Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
Yale J Biol Med ; 96(4): 443-454, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38161580

ABSTRACT

Identifying new hepatocellular carcinoma (HCC)-driven signaling molecules and discovering their molecular mechanisms are crucial for efficient and better outcomes. Recently, OMA1 and YME1L, the inner mitochondrial proteases, were displayed to be associated with tumor progression in various cancers; however, their role in HCC has not yet been studied. Therefore, we evaluated the possible role of OMA1/YME1L in HCC staging and discussed their potential role in cellular apoptosis and proliferation. Our study was performed using four groups of male albino rats: a normal control and three diethyl nitrosamine-treated groups for 8, 16, and 24 weeks. The OMA1 and YME1L, matrix-metalloproteinase-9 (MMP-9), and cyclin D1 content were measured in liver tissues, while alpha-fetoprotein (AFP) level was assessed in serum. Additionally, Ki-67 expression was evaluated by immunohistochemistry. The relative hepatic expression of Bax, and tissue inhibitor matrix metalloproteinase (TIMP-3) was measured. Herein, we confirmed for the first time that OMA1 is down-regulated while YME1L is up-regulated in HCC in the three studied stages with subsequent inhibition of apoptosis and cell cycle progression. Furthermore, these proteases have a possible role in metastasis. These newly recognized results suggested OMA1 and YME1L as possible diagnostic tools and therapeutic targets for HCC management.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Biomarkers, Tumor , Carcinoma, Hepatocellular , Liver Neoplasms , Metalloproteases , Mitochondrial Proteins , Male , Animals , Rats , Diethylnitrosamine/administration & dosage , Metalloproteases/blood , Mitochondrial Proteins/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/blood , Liver Neoplasms/diagnosis , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Neoplasm Staging , ATPases Associated with Diverse Cellular Activities/blood , Apoptosis , Neoplasm Metastasis , Oxidative Stress , Liver/pathology , Biomarkers, Tumor/blood
2.
Asian Pac J Cancer Prev ; 23(1): 271-279, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-35092397

ABSTRACT

OBJECTIVE: We aimed to investigate the signalling crosstalk of TNF-related apoptosis-inducing ligand, TRAIL death receptors, tumour protein p53, and programmed cell death (PDCD5) with IQGAPs. Also, we targeted the crosstalk between IQGAPs genes with decoy receptors, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and interleukins -8 (IL-8) and its receptor genes in a designed model of hepatocellular carcinoma induced in male Balb/c mice. METHODS: The presence of HCC was confirmed by histological and morphological alterations. In parallel to the incidence of hepatic cancer, we found lung, heart, and kidney cancer after treatment with DEN. RESULTS: Our results show that the expression of mRNA of IQGAP1, TRAIL decoy receptors, NF-κB, and IL-8 genes was elevated in hepatocellular carcinoma, as compared to normal liver tissue, while their expression was further up-regulated by increasing the dose of diethylnitrosamine. The expression of IQGAP2, TRAIL death receptors, p53, and PDCD5 was significantly down-regulated in HCC (p˂0.05). For confirmation of gene expression, protein levels of both IQGAP1 and P53 were measured by western blot analysis, which showed that diethylnitrosamine enhanced protein expression of IQGAP1 and diminished that of p53. CONCLUSION: IQGAPs have a direct signaling relationship with p53, IL-8, and TRAIL family. This interaction is recognized as a key signalling pathway for hepatocellular carcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/metabolism , ras GTPase-Activating Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Diethylnitrosamine/administration & dosage , Disease Models, Animal , Down-Regulation , Interleukin-8/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/genetics , Up-Regulation
3.
Gene ; 813: 146124, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34921950

ABSTRACT

The knowledge of RNA editing modifications and its subsequent proteomic diversity in is still limited and represents only the tip of the iceberg. Adenosine to inosine (A-to-I) RNA editing is the most prevalent in RNA editome with a rising role for ADARgene family as a major regulator of the dynamic landscape of RNA editing. This study aimed at evaluating the potential chemopreventive effects of the epigenetic regulator "pterostilbene" in diethylnitrosamine (DEN)-exposedrat model. Consequently, the hepatic Adars expression was investigated as a possible mechanism for mediation of the putative pterostilbene-induced chemopreventive effect. The effects of administration of pterostilbene were investigated on the structural changes, immunohistochemical staining, liver function test, serum alpha feto-protein (AFP), IL-6, and hepatic Adar1 and Adar2 relative gene expression at the beginning and at the 6th week of the study. Pterostilbene attenuated DEN-induced liver injury, improves hepatocyte parrafin-1 (Hep Par-1), decreases heat shock protein 70 (HSP70), improved AFP, serum albumin, transaminases, IL-6 with alleviation of disturbed hepatic Adar1 and Adar2 expression. This study spotlights the role of pterostilbene in attenuation of DEN-induced liver injury which could be mediated, at least partially, through the alleviation of the aberrant expression of Adar enzymes. Yet, more in-depth studies are needed to further elucidate the molecular mechanisms underlying the effects of pterostilbene on RNA editing enzymes.


Subject(s)
Adenosine Deaminase/biosynthesis , Liver Cirrhosis/drug therapy , Stilbenes/pharmacology , Adenosine Deaminase/genetics , Adenosine Deaminase Inhibitors/pharmacology , Animals , Diethylnitrosamine/administration & dosage , Gene Expression , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/enzymology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Proteomics , RNA Editing , RNA-Binding Proteins/genetics , Rats , Rats, Wistar , Transcriptome
4.
Nat Commun ; 12(1): 5725, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593796

ABSTRACT

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.


Subject(s)
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
5.
Hepatology ; 74(6): 3037-3055, 2021 12.
Article in English | MEDLINE | ID: mdl-34292642

ABSTRACT

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.


Subject(s)
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
6.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33753481

ABSTRACT

The CXC chemokine receptor type 4 (CXCR4) receptor and its ligand, CXCL12, are overexpressed in various cancers and mediate tumor progression and hypoxia-mediated resistance to cancer therapy. While CXCR4 antagonists have potential anticancer effects when combined with conventional anticancer drugs, their poor potency against CXCL12/CXCR4 downstream signaling pathways and systemic toxicity had precluded clinical application. Herein, BPRCX807, known as a safe, selective, and potent CXCR4 antagonist, has been designed and experimentally realized. In in vitro and in vivo hepatocellular carcinoma mouse models it can significantly suppress primary tumor growth, prevent distant metastasis/cell migration, reduce angiogenesis, and normalize the immunosuppressive tumor microenvironment by reducing tumor-associated macrophages (TAMs) infiltration, reprogramming TAMs toward an immunostimulatory phenotype and promoting cytotoxic T cell infiltration into tumor. Although BPRCX807 treatment alone prolongs overall survival as effectively as both marketed sorafenib and anti-PD-1, it could synergize with either of them in combination therapy to further extend life expectancy and suppress distant metastasis more significantly.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Receptors, CXCR4/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Drug Synergism , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Molecular Docking Simulation , Rats , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Sorafenib/pharmacology , Sorafenib/therapeutic use , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Xenograft Model Antitumor Assays
7.
Eur Rev Med Pharmacol Sci ; 25(2): 710-721, 2021 01.
Article in English | MEDLINE | ID: mdl-33577025

ABSTRACT

OBJECTIVE: To study the association of inflammatory factors and hepatocarcinoma stem cells of induced liver cancer rats. MATERIALS AND METHODS: 30 SD male healthy rats were selected. 10 rats were given water as normal control group. 10 rats only were implemented laparotomy as sham operation group. The remaining 10 rats were the liver cancer model group and treated with diethylnitrosamine (DEN) to induce liver cancer. Real-time quantitative PCR was used to detect the related inflammatory factors in HCC tissues, including interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), transforming growth factor-ß1 (TGF-ß), human interleukin-1α (IL-1α), human interleukin 1ß (IL-1ß) and levels of hepatocarcinoma stem cells indicators CD90, CD133, Alpha-fetoprotein (AFP). Correlation analysis was used to analyze the correlation between inflammatory factors and hepatocarcinoma stem cells markers CD90 and CD133. RESULTS: The expression levels of IL-6, MCP-1 and TGF-ß of HCC tissues in liver cancer model group were significantly higher than those in the control group and the sham operation group. The expression levels of CD90 and CD133 of tissues in the liver cancer model group were significantly higher than those in the control group and the sham operation group. The differences were statistically significant (p<0.001). By inhibiting related inflammatory factors, the growth, migration and invasion of liver cancer cells were significantly inhibited, and apoptosis was promoted. Correlation analysis results showed that the expression changes of IL-6, MCP-1 and TGF-ß were significantly positively correlated with CD90 up-regulation (p<0.05), while the expression changes of IL-6, MCP-1 and TGF-ß were significantly positively correlated with CD133 up-regulation (p<0.05). CONCLUSIONS: The inflammatory factors IL-6, MCP-1 and TGF-ß are closely related to hepatocarcinoma stem cells, which play an important role in promoting the occurrence and deterioration of liver cancer.


Subject(s)
Biomarkers, Tumor/genetics , Chemokine CCL2/genetics , Interleukin-6/genetics , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Transforming Growth Factor beta1/genetics , Administration, Oral , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Cell Proliferation , Chemokine CCL2/metabolism , Diethylnitrosamine/administration & dosage , Disease Models, Animal , Interleukin-6/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Male , Neoplastic Stem Cells/pathology , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism
8.
Cell Mol Gastroenterol Hepatol ; 11(2): 407-431, 2021.
Article in English | MEDLINE | ID: mdl-32932015

ABSTRACT

BACKGROUND & AIMS: Cancer-associated fibroblasts (CAFs) play a key role in the cancer process, but the research progress is hampered by the paucity of preclinical models that are essential for mechanistic dissection of cancer cell-CAF interactions. Here, we aimed to establish 3-dimensional (3D) organotypic co-cultures of primary liver tumor-derived organoids with CAFs, and to understand their interactions and the response to treatment. METHODS: Liver tumor organoids and CAFs were cultured from murine and human primary liver tumors. 3D co-culture models of tumor organoids with CAFs and Transwell culture systems were established in vitro. A xenograft model was used to investigate the cell-cell interactions in vivo. Gene expression analysis of CAF markers in our hepatocellular carcinoma cohort and an online liver cancer database indicated the clinical relevance of CAFs. RESULTS: To functionally investigate the interactions of liver cancer cells with CAFs, we successfully established murine and human 3D co-culture models of liver tumor organoids with CAFs. CAFs promoted tumor organoid growth in co-culture with direct cell-cell contact and in a Transwell system via paracrine signaling. Vice versa, cancer cells secrete paracrine factors regulating CAF physiology. Co-transplantation of CAFs with liver tumor organoids of mouse or human origin promoted tumor growth in xenograft models. Moreover, tumor organoids conferred resistance to clinically used anticancer drugs including sorafenib, regorafenib, and 5-fluorouracil in the presence of CAFs, or the conditioned medium of CAFs. CONCLUSIONS: We successfully established murine and human 3D co-culture models and have shown robust effects of CAFs in liver cancer nurturing and treatment resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/pathology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Organoids/pathology , Animals , Antineoplastic Agents/therapeutic use , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Hepatocellular/pathology , Coculture Techniques , Culture Media, Conditioned/metabolism , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Drug Resistance, Neoplasm , Humans , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Mice , Organoids/drug effects , Paracrine Communication , Primary Cell Culture , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays
9.
Cell Mol Gastroenterol Hepatol ; 11(2): 597-621, 2021.
Article in English | MEDLINE | ID: mdl-32987153

ABSTRACT

BACKGROUND & AIMS: Tristetraprolin (TTP) is a key post-transcriptional regulator of inflammatory and oncogenic transcripts. Accordingly, TTP was reported to act as a tumor suppressor in specific cancers. Herein, we investigated how TTP contributes to the development of liver inflammation and fibrosis, which are key drivers of hepatocarcinogenesis, as well as to the onset and progression of hepatocellular carcinoma (HCC). METHODS: TTP expression was investigated in mouse/human models of hepatic metabolic diseases and cancer. The role of TTP in nonalcoholic steatohepatitis and HCC development was further examined through in vivo/vitro approaches using liver-specific TTP knockout mice and a panel of hepatic cancer cells. RESULTS: Our data demonstrate that TTP loss in vivo strongly restrains development of hepatic steatosis and inflammation/fibrosis in mice fed a methionine/choline-deficient diet, as well as HCC development induced by the carcinogen diethylnitrosamine. In contrast, low TTP expression fostered migration and invasion capacities of in vitro transformed hepatic cancer cells likely by unleashing expression of key oncogenes previously associated with these cancerous features. Consistent with these data, TTP was significantly down-regulated in high-grade human HCC, a feature further correlating with poor clinical prognosis. Finally, we uncover hepatocyte nuclear factor 4 alpha and early growth response 1, two key transcription factors lost with hepatocyte dedifferentiation, as key regulators of TTP expression. CONCLUSIONS: Although TTP importantly contributes to hepatic inflammation and cancer initiation, its loss with hepatocyte dedifferentiation fosters cancer cells migration and invasion. Loss of TTP may represent a clinically relevant biomarker of high-grade HCC associated with poor prognosis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Tristetraprolin/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Datasets as Topic , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/immunology , Hepatocytes , Humans , Liver/immunology , Liver/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemistry , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Male , Mice , Non-alcoholic Fatty Liver Disease , Primary Cell Culture , Prognosis , RNA-Seq , Survival Analysis , Tristetraprolin/genetics
10.
Clin Cancer Res ; 26(17): 4581-4589, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32499234

ABSTRACT

PURPOSE: Targeted therapies for cancer have accelerated the need for functional imaging strategies that inform therapeutic efficacy. This study assesses the potential of functional genetic screening to integrate therapeutic target identification with imaging probe selection through a proof-of-principle characterization of a therapy-probe pair using dynamic nuclear polarization (DNP)-enhanced magnetic resonance spectroscopic imaging (MRSI). EXPERIMENTAL DESIGN: CRISPR-negative selection screens from a public dataset were used to identify the relative dependence of 625 cancer cell lines on 18,333 genes. Follow-up screening was performed in hepatocellular carcinoma with a focused CRISPR library targeting imaging-related genes. Hyperpolarized [1-13C]-pyruvate was injected before and after lactate dehydrogenase inhibitor (LDHi) administration in male Wistar rats with autochthonous hepatocellular carcinoma. MRSI evaluated intratumoral pyruvate metabolism, while T2-weighted segmentations quantified tumor growth. RESULTS: Genetic screening data identified differential metabolic vulnerabilities in 17 unique cancer types that could be imaged with existing probes. Among these, hepatocellular carcinoma required lactate dehydrogenase (LDH) for growth more than the 29 other cancer types in this database. LDH inhibition led to a decrease in lactate generation (P < 0.001) and precipitated dose-dependent growth inhibition (P < 0.01 overall, P < 0.05 for dose dependence). Intratumoral alanine production after inhibition predicted the degree of growth reduction (P < 0.001). CONCLUSIONS: These findings demonstrate that DNP-MRSI of LDH activity using hyperpolarized [1-13C]-pyruvate is a theranostic strategy for hepatocellular carcinoma, enabling quantification of intratumoral LDHi pharmacodynamics and therapeutic efficacy prediction. This work lays the foundation for a novel theranostic platform wherein functional genetic screening informs imaging probe selection to quantify therapeutic efficacy on a cancer-by-cancer basis.


Subject(s)
Carbon-13 Magnetic Resonance Spectroscopy/methods , Carcinoma, Hepatocellular/diagnosis , Liver Neoplasms, Experimental/diagnosis , Liver Neoplasms/diagnosis , Molecular Imaging/methods , Animals , CRISPR-Cas Systems/genetics , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Datasets as Topic , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Early Detection of Cancer/methods , Humans , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Liver/diagnostic imaging , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Molecular Probes/administration & dosage , Molecular Probes/pharmacokinetics , Precision Medicine/methods , Proof of Concept Study , Pyruvic Acid/metabolism , Rats
11.
Anal Chem ; 92(10): 6993-7002, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32311262

ABSTRACT

Diversified oxidized-lipid molecules are responsible for inflammation and cell death, including ferroptosis. Lipid radicals are the source of these oxidized lipids, which are the initial key molecules in the lipid peroxidation chain reaction. However, owing to their extremely high reactivity and short half-life, an established detection technique is not available. Here, we propose a high-performance liquid chromatography fluorometry and high-resolution tandem mass spectrometry system combined with a fluorescent probe as a structural analysis method for lipid-derived radicals. We detected 132 lipid-derived radicals, including 111 new species, from five polyunsaturated fatty acids. In addition, a database was constructed for which the initial fatty acid could be determined using the radical structure. Further, 12 endogenous lipid-derived radicals were identified in carcinogen-induced liver cancer mouse models. Therefore, this method and its corresponding database will provide novel insights into mechanisms underlying the lipid peroxidation, including the associated inflammation and ferroptosis.


Subject(s)
Lipids/analysis , Liver Neoplasms/diagnosis , Animals , Diethylnitrosamine/administration & dosage , Disease Models, Animal , Free Radicals/analysis , Injections, Intraperitoneal , Liver Neoplasms/chemically induced , Male , Mice , Mice, Inbred C57BL , Molecular Structure
12.
Bioorg Chem ; 99: 103818, 2020 06.
Article in English | MEDLINE | ID: mdl-32276135

ABSTRACT

Targeted therapy is a new strategy for cancer treatment that targets chemical entities specific to cancer cells than normal ones. One of the features associated with malignancy is the elevated copper which plays an integral role in angiogenesis. Work is in progress in our lab to identify new copper chelators to target elevated copper under targeted therapy for the killing of cancer cells. Recently, a coumarin-based copper chelator, di(2-picolyl)amine-3(bromoacetyl)coumarin hybrid molecule (ligand-L) has been synthesized by us, and also studied its copper-dependent macromolecular damage response in copper overloaded lymphocytes. The present study investigates the anticancer activity of ligand-L and its mode of action in rat model of diethylnitrosamine (DEN) induced hepatocellular carcinoma. It has been found that liver tissue has a marked increase in copper levels in DEN induced hepatocellular carcinoma. Ex vivo results showed that ligand-L inhibited cell viability, induced reactive oxygen species (ROS) generation, DNA damage, loss of mitochondrial membrane potential and caspase-3 activation in isolated hepatocellular carcinoma cells (HCC). All these effects induced by ligand-L were abrogated by neocuproine and N-acetylcysteine (ROS scavenger). Further, ligand-L treatment of animals bearing hepatocellular carcinoma results in an increment in the cellular redox scavengers, lipid peroxidation and DNA breakage in malignant hepatocytes. In vivo studies using ligand-L also showed that ligand-L possesses anticancer properties as evidenced by improvement in liver marker enzymes and liver surface morphology, and reduced alpha-fetoprotein in the treated group compared to untreated cancer-induced group. Overall, this study suggests that copper-ligand-L interaction leads to ROS generation which caused DNA damage and apoptosis in malignant cells. This study provides enough support to establish ligand-L as a clinically relevant lead molecule for the treatment of different malignancies.


Subject(s)
Aminocoumarins/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Carcinoma, Hepatocellular/drug therapy , Copper/pharmacology , Liver Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Aminocoumarins/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antioxidants/chemical synthesis , Antioxidants/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Death/drug effects , Cell Proliferation/drug effects , Copper/chemistry , DNA Damage , Diethylnitrosamine/administration & dosage , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hepatocytes/drug effects , Hepatocytes/metabolism , Injections, Intraperitoneal , Lipid Peroxidation/drug effects , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Molecular Structure , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/analysis , Structure-Activity Relationship
13.
Cancer Biol Med ; 17(1): 142-153, 2020 02 15.
Article in English | MEDLINE | ID: mdl-32296582

ABSTRACT

Objective: Epithelial cancers often originate from progenitor cells, while the origin of hepatocellular carcinoma (HCC) is still controversial. HCC, one of the deadliest cancers, is closely linked with liver injuries and chronic inflammation, which trigger massive infiltration of bone marrow-derived cells (BMDCs) during liver repair. Methods: To address the possible roles of BMDCs in HCC origination, we established a diethylnitrosamine (DEN)-induced HCC model in bone marrow transplanted mice. Immunohistochemistry and frozen tissue immunofluorescence were used to verify DEN-induced HCC in the pathology of the disease. The cellular origin of DEN-induced HCC was further studied by single cell sequencing, single-cell nested PCR, and immunofluorescence-fluorescence in situ hybridization. Results: Studies by using single cell sequencing and biochemical analysis revealed that HCC cells in these mice were coming from donor mice BMDCs, and not from recipient mice. Furthermore, the copy numbers of mouse orthologs of several HCC-related genes previously reported in human HCC were also altered in our mouse model. DEN-induced HCCs exhibited a similar histological phenotype and genomic profile as human HCCs. Conclusions: These results suggested that BMDCs are an important origin of HCC, which provide important clues to HCC prevention, detection, and treatments.


Subject(s)
Bone Marrow Cells/pathology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms/pathology , Liver/cytology , Animals , Biomarkers, Tumor/genetics , Bone Marrow Transplantation , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Cell Separation/methods , DNA Copy Number Variations , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Liver/drug effects , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Male , Mice, Transgenic , Single-Cell Analysis/methods , Transplantation Chimera , Whole Genome Sequencing
14.
Life Sci ; 243: 117274, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31927046

ABSTRACT

AIMS: 2,3,5,4'-Tetrahydroxystilbene-2-O-ß-d-glucoside (TSG) is the key bioactive ingredient extracted from Polygonum multiflorum Thumb. Pharmacological studies suggest that it exerts numerous biological effects, including anti-oxidant, anti-aging, and anti-inflammation. This study aimed at investigating the effect of TSG on diethylnitrosamine (DEN)-induced acute hepatotoxicity and DNA damage. MAIN METHODS: Fifty male C57BL/6 mice were randomly divided into 5 groups (n = 10 each): control, DEN, DEN+TSG (low), DEN+TSG (high) and TSG (high) groups. DEN (100 mg/kg) was injected intraperitoneally (i.p.) alone or with TSG (30 or 60 mg/kg, i.p.) for 5 consecutive days. KEY FINDINGS: TSG inhibited liver injury and inflammatory cell infiltration in DEN-treated mice. It also attenuated DEN-induced accumulation of reactive oxygen species (ROS), proinflammatory cytokines, and DNA damage. Moreover, TSG promoted the expression of nuclear erythroid 2-related factor 2 (Nrf2) target antioxidant genes by enhancing Nrf2 protein phosphorylation and nuclear translocation. As major phase I detoxification enzymes, cytochrome P450 family 2 subfamily E member 1 (CYP2E1) and cytochrome P450 1 subfamily A member 1 (CYP1A1) are responsible for the metabolic activation of DEN. We found that TSG administration inhibited CYP2E1 and CYP1A1 induction in DEN-treated mice. SIGNIFICANCE: These results indicate that TSG can alleviate DEN-induced acute hepatotoxicity by modulating the Nrf2-related antioxidant system and metabolic activation of DEN. Therefore, TSG might be a promising medication for DEN-induced liver injury treatment.


Subject(s)
Alkylating Agents/toxicity , DNA Damage , Diethylnitrosamine/toxicity , Glucosides/pharmacology , Liver/drug effects , Stilbenes/pharmacology , Animals , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP2E1/metabolism , Cytokines/metabolism , Diethylnitrosamine/administration & dosage , Dose-Response Relationship, Drug , Glucosides/administration & dosage , Inflammation Mediators/metabolism , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Stilbenes/administration & dosage
15.
Int J Cancer ; 146(12): 3410-3422, 2020 06 15.
Article in English | MEDLINE | ID: mdl-31721195

ABSTRACT

RuvBL1 is an AAA+ ATPase whose expression in hepatocellular carcinoma (HCC) correlates with a poor prognosis. In vitro models suggest that targeting RuvBL1 could be an effective strategy against HCC. However, the role of RuvBL1 in the onset and progression of HCC remains unknown. To address this question, we developed a RuvBL1hep+/- mouse model and evaluated the outcome of DEN-induced liver carcinogenesis up to 12 months of progression. We found that RuvBL1 haploinsufficiency initially delayed the onset of liver cancer, due to a reduced hepatocyte turnover in RuvBL1hep+/- mice. However, RuvBL1hep+/- mice eventually developed HCC nodules that, with aging, grew larger than in the control mice. Moreover, RuvBL1hep+/- mice developed hepatic insulin resistance and impaired glucose homeostasis. We could determine that RuvBL1 regulates insulin signaling through the Akt/mTOR pathway in liver physiology in vivo as well as in normal hepatocytic and HCC cells in vitro. Whole transcriptome analysis of mice livers confirmed the major role of RuvBL1 in the regulation of hepatic glucose metabolism. Finally, RuvBL1 expression was found significantly correlated to glucose metabolism and mTOR signaling by bioinformatic analysis of human HCC sample from the publicly available TGCA database. These data uncover a role of RuvBL1 at the intersection of liver metabolism, hepatocyte proliferation and HCC development, providing a molecular rationale for its overexpression in liver cancer.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Carcinoma, Hepatocellular/genetics , Carrier Proteins/genetics , DNA Helicases/genetics , Insulin Resistance/genetics , Liver Neoplasms/genetics , Liver/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cohort Studies , DNA Helicases/metabolism , Datasets as Topic , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Disease Models, Animal , Disease Progression , Disease-Free Survival , Glucose/metabolism , Haploinsufficiency , Hepatocytes/metabolism , Humans , Insulin/metabolism , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
16.
FEBS Open Bio ; 9(11): 1900-1908, 2019 11.
Article in English | MEDLINE | ID: mdl-31433574

ABSTRACT

RNA-sequencing (RNA-Seq) is a useful method to explore the molecular events in cells and tissues at the transcriptional level. However, comprehensive transcriptome analysis of hepatocarcinogenesis and progression is lacking. In this study, we aimed to characterize a dimethylnitrosamine (DEN) and carbon tetrachloride (CCl4 ; DEN+CCl4 )-induced hepatocellular carcinoma (HCC) mouse model by RNA-Seq. In total, 2033 genes were up-regulated and 841 genes were down-regulated after DEN and CCl4 stimulation. The differentially expressed genes were highly enriched for the Gene Ontology terms oxoacid metabolic process, carboxylic acid metabolic process, and organic acid metabolic process. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that the top five significantly overrepresented pathways were metabolic pathways, chemical carcinogenesis, steroid hormone biosynthesis, retinol metabolism and metabolism of xenobiotics by cytochrome P450. Moreover, a protein-protein interaction network analysis indicated that Rous sarcoma oncogene (Src) may play a key role in DEN+CCl4 -induced HCC. These results provide a comprehensive overview of transcriptome events in DEN+CCl4 -induced HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , Sequence Analysis, RNA , Animals , Carcinoma, Hepatocellular/chemically induced , Diethylnitrosamine/administration & dosage , Gene Expression Profiling , Injections, Intraperitoneal , Liver Neoplasms/chemically induced , Male , Mice , Mice, Inbred C57BL
17.
Cancer Sci ; 110(9): 2748-2759, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31301081

ABSTRACT

In hepatocarcinogenesis induced by diethylnitrosamine (DEN) in B6C3F1 mice, the BrafV637E mutation, corresponding to the human BRAFV600E mutation, plays a pivotal role. The livers of transgenic mice with a hepatocyte-specific human BRAFV600E mutation weighed 4.5 times more than that of normal mice and consisted entirely of hepatocytes, resembling DEN-induced preneoplastic hepatocytes. However, these transgenic mice spontaneously died 7 wk after birth, therefore this study aimed to clarify the causes of death. In the transgenic mice, the liver showed thrombopoietin (TPO) overexpression, which is associated with eventual megakaryocytosis and thrombocytosis, and activated platelets were deposited in hepatic sinusoids. TPO was also overexpressed in the DEN-induced hepatic tumors, and sinusoidal platelet deposition was observed in the hepatic tumors of humans and mice. Podoplanin was expressed in some of the Kupffer cells in the liver of the transgenic mice, indicating that platelet activation occurred via the interaction of podoplanin with C-type lectin receptor 2 (CLEC-2) on the platelet membrane. Additionally, erythrocyte dyscrasia and glomerulonephropathy/interstitial pneumonia associated with platelet deposition were observed. In the transgenic mice, aspirin (Asp) administration prevented platelet activation, reduced the liver/body weight ratio, decreased the platelet deposition in the liver, kidney, and lung, and prevented erythrocyte dyscrasia and ameliorated the renal/pulmonary changes. Thrombopoietin overproduction by BRAFV600E-mutated hepatocytes may contribute to hepatocyte proliferation via thrombocytosis, platelet activation, and the interaction of platelets with hepatic sinusoidal cells, while hematologic, renal, and pulmonary disorders due to aberrant platelet activation may lead to spontaneous death in the transgenic mice.


Subject(s)
Carcinogenesis/genetics , Liver Neoplasms, Experimental/pathology , Liver/pathology , Proto-Oncogene Proteins B-raf/genetics , Thrombopoietin/metabolism , Animals , Biopsy , Blood Platelets/pathology , Bone Marrow/pathology , Capillaries/pathology , Carcinogens/administration & dosage , Carcinogens/toxicity , Cell Proliferation/genetics , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Female , Gene Expression Regulation, Neoplastic , Hepatectomy , Hepatocytes/pathology , Humans , Liver/blood supply , Liver/cytology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Platelet Activation/genetics , Primary Cell Culture , Proto-Oncogene Proteins B-raf/metabolism , Tumor Cells, Cultured
18.
Toxicol Appl Pharmacol ; 378: 114611, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31176654

ABSTRACT

Hepatocellular carcinoma (HCC) arises after a long period of exposition to etiological factors that might be either independent or collectively contributing. Several rodent models resemble human HCC; however, the major limitation of these models is the lack of chronic injury that reproducibly mimics the molecular alterations as it occurs in humans. Thus, we hypothesized that chronic administration of different DEN treatments identifies the best-fit dose to induce the HCC and/or to determine whether small DEN doses act synergistically with other known hepatotoxins to induce HCC in mice. C57BL/6 J male mice were intraperitoneally injected twice a week for 6 weeks with different DEN doses ranging from 2.5 to 40 mg/kg body weight; then, selected doses (2.5, 5 and 20 mg/kg) for 6, 10, 14, and 18 weeks. We demonstrated that DEN at 20 mg/kg promoted reactive oxygen species and 4-hydroxynonenal production, cell proliferation inflammatory infiltrate, and fibrosis, which in turn induced liver cancer by week 18. These parameters were established by evaluating histopathological changes, HCC markers such as glutathione S-transferase placental-1 (Gstp1), Cytokeratin-19 (Ck19) and prostaglandin reductase-1 (Ptgr1); that of Cyp2e1, a DEN metabolizing enzyme; and the expression of the proliferation marker Ki67. While DEN at 2.5 and 5 mg/kg increased Gstp1 and Ck19, DEN at 20 mg/kg decreased them and Cyp2e1 expression and activity. In summary, our results demonstrate that DEN chronically administrated at 20 mg/kg induces the HCC, while DEN at 2.5 and 5 mg/kg could be useful in elucidating its synergistic effect with other hepatotoxic agents in mice.


Subject(s)
Carcinogenesis/drug effects , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/adverse effects , Liver Neoplasms/chemically induced , Liver/drug effects , Animals , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Drug Synergism , Fibrosis/chemically induced , Fibrosis/metabolism , Inflammation/metabolism , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
19.
Environ Toxicol ; 34(9): 1025-1033, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31087429

ABSTRACT

Hepatocellular carcinoma is considered one of the most prevalent and lethal malignancies worldwide. Chemotherapy with cytotoxic agents showed a low response rate with possible toxic effects. Recently, some emphases have been placed on the anticancer properties of bovine whey protein and its components, especially lactoferrin. The present study aimed to evaluate and compare the antihepatocarcinogenic activity of bovine whey protein concentrate (WPC, 300 and 600 mg/kg body weight) and lactoferrin (30 and 60 mg/kg body weight), orally and daily for 14 weeks, in the mice model of diethylnitrosamine (DEN)-induced hepatocarcinogenesis. The results showed that both WPC and lactoferrin (in a dose-dependent manner) alleviated significantly (P < .001) the elevation in serum markers of liver carcinoma and inflammation in the DEN-treated mice. Also, they exhibited a great amelioration in the livers' histological structure of the DEN-treated mice by 37.0% to 66.7%. In addition, they decreased significantly (P < .001) the hepatic DNA fragmentation in the DEN-treated mice by 23.1% to 32.7%. Only, the high doses of WPC and lactoferrin completely modulated the decrease in the activity of liver enzymic antioxidant defense system (catalase, glutathione peroxidase, and superoxide dismutase) and improved significantly (P < .01-.001) the concentration of hepatic reduced glutathione of the DEN-treated mice. Moreover, the high doses of WPC and lactoferrin reduced significantly (P < .05-.001) the elevation in the concentrations of hepatic active caspases 3, 8, and 9 of the DEN-treated mice. In conclusion, both WPC and lactoferrin were effective in inhibiting the hepatocarcinogenic activity of DEN in mice model through their ability to alleviate the hepatic inflammation and apoptosis.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Liver Neoplasms, Experimental/drug therapy , Liver/drug effects , Whey Proteins/therapeutic use , Animals , Anticarcinogenic Agents/administration & dosage , Antioxidants/metabolism , Cattle , Diethylnitrosamine/administration & dosage , Dose-Response Relationship, Drug , Lactoferrin/administration & dosage , Lactoferrin/therapeutic use , Liver/enzymology , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Whey Proteins/administration & dosage
20.
Elife ; 82019 04 16.
Article in English | MEDLINE | ID: mdl-30990169

ABSTRACT

Hepatocellular carcinoma (HCC) is a deadly human cancer associated with chronic inflammation. The cytosolic pathogen sensor NLRP12 has emerged as a negative regulator of inflammation, but its role in HCC is unknown. Here we investigated the role of NLRP12 in HCC using mouse models of HCC induced by carcinogen diethylnitrosamine (DEN). Nlrp12-/- mice were highly susceptible to DEN-induced HCC with increased inflammation, hepatocyte proliferation, and tumor burden. Consistently, Nlrp12-/- tumors showed higher expression of proto-oncogenes cJun and cMyc and downregulation of tumor suppressor p21. Interestingly, antibiotics treatment dramatically diminished tumorigenesis in Nlrp12-/- mouse livers. Signaling analyses demonstrated higher JNK activation in Nlrp12-/- HCC and cultured hepatocytes during stimulation with microbial pattern molecules. JNK inhibition or NLRP12 overexpression reduced proliferative and inflammatory responses of Nlrp12-/- hepatocytes. In summary, NLRP12 negatively regulates HCC pathogenesis via downregulation of JNK-dependent inflammation and proliferation of hepatocytes.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Down-Regulation , Hepatocytes/enzymology , Hepatocytes/physiology , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/biosynthesis , Liver Neoplasms/physiopathology , Animals , Carcinogens/administration & dosage , Carcinoma, Hepatocellular/chemically induced , Cell Proliferation , Diethylnitrosamine/administration & dosage , Disease Models, Animal , Gene Knockout Techniques , Intracellular Signaling Peptides and Proteins/deficiency , Liver Neoplasms/chemically induced , Mice, Knockout , Proto-Oncogene Proteins c-myc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...