellular , Epiregulin , ErbB Receptors , Lipopolysaccharides , Liver Neoplasms , Signal Transduction , Tumor Microenvironment , Epiregulin/metabolism , Epiregulin/genetics , Animals , ErbB Receptors/metabolism , ErbB Receptors/genetics , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Mice , Cell Line, Tumor , Neovascularization, Pathologic/metabolism , Carcinogenesis/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Interleukin-8/metabolism , Interleukin-8/genetics , Cell Proliferation , Male , Hepatic Stellate Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects
15.
Toxicol Mech Methods ; 34(7): 750-760, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38577837

ABSTRACT

This study aimed to investigate the effects of Tarantula cubensis alcohol extract (TCAE, Theranekron) and Sorafenib (S) treatments on carcinogenesis, apoptosis and biochemical profile of rats with experimentally induced hepatocellular carcinoma (HCC). In the presented study, 58 male rats were divided into 7 groups; Negative Control (NC, n = 6), NC + TCAE (NCT, n = 6), NC + Sorafenib (NCS, n = 6), Positive Control (PC, n = 10), Positive Control + TCAE (PCT, n = 10), Positive Control + Sorafenib (PCS, n = 10), Positive Control + TCAE + Sorafenib (PCTS, n = 10). The active ingredients Diethylnitrosamine (DEN, 120 mg/kg, single dose) and Nitrosomorpholine (NMOR, 50 ppm, 21 weeks orally) were used to induce HCC in rats. At the end of the experiment, the animals were euthanized under appropriate conditions and samples were collected for biochemical and pathological investigations. In the PC group, alanine aminotransferase (ALT), aspartate aminotransferase (AST) and gamma-glutamyl transpeptidase (GGT) levels were higher (p < 0.001) and urea levels were lower (p < 0.001) compared to all other groups. Treatment groups reorganized the relevant markers (ALT, AST, GGT, and urea). A significant increase was detected in Caspase-10, Caspase-3 and Granzyme-B (GrzB) (p < 0.001) in blood and Caspase-10 and GrzB (p < 0.05) in liver tissue in PCT, PCS and PCTS groups compared to the PC group. Histopathological examination revealed that the PC group showed cancer morphology, and the treatment groups caused a decrease in tumor incidence and size. Our current findings suggest that the mechanism of action of TCAE in HCC is through the NKs/CTLs-GrzB-Casp10-Casp3 signaling pathway and can be used in combination with chemotherapy drugs for the development of future drug designs.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular , Diethylnitrosamine , Sorafenib , Animals , Sorafenib/pharmacology , Male , Apoptosis/drug effects , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Diethylnitrosamine/toxicity , Plant Extracts/pharmacology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Rats , Liver/drug effects , Liver/pathology , Liver/metabolism , Antineoplastic Agents/pharmacology , Nitrosamines/toxicity
16.
EMBO J ; 38(15): e101964, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31267557

ABSTRACT

The IGF1R signaling is important in the malignant progression of cancer. However, overexpression of IGF1R has not been properly assessed in HCC. Here, we revealed that GSTZ1-1, the enzyme in phenylalanine/tyrosine catabolism, is downregulated in HCC, and its expression was negatively correlated with IGF1R. Mechanistically, GSTZ1-1 deficiency led to succinylacetone accumulation, alkylation modification of KEAP1, and NRF2 activation, thus promoting IGF1R transcription by recruiting SP1 to its promoter. Moreover, inhibition of IGF1R or NRF2 significantly inhibited tumor-promoting effects of GSTZ1 knockout in vivo. These findings establish succinylacetone as an oncometabolite, and GSTZ1-1 as an important tumor suppressor by inhibiting NRF2/IGF1R axis in HCC. Targeting NRF2 or IGF1R may be a promising treatment approach for this subset HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Diethylnitrosamine/adverse effects , Down-Regulation , Glutathione Transferase/genetics , Heptanoates/metabolism , Liver Neoplasms/pathology , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hep G2 Cells , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , NF-E2-Related Factor 2/metabolism , Neoplasms, Experimental , Prognosis , Receptor, IGF Type 1/metabolism , Signal Transduction , Survival Analysis
17.
Mol Med ; 29(1): 156, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37946098

ABSTRACT

BACKGROUND: Krüppel-like factor 10 (KLF10) is involved in a positive feedback loop that regulates transforming growth factor ß (TGFß) signaling, and TGFß plays an important role in the pathogenesis of liver disease. Here, we investigated whether KLF10 deletion affects the development of liver fibrosis and hepatocellular carcinoma (HCC). METHODS: We induced KLF10 deletion in C57BL/6 mice. Liver fibrosis was induced by feeding a diet high in fat and sucrose (high-fat diet [HFD]), whereas HCC was produced by intraperitoneal administration of N-diethylnitrosamine (DEN). An in vitro experiment was performed to evaluate the role of KLF10 in the cancer microenvironment using Hep3B and LX2 cells. An immunohistochemical study of KLF10 expression was performed using human HCC samples from 60 patients who had undergone liver resection. RESULTS: KLF10 deletion resulted in an increased DEN-induced HCC burden with significant upregulation of SMAD2, although loss of KLF10 did not alter HFD-induced liver fibrosis. DEN-treated mice with KLF10 deletion exhibited increased levels of mesenchymal markers (N-cadherin and SNAI2) and tumor metastasis markers (matrix metalloproteinases 2 and 9). KLF10 depletion in Hep3B and LX2 cells using siRNA was associated with increased invasiveness. Compared with co-culture of KLF10-preserved Hep3B cells and KLF10-intact LX2 cells, co-culture of KLF10-preserved Hep3B cells and KLF10-depleted LX2 cells resulted in significantly enhanced invasion. Low KLF10 expression in resected human HCC specimens was associated with poor survival. CONCLUSION: The results of this study suggest that loss of KLF10 facilitates liver cancer development with alteration in TGFß signaling.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Mice, Inbred C57BL , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
18.
Anal Chem ; 95(27): 10241-10248, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37358189

ABSTRACT

Herein, we present molecular nanoparticles of ferric-tannic complexes (so called ferric-tannic nanoparticles, FT NPs) used to enhance the MRI signal in the early stage of hepatocarcinoma. FT NPs were found to accumulate in the hepatic parenchyma without tumor nodules of Wistar rats in which hepatocarcinogenicity had been induced using diethylnitrosamine (DEN). The MRI enhancement and accumulation of FT NPs were clearly observed in the early phase of hepatocarcinogenicity, which was possibly modulated by various solute carrier family members present in the entire hepatic parenchyma of the DEN-induced rats. These findings suggest that MRI with FT NPs is promising for the assessment of the early stage of hepatocarcinoma.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Rats , Animals , Liver Neoplasms/chemically induced , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/pathology , Carcinogenesis , Rats, Wistar , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/diagnostic imaging , Magnetic Resonance Imaging , Iron
19.
BMC Cancer ; 23(1): 175, 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36809998

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death, so we should be concerned and look for effective/less-harmful treatments than chemotherapeutics already clinically in application. Aspirin works well ''in conjunction'' with other therapies for HCC since aspirin can boost the sensitivity of anti-cancer activity. Vitamin C also was shown to have antitumor effects. In this study, we examined the anti-HCC activities of synergistic combination (aspirin and vitamin C) vs. doxorubicin on HCC-bearing rats and hepatocellular carcinoma (HepG-2) cells. METHODS: In vitro, we evaluated IC50 and selectivity index (SI) using HepG-2 and human lung fibroblast (WI-38) cell lines. In vivo, four rat groups were used: Normal, HCC (intraperitoneally (i.p.) administered 200 mg thioacetamide/kg/twice a week), HCC + DOXO (HCC-bearing rats i.p. administered 0.72 mg doxorubicin (DOXO)/rat/once a week), and HCC + Aspirin + Vit. C (i.p. administered vitamin C (Vit. C) 4 g/kg/day after day concomitant with aspirin 60 mg/kg/orally day after day). We evaluated biochemical factors [aminotransferases (ALT and AST), albumin, and bilirubin (TBIL) spectrophotometrically, caspase 8 (CASP8), p53, Bcl2 associated X protein (BAX), caspase 3 (CASP3), alpha-fetoprotein (AFP), cancer antigen 19.9 (CA19.9), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) using ELISA], and liver histopathologically. RESULTS: HCC induction was accompanied by significant time-dependent elevations in all measured biochemical parameters except the p53 level significantly declined. Liver tissue architecture organization appeared disturbed with cellular infiltration, trabeculae, fibrosis, and neovascularization. Following drug medication, all biochemical levels significantly reversed toward normal, with fewer signs of carcinogenicity in liver tissues. Compared to doxorubicin, aspirin & vitamin C therapy ameliorations were more appreciated. In vitro, combination therapy (aspirin & vitamin C) exhibited potent cytotoxicity (HepG-2 IC50 of 17.41 ± 1.4 µg/mL) and more excellent safety with a SI of 3.663. CONCLUSIONS: Based on our results, aspirin plus vitamin C can be considered reliable, accessible, and efficient synergistic anti-HCC medication.


Subject(s)
Ascorbic Acid , Aspirin , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Rats , Ascorbic Acid/therapeutic use , Aspirin/therapeutic use , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/therapeutic use , Liver Neoplasms/chemically induced , Liver Neoplasms/drug therapy , Thioacetamide/adverse effects , Tumor Suppressor Protein p53/metabolism , Vitamins
20.
BMC Cancer ; 23(1): 59, 2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36650455

ABSTRACT

BACKGROUND: Saroglitazar is a novel PPAR-α/γ agonist with predominant PPAR-α activity. In various preclinical models, saroglitazar has been shown to prevent & reverse symptoms of NASH. In view of these observations, and the fact that NASH is a progressive disease leading to HCC, we hypothesized that saroglitazar may prevent the development of HCC in rodents. METHODS: HCC was induced in C57BL/6 mice by a single intraperitoneal injection of 25 mg/kg diethylnitrosamine (DEN) at the age of 4 weeks and then feeding the animal a choline-deficient, L-amino acid- defined, high-fat diet (CDAHFD) for the entire study duration. Eight weeks after initiation of CDAHFD, saroglitazar (1 and 3 mg/kg) treatment was started and continued for another 27 weeks. RESULTS: Saroglitazar treatment significantly reduced the liver injury markers (serum ALT and AST), reversed hepatic steatosis and decreased the levels of pro-inflammatory cytokines like TNF-α in liver. It also resulted in a marked increase in serum adiponectin and osteopontin levels. All disease control animals showed hepatic tumors, which was absent in saroglitazar (3 mg/kg)- treatment group indicating 100% prevention of hepatic tumorigenesis. This is the first study demonstrating a potent PPARα agonist causing suppression of liver tumors in rodents, perhaps due to a strong anti-NASH activity of Saroglitazar that overrides its rodent-specific peroxisome proliferation activity. CONCLUSION: The data reveals potential of saroglitazar for chemoprevention of hepatocellular carcinoma in patients with NAFLD/NASH.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Mice , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/prevention & control , Choline , Diethylnitrosamine/toxicity , Liver Neoplasms/chemically induced , Liver Neoplasms/drug therapy , Liver Neoplasms/prevention & control , Injections, Intraperitoneal , Diet, High-Fat/adverse effects , Amino Acids , Peroxisome Proliferator-Activated Receptors , Mice, Inbred C57BL , Liver/pathology , Non-alcoholic Fatty Liver Disease/etiology , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL