Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 81(5): 1252-1264, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33414169

ABSTRACT

Acetyl-CoA is a vitally important and versatile metabolite used for many cellular processes including fatty acid synthesis, ATP production, and protein acetylation. Recent studies have shown that cancer cells upregulate acetyl-CoA synthetase 2 (ACSS2), an enzyme that converts acetate to acetyl-CoA, in response to stresses such as low nutrient availability and hypoxia. Stressed cancer cells use ACSS2 as a means to exploit acetate as an alternative nutrient source. Genetic depletion of ACSS2 in tumors inhibits the growth of a wide variety of cancers. However, there are no studies on the use of an ACSS2 inhibitor to block tumor growth. In this study, we synthesized a small-molecule inhibitor that acts as a transition-state mimetic to block ACSS2 activity in vitro and in vivo. Pharmacologic inhibition of ACSS2 as a single agent impaired breast tumor growth. Collectively, our findings suggest that targeting ACSS2 may be an effective therapeutic approach for the treatment of patients with breast cancer. SIGNIFICANCE: These findings suggest that targeting acetate metabolism through ACSS2 inhibitors has the potential to safely and effectively treat a wide range of patients with cancer.


Subject(s)
Acetate-CoA Ligase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Acetate-CoA Ligase/genetics , Acetate-CoA Ligase/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Drug Stability , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fatty Acids/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Mice, Inbred Strains , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Br J Cancer ; 122(6): 868-884, 2020 03.
Article in English | MEDLINE | ID: mdl-31942031

ABSTRACT

BACKGROUND: Recent studies have suggested that fatty acid oxidation (FAO) is a key metabolic pathway for the growth of triple negative breast cancers (TNBCs), particularly those that have high expression of MYC. However, the underlying mechanism by which MYC promotes FAO remains poorly understood. METHODS: We used a combination of metabolomics, transcriptomics, bioinformatics, and microscopy to elucidate a potential mechanism by which MYC regulates FAO in TNBC. RESULTS: We propose that MYC induces a multigenic program that involves changes in intracellular calcium signalling and fatty acid metabolism. We determined key roles for fatty acid transporters (CD36), lipases (LPL), and kinases (PDGFRB, CAMKK2, and AMPK) that each contribute to promoting FAO in human mammary epithelial cells that express oncogenic levels of MYC. Bioinformatic analysis further showed that this multigenic program is highly expressed and predicts poor survival in the claudin-low molecular subtype of TNBC, but not other subtypes of TNBCs, suggesting that efforts to target FAO in the clinic may best serve claudin-low TNBC patients. CONCLUSION: We identified critical pieces of the FAO machinery that have the potential to be targeted for improved treatment of patients with TNBC, especially the claudin-low molecular subtype.


Subject(s)
Claudins/metabolism , Fatty Acids/metabolism , Metabolomics/methods , Proto-Oncogene Proteins c-myc/genetics , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Transfection
3.
Metabolism ; 83: 50-59, 2018 06.
Article in English | MEDLINE | ID: mdl-29410347

ABSTRACT

OBJECTIVE: As the leading risk factor for the development of liver cancer, chronic infection with hepatitis B virus (HBV) represents a significant global health concern. Although an effective HBV vaccine exists, at least 240 million people are chronically infected with HBV worldwide. Therapeutic options for the treatment of chronic HBV remain limited, and none achieve an absolute cure. To develop novel therapeutic targets, a better understanding of the complex network of virus-host interactions is needed. Because of the central metabolic role of the liver, we assessed the metabolic impact of HBV infection as a means to identify viral dependency factors and metabolic pathways that could serve as novel points of therapeutic intervention. METHODS: Primary rat hepatocytes were infected with a control adenovirus, an adenovirus expressing a greater-than-unit-length copy of the HBV genome, or an adenovirus expressing the HBV X protein (HBx). A panel of 369 metabolites was analyzed for HBV- or HBx-induced changes 24 and 48 h post infection. Pathway analysis was used to identify key metabolic pathways altered in the presence of HBV or HBx expression, and these findings were further supported through integration of publically available gene expression data. RESULTS: We observed distinct changes to multiple metabolites in the context of HBV replication or HBx expression. Interestingly, a panel of 7 metabolites (maltotriose, maltose, myristate [14:0], arachidate [20:0], 3-hydroxybutyrate [BHBA], myo-inositol, and 2-palmitoylglycerol [16,0]) were altered by both HBV and HBx at both time points. In addition, incorporation of data from a transcriptome-based dataset allowed us to identify metabolic pathways, including long chain fatty acid metabolism, glycolysis, and glycogen metabolism, that were significantly altered by HBV and HBx. CONCLUSIONS: Because the liver is a central regulator of metabolic processes, it is important to understand how HBV replication and HBV protein expression affects the metabolic function of hepatocytes. Through analysis of a broad panel of metabolites we investigated this metabolic impact. The results of these studies have defined metabolic consequences of an HBV infection of hepatocytes and will help to lay the groundwork for novel research directions and, potentially, development of novel anti-HBV therapeutics.


Subject(s)
Hepatitis B virus/pathogenicity , Hepatitis B/genetics , Hepatitis B/metabolism , Hepatocytes/physiology , Hepatocytes/virology , Metabolic Networks and Pathways , Animals , Cells, Cultured , Gene Expression Profiling , Hepatitis B/pathology , Hepatitis B virus/physiology , Hepatocytes/metabolism , Hepatocytes/pathology , Male , Metabolic Networks and Pathways/genetics , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Transcriptome
4.
Virus Res ; 246: 23-27, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29307794

ABSTRACT

Worldwide, approximately 240 million people are chronically infected with the hepatitis B virus (HBV); chronic HBV infection is associated with the development of life-threatening liver diseases. The HBV HBx protein alters hepatocyte physiology to promote HBV replication. We previously reported that HBx modulates calcium signaling to stimulate HBV replication in human hepatoblastoma HepG2 cells and primary rat hepatocytes. Whether HBx modulates calcium signaling in a primary human hepatocyte, the natural site of an HBV infection, has not been determined. Here, we report the effect of HBx on calcium signaling in primary human hepatocytes and show that HBx modulates calcium signaling via enhanced calcium entry through store-operated calcium channels and elevated mitochondrial calcium, similar to HBx effects in HepG2 cells and primary rat hepatocytes. In addition to demonstrating that HBV and HBx affect calcium signaling in human hepatocytes, these studies also show that HBV and HBx regulation of calcium signaling is identical in primary human and rat hepatocytes, further validating the use of cultured primary rat hepatocytes for HBV studies.


Subject(s)
Calcium/metabolism , Hepatitis B virus/metabolism , Hepatocytes/metabolism , Mitochondria/metabolism , Trans-Activators/metabolism , Virus Replication , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling , Gene Expression , Hepatitis B virus/genetics , Hepatitis B virus/growth & development , Hepatocytes/virology , Humans , Mitochondria/virology , Primary Cell Culture , Rats , Single-Cell Analysis , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins
5.
PLoS One ; 12(2): e0168328, 2017.
Article in English | MEDLINE | ID: mdl-28151934

ABSTRACT

Many viruses modulate calcium (Ca2+) signaling to create a cellular environment that is more permissive to viral replication, but for most viruses that regulate Ca2+ signaling, the mechanism underlying this regulation is not well understood. The hepatitis B virus (HBV) HBx protein modulates cytosolic Ca2+ levels to stimulate HBV replication in some liver cell lines. A chronic HBV infection is associated with life-threatening liver diseases, including hepatocellular carcinoma (HCC), and HBx modulation of cytosolic Ca2+ levels could have an important role in HBV pathogenesis. Whether HBx affects cytosolic Ca2+ in a normal hepatocyte, the natural site of an HBV infection, has not been addressed. Here, we report that HBx alters cytosolic Ca2+ signaling in cultured primary hepatocytes. We used single cell Ca2+ imaging of cultured primary rat hepatocytes to demonstrate that HBx elevates the cytosolic Ca2+ level in hepatocytes following an IP3-linked Ca2+ response; HBx effects were similar when expressed alone or in the context of replicating HBV. HBx elevation of the cytosolic Ca2+ level required extracellular Ca2+ influx and store-operated Ca2+ (SOC) entry and stimulated HBV replication in hepatocytes. We used both targeted RT-qPCR and transcriptome-wide RNAseq analyses to compare levels of SOC channel components and other Ca2+ signaling regulators in HBV-expressing and control hepatocytes and show that the transcript levels of these various proteins are not affected by HBV. We also show that HBx regulation of SOC-regulated Ca2+ accumulation is likely the consequence of HBV modulation of a SOC channel regulatory mechanism. In support of this, we link HBx enhancement of SOC-regulated Ca2+ accumulation to Ca2+ uptake by mitochondria and demonstrate that HBx stimulates mitochondrial Ca2+ uptake in primary hepatocytes. The results of our study may provide insights into viral mechanisms that affect Ca2+ signaling to regulate viral replication and virus-associated diseases.


Subject(s)
Calcium Signaling/physiology , Hepatitis B virus/physiology , Hepatocytes/metabolism , Hepatocytes/virology , Virus Replication/physiology , Animals , Calcium Signaling/genetics , Cells, Cultured , Hepatitis B virus/genetics , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Rats , Trans-Activators/genetics , Trans-Activators/physiology , Viral Proteins/physiology , Viral Regulatory and Accessory Proteins , Virus Replication/genetics
6.
Methods Mol Biol ; 1540: 143-155, 2017.
Article in English | MEDLINE | ID: mdl-27975314

ABSTRACT

Chronic infection with hepatitis B virus (HBV) remains a major worldwide health concern and is the leading cause of hepatocellular carcinoma (HCC). The HBV X protein (HBx) is the only regulatory protein encoded in the HBV genome; HBx stimulates HBV replication in vivo and in vitro. HBx also regulates cytosolic Ca2+ signaling, and altered Ca2+ signaling is associated with the development of many diseases, including HCC. Importantly, many HBx functions, including HBx modulation of cell proliferation, apoptosis, and transcription pathways, have been linked to changes in cytosolic Ca2+ signaling. Additionally, several stages of HBV replication, including capsid formation and activation of the HBV polymerase, are dependent on intracellular Ca2+. Consequently, defining the molecular mechanism that underlies HBV and HBx modulation of cytosolic Ca2+ levels is important for understanding HBV pathogenesis and the role of HBx in HBV replication. Here, we describe a single-cell Ca2+-imaging protocol that we use to investigate HBV and HBx effects on the level of cytosolic Ca2+. We specifically outline two methods that we use to evaluate HBV and HBx regulation of cytosolic Ca2+ levels in cultured primary hepatocytes. This protocol can also be adapted for use in liver cell lines.


Subject(s)
Calcium/metabolism , Gene Expression Regulation, Viral , Hepatitis B virus/genetics , Hepatitis B/metabolism , Hepatitis B/virology , Hepatocytes/metabolism , Hepatocytes/virology , Trans-Activators/genetics , Animals , Calcium Signaling , Cells, Cultured , Cytosol/metabolism , Humans , Microscopy, Fluorescence , Primary Cell Culture , Rats , Viral Regulatory and Accessory Proteins
7.
Sci Rep ; 6: 27965, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27328854

ABSTRACT

Alterations in N-linked glycosylation have long been associated with cancer but for the most part, the reasons why have remained poorly understood. Here we show that increased core fucosylation is associated with de-differentiation of primary hepatocytes and with the appearance of markers indicative of a transition of cells from an epithelial to a mesenchymal state. This increase in core fucosylation was associated with increased levels of two enzymes involved in α-1,6 linked fucosylation, GDP-mannose 4, 6-dehydratase (Gmds) and to a lesser extent fucosyltransferase 8 (Fut8). In addition, the activation of cancer-associated cellular signaling pathways in primary rat hepatocytes can increase core fucosylation and induce additional glycoform alterations on hepatocyte proteins. Specifically, we show that increased levels of protein sialylation and α-1,6-linked core fucosylation are observed following activation of the ß-catenin pathway. Activation of the Akt signaling pathway or induction of hypoxia also results in increased levels of fucosylation and sialylation. We believe that this knowledge will help in the better understanding of the genetic factors associated with altered glycosylation and may allow for the development of more clinically relevant biomarkers.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Dedifferentiation/physiology , Epithelial-Mesenchymal Transition/physiology , Fucosyltransferases/genetics , Hydro-Lyases/metabolism , Liver Neoplasms/pathology , beta Catenin/metabolism , Animals , Biomarkers/metabolism , Carcinoma, Hepatocellular/diagnosis , Cells, Cultured , Fucosyltransferases/metabolism , Glycosylation , Hepatocytes/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/diagnosis , Mesenchymal Stem Cells/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/physiology , Transcriptional Activation , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...