Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Hepatology ; 74(2): 776-796, 2021 08.
Article in English | MEDLINE | ID: mdl-33619771

ABSTRACT

BACKGROUND AND AIMS: HCC undergoes active metabolic reprogramming. Reactive oxygen species (ROS) are excessively generated in cancer cells and are neutralized by NADPH. Malic enzymes (MEs) are the less studied NADPH producers in cancer. APPROACH AND RESULTS: We found that ME1, but not ME3, was regulated by the typical oxidative stress response pathway mediated by kelch-like ECH associated protein 1/nuclear factor erythroid 2-related factor (NRF2). Surprisingly, ME3 was constitutively induced by superenhancers. Disruption of any ME regulatory pathways decelerated HCC progression and sensitized HCC to sorafenib. Therapeutically, simultaneous blockade of NRF2 and a superenhancer complex completely impeded HCC growth. We show that superenhancers allow cancer cells to counteract the intrinsically high level of ROS through constitutively activating ME3 expression. When HCC cells encounter further episodes of ROS insult, NRF2 allows cancer cells to adapt by transcriptionally activating ME1. CONCLUSIONS: Our study reveals the complementary regulatory mechanisms which control MEs and provide cancer cells multiple layers of defense against oxidative stress. Targeting both regulatory mechanisms represents a potential therapeutic approach for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Malate Dehydrogenase/genetics , NAD (+) and NADP (+) Dependent Alcohol Oxidoreductases/genetics , NF-E2-Related Factor 2/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hepatocytes , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Liver Neoplasms/genetics , Malate Dehydrogenase/metabolism , Metabolomics , Mice , NAD (+) and NADP (+) Dependent Alcohol Oxidoreductases/metabolism , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Transcriptional Activation , Xenograft Model Antitumor Assays
2.
Hepatology ; 69(5): 2013-2030, 2019 05.
Article in English | MEDLINE | ID: mdl-30516846

ABSTRACT

Hepatocellular carcinoma (HCC) is the third most lethal cancer worldwide. Increasing evidence shows that epigenetic alterations play an important role in human carcinogenesis. Deregulation of DNA methylation and histone modifications have recently been characterized in HCC, but the significance of chromatin remodeling in liver carcinogenesis remains to be explored. In this study, by systematically analyzing the expression of chromatin remodeling genes in human HCCs, we found that helicase, lymphoid-specific (HELLS), an SWI2/SNF2 chromatin remodeling enzyme, was remarkably overexpressed in HCC. Overexpression of HELLS correlated with more aggressive clinicopathological features and poorer patient prognosis compared to patients with lower HELLS expression. We further showed that up-regulation of HELLS in HCC was conferred by hyperactivation of transcription factor specificity protein 1 (SP1). To investigate the functions of HELLS in HCC, we generated both gain-of-function and loss-of-function models by the CRISPR activation system, lentiviral short hairpin RNA, and the CRISPR/Cas9 genome editing system. We demonstrated that overexpression of HELLS augmented HCC cell proliferation and migration. In contrast, depletion of HELLS reduced HCC growth and metastasis both in vitro and in vivo. Moreover, inactivation of HELLS led to metabolic reprogramming and reversed the Warburg effect in HCC cells. Mechanistically, by integrating analysis of RNA sequencing and micrococcal nuclease sequencing, we revealed that overexpression of HELLS increased nucleosome occupancy, which obstructed the accessibility of enhancers and hindered formation of the nucleosome-free region (NFR) at the transcription start site. Though this mechanism, up-regulation of HELLS mediated epigenetic silencing of multiple tumor suppressor genes including E-cadherin, FBP1, IGFBP3, XAF1 and CREB3L3 in HCC. Conclusion: Our data reveal that HELLS is a key epigenetic driver of HCC; by altering the nucleosome occupancy at the NFR and enhancer, HELLS epigenetically suppresses multiple tumor suppressor genes to promote HCC progression.


Subject(s)
Carcinoma, Hepatocellular/enzymology , DNA Helicases/metabolism , Liver Neoplasms, Experimental/enzymology , Nucleosomes/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Carcinoma, Hepatocellular/etiology , Cell Line, Tumor , Chromatin Assembly and Disassembly , DNA Helicases/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Liver Neoplasms, Experimental/etiology , Mice, Knockout , Mice, Nude , Neoplasm Metastasis , Sp1 Transcription Factor/metabolism
3.
Hepatology ; 69(4): 1768-1786, 2019 04.
Article in English | MEDLINE | ID: mdl-30561826

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most prevalent and lethal cancers worldwide which lacks effective treatment. Cancer cells experience high levels of oxidative stress due to increased generation of reactive oxygen species (ROS). Increased antioxidant-producing capacity is therefore found in cancer cells to counteract oxidative stress. The thioredoxin system is a ubiquitous mammalian antioxidant system which scavenges ROS, and we demonstrate that it is vital for HCC growth as it maintains intracellular reduction-oxidation (redox) homeostasis. Transcriptome sequencing in human HCC samples revealed significant overexpression of thioredoxin reductase 1 (TXNRD1), the cytosolic subunit and key enzyme of the thioredoxin system, with significant correlations to poorer clinicopathological features and patient survival. Driven by the transcriptional activation of nuclear factor (erythroid-derived 2)-like 2, the master protector against oxidative stress, TXNRD1 counteracts intracellular ROS produced in human HCC. Inhibition of TXNRD1 through genetic inhibition hindered the proliferation of HCC cells and induced apoptosis in vitro. Administration of the pharmacological TXNRD1 inhibitor auranofin (AUR) effectively suppressed the growth of HCC tumors induced using the hydrodynamic tail vein injection and orthotopic implantation models in vivo. Furthermore, AUR sensitized HCC cells toward the conventional therapeutic sorafenib. Conclusion: Our study highlights the reliance of HCC cells on antioxidants for redox homeostasis and growth advantage; targeting TXNRD1 resulted in dramatic accumulation of ROS, which was found to be an effective approach for the suppression of HCC tumor growth.


Subject(s)
Auranofin/therapeutic use , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Oxidative Stress/drug effects , Thioredoxin Reductase 1/metabolism , Animals , Antineoplastic Agents/therapeutic use , Auranofin/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Liver Neoplasms/drug therapy , Mice , NF-E2-Related Factor 2/metabolism , Sorafenib/therapeutic use , Thioredoxin Reductase 1/antagonists & inhibitors
4.
Nat Commun ; 8(1): 517, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28894087

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) possess immunosuppressive activities, which allow cancers to escape immune surveillance and become non-responsive to immune checkpoints blockade. Here we report hypoxia as a cause of MDSC accumulation. Using hepatocellular carcinoma (HCC) as a cancer model, we show that hypoxia, through stabilization of hypoxia-inducible factor-1 (HIF-1), induces ectoenzyme, ectonucleoside triphosphate diphosphohydrolase 2 (ENTPD2/CD39L1), in cancer cells, causing its overexpression in HCC clinical specimens. Overexpression of ENTPD2 is found as a poor prognostic indicator for HCC. Mechanistically, we demonstrate that ENTPD2 converts extracellular ATP to 5'-AMP, which prevents the differentiation of MDSCs and therefore promotes the maintenance of MDSCs. We further find that ENTPD2 inhibition is able to mitigate cancer growth and enhance the efficiency and efficacy of immune checkpoint inhibitors. Our data suggest that ENTPD2 may be a good prognostic marker and therapeutic target for cancer patients, especially those receiving immune therapy.Myeloid-derived suppressor cells (MDSCs) promote tumor immune escape. Here, the authors show that in hepatocellular carcinoma, hypoxia induces the expression of ENTPD2 on cancer cells leading to elevated extracellular 5'-AMP, which in turn promote the maintenance of MDSCs by preventing their differentiation.


Subject(s)
Adenosine Triphosphatases/metabolism , Carcinoma, Hepatocellular/enzymology , Hypoxia-Inducible Factor 1/metabolism , Liver Neoplasms/enzymology , Myeloid-Derived Suppressor Cells/enzymology , Adenosine Triphosphatases/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/physiopathology , Cell Differentiation , Cell Proliferation , Humans , Hypoxia/enzymology , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1/genetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/metabolism
5.
J Clin Invest ; 127(5): 1856-1872, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28394261

ABSTRACT

Cancer cells preferentially utilize glucose and glutamine, which provide macromolecules and antioxidants that sustain rapid cell division. Metabolic reprogramming in cancer drives an increased glycolytic rate that supports maximal production of these nutrients. The folate cycle, through transfer of a carbon unit between tetrahydrofolate and its derivatives in the cytoplasmic and mitochondrial compartments, produces other metabolites that are essential for cell growth, including nucleotides, methionine, and the antioxidant NADPH. Here, using hepatocellular carcinoma (HCC) as a cancer model, we have observed a reduction in growth rate upon withdrawal of folate. We found that an enzyme in the folate cycle, methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L), plays an essential role in support of cancer growth. We determined that MTHFD1L is transcriptionally activated by NRF2, a master regulator of redox homeostasis. Our observations further suggest that MTHFD1L contributes to the production and accumulation of NADPH to levels that are sufficient to combat oxidative stress in cancer cells. The elevation of oxidative stress through MTHFD1L knockdown or the use of methotrexate, an antifolate drug, sensitizes cancer cells to sorafenib, a targeted therapy for HCC. Taken together, our study identifies MTHFD1L in the folate cycle as an important metabolic pathway in cancer cells with the potential for therapeutic targeting.


Subject(s)
Aminohydrolases/metabolism , Carcinoma, Hepatocellular/enzymology , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/metabolism , Liver Neoplasms/enzymology , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multienzyme Complexes/metabolism , Neoplasm Proteins/metabolism , Aminohydrolases/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Folic Acid/genetics , Formate-Tetrahydrofolate Ligase/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/genetics
6.
Hepatology ; 64(3): 797-813, 2016 09.
Article in English | MEDLINE | ID: mdl-27228567

ABSTRACT

UNLABELLED: A population of stromal cells, myeloid-derived suppressor cells (MDSCs), is present in tumors. Though studies have gradually revealed the protumorigenic functions of MDSCs, the molecular mechanisms guiding MDSC recruitment remain largely elusive. Hypoxia, O2 deprivation, is an important factor in the tumor microenvironment of solid cancers, whose growth often exceeds the growth of functional blood vessels. Here, using hepatocellular carcinoma as the cancer model, we show that hypoxia is an important driver of MDSC recruitment. We observed that MDSCs preferentially infiltrate into hypoxic regions in human hepatocellular carcinoma tissues and that hypoxia-induced MDSC infiltration is dependent on hypoxia-inducible factors. We further found that hypoxia-inducible factors activate the transcription of chemokine (C-C motif) ligand 26 in cancer cells to recruit chemokine (C-X3-C motif) receptor 1-expressing MDSCs to the primary tumor. Knockdown of chemokine (C-C motif) ligand 26 in cancer cells profoundly reduces MDSC recruitment, angiogenesis, and tumor growth. Therapeutically, blockade of chemokine (C-C motif) ligand 26 production in cancer cells by the hypoxia-inducible factor inhibitor digoxin or blockade of chemokine (C-X3-C motif) receptor 1 in MDSCs by chemokine (C-X3-C motif) receptor 1 neutralizing antibody could substantially suppress MDSC recruitment and tumor growth. CONCLUSION: This study unprecedentedly reveals a novel molecular mechanism by which cancer cells direct MDSC homing to primary tumor and suggests that targeting MDSC recruitment represents an attractive therapeutic approach against solid cancers. (Hepatology 2016;64:797-813).


Subject(s)
Carcinoma, Hepatocellular/metabolism , Chemokines, CC/metabolism , Hypoxia/metabolism , Liver Neoplasms, Experimental/metabolism , Myeloid-Derived Suppressor Cells/physiology , Animals , Base Sequence , CX3C Chemokine Receptor 1 , Cell Line, Tumor , Chemokine CCL26 , Digoxin , Humans , Hypoxia-Inducible Factor 1/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Molecular Sequence Data , Neovascularization, Pathologic , Receptors, Chemokine/antagonists & inhibitors , Tumor Microenvironment
7.
Clin Cancer Res ; 22(12): 3105-17, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26819450

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC) lacks effective curative therapy. Hypoxia is commonly found in HCC. Hypoxia elicits a series of protumorigenic responses through hypoxia-inducible factor-1 (HIF1). Better understanding of the metabolic adaptations of HCC cells during hypoxia is essential to the design of new therapeutic regimen. EXPERIMENTAL DESIGN: Expressions of genes involved in the electron transport chain (ETC) in HCC cell lines (20% and 1% O2) and human HCC samples were analyzed by transcriptome sequencing. Expression of NDUFA4L2, a less active subunit in complex I of the ETC, in 100 pairs of HCC and nontumorous liver tissues were analyzed by qRT-PCR. Student t test and Kaplan-Meier analyses were used for clinicopathologic correlation and survival studies. Orthotopic HCC implantation model was used to evaluate the efficiency of HIF inhibitor. RESULTS: NDUFA4L2 was drastically overexpressed in human HCC and induced by hypoxia. NDUFA4L2 overexpression was closely associated with tumor microsatellite formation, absence of tumor encapsulation, and poor overall survival in HCC patients. We confirmed that NDUFA4L2 was HIF1-regulated in HCC cells. Inactivation of HIF1/NDUFA4L2 increased mitochondrial activity and oxygen consumption, resulting in ROS accumulation and apoptosis. Knockdown of NDUFA4L2 markedly suppressed HCC growth and metastasis in vivo HIF inhibitor, digoxin, significantly suppressed growth of tumors that expressed high level of NDUFA4L2. CONCLUSIONS: Our study has provided the first clinical relevance of NDUFA4L2 in human cancer and suggested that HCC patients with NDUFA4L2 overexpression may be suitable candidates for HIF inhibitor treatment. Clin Cancer Res; 22(12); 3105-17. ©2016 AACR.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/pathology , NADH Dehydrogenase/metabolism , Oxidative Stress/genetics , Animals , Apoptosis/physiology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Digoxin/pharmacology , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microsatellite Repeats/genetics , Middle Aged , Mitochondria/metabolism , NADH Dehydrogenase/genetics , Oxidation-Reduction , Oxygen Consumption/physiology , RNA Interference , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
8.
Proc Natl Acad Sci U S A ; 113(6): E725-34, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26811478

ABSTRACT

Cancer cells experience an increase in oxidative stress. The pentose phosphate pathway (PPP) is a major biochemical pathway that generates antioxidant NADPH. Here, we show that transketolase (TKT), an enzyme in the PPP, is required for cancer growth because of its ability to affect the production of NAPDH to counteract oxidative stress. We show that TKT expression is tightly regulated by the Nuclear Factor, Erythroid 2-Like 2 (NRF2)/Kelch-Like ECH-Associated Protein 1 (KEAP1)/BTB and CNC Homolog 1 (BACH1) oxidative stress sensor pathway in cancers. Disturbing the redox homeostasis of cancer cells by genetic knockdown or pharmacologic inhibition of TKT sensitizes cancer cells to existing targeted therapy (Sorafenib). Our study strengthens the notion that antioxidants are beneficial to cancer growth and highlights the therapeutic benefits of targeting pathways that generate antioxidants.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Oxidative Stress , Transketolase/metabolism , Animals , Base Sequence , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Knockdown Techniques , Glucose/metabolism , Glutathione/metabolism , Glycolysis/drug effects , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Metabolome/drug effects , Mice, Nude , Molecular Sequence Data , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Oxidative Stress/drug effects , Pentose Phosphate Pathway/drug effects , Peroxides/pharmacology , Phenylurea Compounds/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Sorafenib , Transketolase/antagonists & inhibitors , Transketolase/genetics , Up-Regulation/drug effects
9.
PLoS One ; 9(12): e115036, 2014.
Article in English | MEDLINE | ID: mdl-25541689

ABSTRACT

Hepatocellular carcinoma (HCC) is an aggressive tumor, with a high mortality rate due to late symptom presentation and frequent tumor recurrences and metastasis. It is also a rapidly growing tumor supported by different metabolic mechanisms; nevertheless, the biological and molecular mechanisms involved in the metabolic reprogramming in HCC are unclear. In this study, we found that pyruvate kinase M2 (PKM2) was frequently over-expressed in human HCCs and its over-expression was associated with aggressive clinicopathological features and poor prognosis of HCC patients. Furthermore, knockdown of PKM2 suppressed aerobic glycolysis and cell proliferation in HCC cell lines in vitro. Importantly, knockdown of PKM2 hampered HCC growth in both subcutaneous injection and orthotopic liver implantation models, and reduced lung metastasis in vivo. Of significance, PKM2 over-expression in human HCCs was associated with a down-regulation of a liver-specific microRNA, miR-122. We further showed that miR-122 interacted with the 3UTR of the PKM2 gene. Re-expression of miR-122 in HCC cell lines reduced PKM2 expression, decreased glucose uptake in vitro, and suppressed HCC tumor growth in vivo. Our clinical data and functional studies have revealed a novel biological mechanism involved in HCC metabolic reprogramming.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/secondary , Membrane Proteins/metabolism , MicroRNAs/genetics , Thyroid Hormones/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Glycolysis , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental , Lung Neoplasms/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Nude , MicroRNAs/metabolism , Prognosis , Thyroid Hormones/genetics , Thyroid Hormone-Binding Proteins
10.
Hepatology ; 60(5): 1645-58, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25048396

ABSTRACT

UNLABELLED: Poor prognosis of cancers, including hepatocellular carcinoma (HCC), is mainly associated with metastasis; however, the underlying mechanisms remain poorly understood. This article investigates the role of lysyl oxidase-like 2 (LOXL-2) in the biology of HCC metastasis. First, we showed that HCC metastasis relies on a collagen-modifying enzyme, LOXL2, which was significantly overexpressed in tumorous tissues and sera of HCC patients, indicating that LOXL2 may be a good diagnostic marker for HCC patients. Second, we delineated a complex, interlinked signaling network that involves multiple regulators, including hypoxia, transforming growth factor beta (TGF-ß), and microRNAs (miRNAs), converging to control the expression of LOXL2. We found not only that LOXL2 was regulated by hypoxia/hypoxia-inducible factor 1 alpha (HIF-1α), but also that TGF-ß activated LOXL2 transcription through mothers against decapentaplegic homolog 4 (Smad4), whereas two frequently underexpressed miRNA families, miR-26 and miR-29, cooperatively suppressed LOXL2 transcription through interacting with the 3' untranslated region of LOXL2. Third, we demonstrated the imperative roles of LOXL2 in modifying the extracellular matrix components in the tumor microenvironment and metastatic niche of HCC. LOXL2 promoted intrahepatic metastasis by increasing tissue stiffness, thereby enhancing the cytoskeletal reorganization of HCC cells. Furthermore, LOXL2 facilitated extrahepatic metastasis by enhancing recruitment of bone-marrow-derived cells to the metastatic site. CONCLUSION: These findings integrate the clinical relevance, molecular regulation, and functional implications of LOXL2 in HCC metastasis.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms, Experimental/enzymology , Animals , Case-Control Studies , Cell Adhesion , Cell Line, Tumor , Collagen/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Neoplasm Metastasis , Smad4 Protein/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...