sphingosine-1-phosphate and Liver-Neoplasms

sphingosine-1-phosphate has been researched along with Liver-Neoplasms* in 20 studies

Reviews

3 review(s) available for sphingosine-1-phosphate and Liver-Neoplasms

ArticleYear
The role of glucosylceramide and glucosylceramide synthase in liver disease: from bench to bedside - Review.
    Acta biochimica Polonica, 2020, Dec-27, Volume: 68, Issue:1

    The cell membrane, which is lipid-rich, is not only a simple mechanical barrier but also an important and complex component of the cell. It also communicates with the external environment. Sphingomyelin is an important class of phospholipids in the membrane that performs many functions. Interest in sphingomyelin-based liposomes, which are a critical component of cell membranes, have become the focus of intense study in recent years. Through additional research, the function of sphingomyelin and its derivatives in diseases can be gradually elucidated. Sphingomyelin consists of ceramide and its derivatives including ceramide-1-phosphate glucosylceramide and sphingosine-1-phosphate. The metabolism of glucosylceramide is regulated by glucosylceramide synthase (EC: 2.4.1.80) which is the key enzyme in the glycosylation of ceramide. The activity of glucosylceramide synthase directly affects the level of glucosylceramide in cells which in turn affects the function of cells and may eventually lead to diseases. Recently, the relationship between glucosylceramide and its metabolic enzymes, with diseases has become a relatively new area of study. The purpose of this paper is to address the relationship between glucosylceramide, glucosylceramide synthase, and their possible association with liver diseases at the theoretical level.

    Topics: Apoptosis; Carcinoma, Hepatocellular; Cell Membrane; Ceramides; Glucosylceramides; Glucosyltransferases; Hepatocytes; Humans; Liver Cirrhosis; Liver Neoplasms; Lung Injury; Lysophospholipids; Sphingomyelins; Sphingosine

2020
Role of Sphingosine Kinase 1 and Sphingosine-1-Phosphate Axis in Hepatocellular Carcinoma.
    Handbook of experimental pharmacology, 2020, Volume: 259

    Hepatocellular carcinoma (HCC) is primarily diagnosed in the latter stages of disease progression and is the third leading cause of cancer deaths worldwide. Thus, there is a need to find biomarkers of early HCC as well as the development of more effective treatments for the disease. Sphingosine-1-phosphate (S1P) is a pleiotropic lipid signaling molecule produced by two isoforms of sphingosine kinase (SphK1 and SphK2) that is involved in regulation of many aspects of mammalian physiology and pathophysiology, including inflammation, epithelial and endothelial barrier function, cancer, and metastasis, among many others. Abundant evidence indicates that SphK1 and S1P promote cancer progression and metastasis in multiple types of cancers. However, the role of SphK/S1P in HCC is less well studied. Here, we review the current state of knowledge of SphKs and S1P in HCC, including evidence for the correlation of SphK1 expression and S1P levels with progression of HCC and negative outcomes, and discuss how this information could lead to the design of more effective diagnostic and treatment modalities for HCC.

    Topics: Animals; Carcinoma, Hepatocellular; Liver Neoplasms; Lysophospholipids; Phosphotransferases (Alcohol Group Acceptor); Sphingosine

2020
DNA damage response and sphingolipid signaling in liver diseases.
    Surgery today, 2016, Volume: 46, Issue:9

    Patients with unresectable hepatocellular carcinoma (HCC) cannot generally be cured by systemic chemotherapy or radiotherapy due to their poor response to conventional therapeutic agents. The development of novel and efficient targeted therapies to increase their treatment options depends on the elucidation of the molecular mechanisms that underlie the pathogenesis of HCC. The DNA damage response (DDR) is a network of cell-signaling events that are triggered by DNA damage. Its dysregulation is thought to be one of the key mechanisms underlying the generation of HCC. Sphingosine-1-phosphate (S1P), a lipid mediator, has emerged as an important signaling molecule that has been found to be involved in many cellular functions. In the liver, the alteration of S1P signaling potentially affects the DDR pathways. In this review, we explore the role of the DDR in hepatocarcinogenesis of various etiologies, including hepatitis B and C infection and non-alcoholic steatohepatitis. Furthermore, we discuss the metabolism and functions of S1P that may affect the hepatic DDR. The elucidation of the pathogenic role of S1P may create new avenues of research into therapeutic strategies for patients with HCC.

    Topics: Adaptor Proteins, Signal Transducing; Carcinoma, Hepatocellular; DNA Damage; Hepatitis B; Hepatitis C; Humans; Liver Neoplasms; Lyases; Lysophospholipids; Molecular Targeted Therapy; Non-alcoholic Fatty Liver Disease; Phosphoric Monoester Hydrolases; Phosphotransferases (Alcohol Group Acceptor); Signal Transduction; Sphingosine

2016

Other Studies

17 other study(ies) available for sphingosine-1-phosphate and Liver-Neoplasms

ArticleYear
Conjugated Bile Acids Accelerate Progression of Pancreatic Cancer Metastasis via S1PR2 Signaling in Cholestasis.
    Annals of surgical oncology, 2023, Volume: 30, Issue:3

    Pancreatic cancer (PC) has an extremely high mortality rate, where obstructive jaundice due to cholestasis is a classic symptom. Conjugated bile acids (CBAs) such as taurocholic acid (TCA) have been reported to activate both the ERK1/2 and AKT signaling pathways via S1P receptor 2 (S1PR2) and promote growth of cholangiocarcinoma. Thus, we hypothesize that CBAs, which accumulate in cholestasis, accelerate PC progression via S1PR2.. Murine Panc02-luc and human AsPC-1, MIA PaCa2, and BxPC-3 cells were treated with TCA, S1PR2 agonist CYM5520, S1PR2 antagonist JTE-013, sphingosine-1-phosphate (S1P), and functional S1P receptor antagonist (except S1PR2) FTY720. Bile duct ligation (BDL) was performed on liver implantation or intraperitoneal injection of Panc02-luc cells.. Panc02-luc and AsPC-1 cells predominantly expressed S1PR2, and their growth and migration were stimulated by TCA or CYM5520 in dose-dependent manner, which was blocked by JTE-013. This finding was not seen in PC cell lines expressing other S1P receptors than S1PR2. Panc02-luc growth stimulation by S1P was not blocked by FTY720. BDL significantly increased PC liver metastasis compared with sham. PC peritoneal carcinomatosis was significantly worsened by BDL, confirmed by number of nodules, tumor weight, bioluminescence, Ki-67 stain, ascites, and worse survival compared with sham. CYM5520 significantly worsened PC carcinomatosis, whereas treatment with anti-S1P antibody or FTY720 also worsened progression.. CBAs accelerated growth of S1PR2 predominant PC both in vitro and in vivo. This finding implicates S1PR2 as a potential therapeutic target in metastatic S1PR2 predominant pancreatic cancer.

    Topics: Animals; Bile Acids and Salts; Bile Duct Neoplasms; Bile Ducts, Intrahepatic; Cholestasis; Fingolimod Hydrochloride; Humans; Liver Neoplasms; Mice; Pancreatic Neoplasms; Receptors, Lysosphingolipid; Sphingosine-1-Phosphate Receptors; Steroids

2023
Sphingosine-1-phosphate transporter spinster homolog 2 is essential for iron-regulated metastasis of hepatocellular carcinoma.
    Molecular therapy : the journal of the American Society of Gene Therapy, 2022, 02-02, Volume: 30, Issue:2

    Iron dyshomeostasis is associated with hepatocellular carcinoma (HCC) development. However, the role of iron in HCC metastasis is unknown. This study aimed to elucidate the underlying mechanisms of iron's enhancement activity on HCC metastasis. In addition to the HCC cell lines and clinical samples in vitro, iron-deficient (ID) mouse models were generated using iron-free diet and transferrin receptor protein knockout, followed by administration of HCC tumors through either orthotopic or ectopic route. Clinical metastatic HCC samples showed significant ID status, accompanied by overexpression of sphingosine-1-phosphate transporter spinster homolog 2 (SPNS2). Mechanistically, ID increased SPNS2 expression, leading to HCC metastasis in both cell cultures and mouse models. ID not only altered the anti-tumor immunity, which was indicated by phenotypes of lymphatic subsets in the liver and lung of tumor-bearing mice, but also promoted HCC metastasis in a cancer cell autonomous manner through the SPNS2. Since germline knockout of globe SPNS2 showed significantly reduced HCC metastasis, we further developed hepatic-targeting recombinant adeno-associated virus vectors to knockdown SPNS2 expression and to inhibit iron-regulated HCC metastasis. Our observation indicates the role of iron in HCC pulmonary metastasis and suggests SPNS2 as a potential therapeutic target for the prevention of HCC pulmonary metastasis.

    Topics: Animals; Anion Transport Proteins; Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Movement; Iron; Liver Neoplasms; Lysophospholipids; Mice; Neoplasm Metastasis; Sphingosine

2022
Sphingosine-1-phosphate promotes tumor development and liver fibrosis in mouse model of congestive hepatopathy.
    Hepatology (Baltimore, Md.), 2022, Volume: 76, Issue:1

    Chronic liver congestion reflecting right-sided heart failure (RHF), Budd-Chiari syndrome, or Fontan-associated liver disease (FALD) is involved in liver fibrosis and HCC. However, molecular mechanisms of fibrosis and HCC in chronic liver congestion remain poorly understood.. Here, we first demonstrated that chronic liver congestion promoted HCC and metastatic liver tumor growth using murine model of chronic liver congestion by partial inferior vena cava ligation (pIVCL). As the initial step triggering HCC promotion and fibrosis, gut-derived lipopolysaccharide (LPS) appeared to induce LSECs capillarization in mice and in vitro. LSEC capillarization was also confirmed in patients with FALD. Mitogenic factor, sphingosine-1-phosphate (S1P), was increased in congestive liver and expression of sphingosine kinase 1, a major synthetase of S1P, was increased in capillarized LSECs after pIVCL. Inhibition of S1P receptor (S1PR) 1 (Ex26) and S1PR2 (JTE013) mitigated HCC development and liver fibrosis, respectively. Antimicrobial treatment lowered portal blood LPS concentration, LSEC capillarization, and liver S1P concentration accompanied by reduction of HCC development and fibrosis in the congestive liver.. In conclusion, chronic liver congestion promotes HCC development and liver fibrosis by S1P production from LPS-induced capillarized LSECs. Careful treatment of both RHF and liver cancer might be necessary for patients with RHF with primary or metastatic liver cancer.

    Topics: Animals; Carcinoma, Hepatocellular; Disease Models, Animal; Fibrosis; Heart Failure; Humans; Lipopolysaccharides; Liver Cirrhosis; Liver Neoplasms; Lysophospholipids; Mice; Receptors, Lysosphingolipid; Sphingosine; Vascular Diseases

2022
Sphingosine 1-phosphate lyase facilitates cancer progression through converting sphingolipids to glycerophospholipids.
    Clinical and translational medicine, 2022, Volume: 12, Issue:9

    In addition to potent agonist properties for sphingosine 1-phosphate (S1P) receptors, intracellularly, S1P is an intermediate in metabolic conversion pathway from sphingolipids to glycerolysophospholipids (glyceroLPLs). We hypothesized that this S1P metabolism and its products might possess some novel roles in the pathogenesis of cancer, where S1P lyase (SPL) is a key enzyme.. The mRNA levels of sphingolipid-related and other cancer-related factors were measured in human hepatocellular carcinoma (HCC), colorectal cancer, and esophageal cancer patients' tumours and in their adjacent non-tumour tissues. Phospholipids (PL) and glyceroLPLs were measured by using liquid chromatography-tandem mass spectrometry (LC-MS/MS). In-vitro experiments were performed in Colon 26 cell line with modulation of the SPL and GPR55 expressions. Xenograft model was used for determination of the cancer progression and for pharmacological influence.. Besides high SPL levels in human HCC and colon cancer, SPL levels were specifically and positively linked with levels of glyceroLPLs, including lysophosphatidylinositol (LPI). Overexpression of SPL in Colon 26 cells resulted in elevated levels of LPI and lysophosphatidylglycerol (LPG), which are agonists of GPR55. SPL overexpression-enhanced cell proliferation was inhibited by GPR55 silencing. Conversely, inhibition of SPL led to the opposite outcome and reversed by adding LPI, LPG, and metabolites generated during S1P degradation, which is regulated by SPL. The xenograft model results suggested the contribution of SPL and glyceroLPLs to tumour progression depending on levels of SPL and GPR55. Moreover, the pharmacological inhibition of SPL prevented the progression of cancer. The underlying mechanisms for the SPL-mediated cancer progression are the activation of p38 and mitochondrial function through the LPI, LPG-GPR55 axis and the suppression of autophagy in a GPR55-independent manner.. A new metabolic pathway has been proposed here in HCC and colon cancer, SPL converts S1P to glyceroLPLs, mainly to LPI and LPG, and facilitates cancer development.

    Topics: Carcinoma, Hepatocellular; Chromatography, Liquid; Colonic Neoplasms; Glycerophospholipids; Humans; Liver Neoplasms; Lysophospholipids; RNA, Messenger; Sphingolipids; Sphingosine; Tandem Mass Spectrometry

2022
Sphingosine 1-phosphate regulates proliferation, cell cycle and apoptosis of hepatocellular carcinoma cells via syndecan-1.
    Progress in biophysics and molecular biology, 2019, Volume: 148

    Sphingosine 1-phosphate (S1P) plays an important role in hepatocarcinogenesis. We previously demonstrated that S1P induced epithelial-mesenchymal transition of hepatocellular carcinoma (HCC) cells via an MMP-7/Syndecan-1/TGF-β autocrine loop. In the present study, we investigated the regulative role of S1P in cell survival and progression of HCC cells, and tested whether syndecan-1 is required in the S1P action. After transfected with syndecan-1 shRNA, HepG2 and SMMC7721 cells were treated with S1P for 72 h, and then cell proliferation was detected by CCK8 assay, and cell cycle progression and cell apoptosis were detected by flow cytometry. The levels of apoptosis markers including cleaved-Caspase-3 and cleaved-PARP in SMMC7721 cells were examined by western blotting. Results showed that S1P significantly enhanced cell proliferation in HCC cells, which was significantly inhibited by syndecan-1 shRNA. S1P induced the cell proportion in S phase in HCC cells, whereas S1P decreased the proportion of cells in both early and late apoptosis. Syndecan-1 shRNA induced the G2/M arrest in the presence of S1P. In the syndecan-1 shRNA transfected HCC cells, the proportions of late and early apoptotic cells, and levels of cleaved-Caspase-3 and cleaved-PARP were significantly increased in cells with or without S1P treatment. Thus, S1P augments the proportion of cells in S phase of the cell cycle that might translate to enhance HCC cell proliferation and inhibit the cell apoptosis via syndecan-1.

    Topics: Apoptosis; Carcinoma, Hepatocellular; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Gene Silencing; Hep G2 Cells; Humans; Liver Neoplasms; Lysophospholipids; Sphingosine; Syndecan-1

2019
S1P Stimulates Proliferation by Upregulating CTGF Expression through S1PR2-Mediated YAP Activation.
    Molecular cancer research : MCR, 2018, Volume: 16, Issue:10

    Dysregulation of the Hippo pathway in the liver results in overgrowth and eventually tumorigenesis. To date, several upstream mechanisms have been identified that affect the Hippo pathway, which ultimately regulate YAP, the major downstream effector of the pathway. However, upstream regulators of the Hippo pathway in the liver remain poorly defined. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite that has been shown to stimulate hepatocellular carcinoma (HCC) cell proliferation, but whether the Hippo pathway is involved in S1P-stimulated HCC cell proliferation remains to be determined. Here it is demonstrated that S1P activates YAP and that the S1P receptor 2 (S1PR2/S1P2) mediates S1P-induced YAP activation in both human and mouse HCC cells. S1P promotes YAP-mediated upregulation of cysteine-rich protein 61 and connective tissue growth factor (CTGF), and stimulates HCC cell proliferation. By using siRNA-mediated knockdown approaches, only CTGF was required for S1P-stimulated cell proliferation. Of note, S1P activates YAP in a MST1/2-independent manner suggesting that the canonical Hippo kinase is not required for S1P-mediated proliferation in liver. The upregulation of CTGF and S1P2 were also observed in liver-specific YAP overexpression transgenic mouse hepatocytes. Moreover, YAP regulated liver differentiation-dependent gene expression by influencing the chromatin binding of HNF4α based on ChIP-seq analysis. Finally, results using gain- and loss-of-function approaches demonstrate that HNF4α negatively regulated S1P-induced CTGF expression.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Carcinoma, Hepatocellular; Cell Proliferation; Connective Tissue Growth Factor; Gene Expression Regulation, Neoplastic; Hepatocyte Nuclear Factor 4; Hepatocytes; Humans; Liver; Liver Neoplasms; Lysophospholipids; Mice; Mice, Transgenic; Phosphoproteins; Receptors, Lysosphingolipid; RNA, Small Interfering; Sphingosine; Sphingosine-1-Phosphate Receptors; Transcription Factors; YAP-Signaling Proteins

2018
Alkaline ceramidase 3 promotes growth of hepatocellular carcinoma cells via regulating S1P/S1PR2/PI3K/AKT signaling.
    Pathology, research and practice, 2018, Volume: 214, Issue:9

    Hepatocellular carcinoma (HCC) is one of the cancer types with poor prognosis. To effectively treat HCC, new molecular targets and therapeutic approaches must be identified. Alkaline ceramidase 3 (Acer3) hydrolyzed long-chain unsaturated ceramide to produce free fatty acids and sphingosine. However, whether and how Acer3 modulates progression of HCC remains largely unknown.. Acer3 mRNA levels in different types of human HCC samples or normal tissues were determined from Gene Expression across Normal and Tumor tissue (GENT) database. The expression level of Acer3 in human HCC cell lines were examined by western blot. Overall survival and disease-free survival of HCC patients were determined by Kaplan-Meier analysis. Effects of Acer3 knockdown by lentivirus infection were evaluated on cell growth and apoptosis. The mechanisms involved in HCC cells growth and apoptosis were analyzed by western blot.. In silico analysis of TCGA databases of HCC patients showed that the expression of Acer3 significantly inversely correlates with the overall and disease-free survival of HCC patients. Knockdown expression of Acer3 resulted in decreased cell growth and increased apoptosis. Notably, inhibition of Acer3 resulted in intracellular exhaustion of Sphingosine-1-phosphate (S1P) and inhibited activation of S1PR2/PI3K/AKT signaling. Finally, knockdown of Acer3 induced up-regulation of Bax and down-regulation of Bcl-2.. Our study suggests that Acer3 contributes to HCC propagation, and suggests that inhibition of Acer3 may be novel strategy for treating human HCC.

    Topics: Alkaline Ceramidase; Apoptosis; Carcinoma, Hepatocellular; Cell Proliferation; Disease-Free Survival; Humans; Kaplan-Meier Estimate; Liver Neoplasms; Lysophospholipids; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Receptors, Lysosphingolipid; Signal Transduction; Sphingosine; Sphingosine-1-Phosphate Receptors

2018
Sphingosine 1-phosphate (S1P) reduces hepatocyte growth factor-induced migration of hepatocellular carcinoma cells via S1P receptor 2.
    PloS one, 2018, Volume: 13, Issue:12

    A bioactive lipid, sphingosine 1-phosphate (S1P), acts extracellularly as a potent mediator, and is implicated in the progression of various cancers including hepatocellular carcinoma (HCC). S1P exerts its functions by binding to five types of specific receptors, S1P receptor 1 (S1PR1), S1PR2, S1PR3, S1PR4 and S1PR5 on the plasma membrane. However, the exact roles of S1P and each S1PR in HCC cells remain to be clarified. In the present study, we investigated the effect of S1P on the hepatocyte growth factor (HGF)-induced migration of human HCC-derived HuH7 cells, and the involvement of each S1PR. S1P dose-dependently reduced the HGF-induced migration of HuH7 cells. We found that all S1PRs exist in the HuH7 cells. Among each selective agonist for five S1PRs, CYM5520, a selective S1PR2 agonist, significantly suppressed the HGF-induced HuH7 cell migration whereas selective agonists for S1PR1, S1PR3, S1PR4 or S1PR5 failed to affect the migration. The reduction of the HGF-induced migration by S1P was markedly reversed by treatment of JTE013, a selective antagonist for S1PR2, and S1PR2- siRNA. These results strongly suggest that S1P reduces the HGF-induced HCC cell migration via S1PR2. Our findings may provide a novel potential of S1PR2 to therapeutic strategy for metastasis of HCC.

    Topics: Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Movement; Hepatocyte Growth Factor; Humans; Liver Neoplasms; Lysophospholipids; Receptors, Lysosphingolipid; RNA, Small Interfering; Sphingosine

2018
Melatonin prevents deregulation of the sphingosine kinase/sphingosine 1-phosphate signaling pathway in a mouse model of diethylnitrosamine-induced hepatocellular carcinoma.
    Journal of pineal research, 2017, Volume: 62, Issue:1

    The sphingosine kinase (SphK)/sphingosine 1-phosphate (S1P) pathway is involved in multiple biological processes, including carcinogenesis. Melatonin shows beneficial effects in cell and animal models of hepatocellular carcinoma, but it is unknown if they are associated with the modulation of the SphK/S1P system, along with different downstream signaling pathways modified in cancer. We investigated the effects of melatonin in mice which received diethylnitrosamine (DEN) (35 mg/kg body weight i.p) once a week for 8 weeks. Melatonin was given at 5 or 10 mg/kg/day i.p. beginning 4 weeks after the onset of DEN administration and ending at the sacrifice time (10, 20, 30, or 40 weeks). Melatonin alleviated the distortion of normal hepatic architecture, lowered the incidence of preneoplastic/neoplastic lesions, and inhibited the expression of proliferative/cell cycle regulatory proteins (Ki67, PCNA, cyclin D1, cyclin E, CDK4, and CDK6). S1P levels and expression of SphK1, SphK2, and S1P receptors (S1PR1/S1PR3) were significantly elevated in DEN-treated mice. However, there was a decreased expression of S1P lyase. These effects were significantly abrogated in a time- and dose-dependent manner by melatonin, which also increased S1PR2 expression. Following DEN treatment, mice exhibited increased phosphorylation of PI3K, AKT, mTOR, STAT3, ERK, and p38, and a higher expression of NF-κB p50 and p65 subunits. Melatonin administration significantly inhibited those changes. Data obtained suggest a contribution of the SphK/S1P system and related signaling pathways to the protective effects of melatonin in hepatocarcinogenesis.

    Topics: Animals; Blotting, Western; Carcinogens; Carcinoma, Hepatocellular; Diethylnitrosamine; Disease Models, Animal; Immunohistochemistry; Liver Neoplasms; Lysophospholipids; Male; Melatonin; Mice; Mice, Inbred ICR; Phosphotransferases (Alcohol Group Acceptor); Real-Time Polymerase Chain Reaction; Signal Transduction; Sphingosine

2017
Increased mRNA Levels of Sphingosine Kinases and S1P Lyase and Reduced Levels of S1P Were Observed in Hepatocellular Carcinoma in Association with Poorer Differentiation and Earlier Recurrence.
    PloS one, 2016, Volume: 11, Issue:2

    Although sphingosine 1-phosphate (S1P) has been reported to play an important role in cancer pathophysiology, little is known about S1P and hepatocellular carcinoma (HCC). To clarify the relationship between S1P and HCC, 77 patients with HCC who underwent surgical treatment were consecutively enrolled in this study. In addition, S1P and its metabolites were quantitated by LC-MS/MS. The mRNA levels of sphingosine kinases (SKs), which phosphorylate sphingosine to generate S1P, were increased in HCC tissues compared with adjacent non-HCC tissues. Higher mRNA levels of SKs in HCC were associated with poorer differentiation and microvascular invasion, whereas a higher level of SK2 mRNA was a risk factor for intra- and extra-hepatic recurrence. S1P levels, however, were unexpectedly reduced in HCC compared with non-HCC tissues, and increased mRNA levels of S1P lyase (SPL), which degrades S1P, were observed in HCC compared with non-HCC tissues. Higher SPL mRNA levels in HCC were associated with poorer differentiation. Finally, in HCC cell lines, inhibition of the expression of SKs or SPL by siRNA led to reduced proliferation, invasion and migration, whereas overexpression of SKs or SPL enhanced proliferation. In conclusion, increased SK and SPL mRNA expression along with reduced S1P levels were more commonly observed in HCC tissues compared with adjacent non-HCC tissues and were associated with poor differentiation and early recurrence. SPL as well as SKs may be therapeutic targets for HCC treatment.

    Topics: Aldehyde-Lyases; Carcinoma, Hepatocellular; Cell Differentiation; Cell Line, Tumor; Cell Movement; Cell Proliferation; Gene Expression Regulation, Neoplastic; Humans; Liver Neoplasms; Lysophospholipids; Metabolome; Neoplasm Invasiveness; Neoplasm Recurrence, Local; Phosphotransferases (Alcohol Group Acceptor); Risk Factors; RNA, Messenger; Sphingosine

2016
Serum sphingolipidomic analyses reveal an upregulation of C16-ceramide and sphingosine-1-phosphate in hepatocellular carcinoma.
    Oncotarget, 2016, Apr-05, Volume: 7, Issue:14

    We have recently shown that major alterations of serum sphingolipid metabolites in chronic liver disease associate significantly with the stage of liver fibrosis in corresponding patients. In the current study we assessed via mass spectrometry serum concentrations of sphingolipid metabolites in a series of 122 patients with hepatocellular carcinoma (HCC) compared to an age- and sex-matched series of 127 patients with cirrhosis. We observed a highly significant upregulation of long and very long chain ceramides (C16-C24) in the serum of patients with HCC as compared to patients with cirrhosis (P < 0.001). Accordingly, dihydro-ceramides, synthetic precursors of ceramides and notably sphingosine, sphingosine-1-phosphate (S1P) and sphinganine-1-phosphate (SA1P) were upregulated in patients with HCC (P < 0.001). Especially the diagnostic accuracy of C16-ceramide and S1P, assessed by receiver operating curve (ROC) analysis, showed a higher area under the curve (AUC) value as compared to alpha fetoprotein (AFP) (0.999 and 0.985 versus 0.823, P < 0.001 respectively). In conclusion, serum levels of sphingolipid metabolites show a significant upregulation in patients with HCC as compared to patients with cirrhosis. Particularly C16-ceramide and S1P may serve as novel diagnostic markers for the identification of HCC in patients with liver diseases. Our data justify further investigations on the role of sphingolipids in HCC.

    Topics: Adult; Aged; Aged, 80 and over; Biomarkers, Tumor; Carcinoma, Hepatocellular; Ceramides; Female; Humans; Liver Cirrhosis; Liver Neoplasms; Lysophospholipids; Male; Middle Aged; Sphingosine; Up-Regulation

2016
Aldo-keto Reductase Family 1 Member B 10 Mediates Liver Cancer Cell Proliferation through Sphingosine-1-Phosphate.
    Scientific reports, 2016, Mar-07, Volume: 6

    AKR1B10 is involved in hepatocarcinogenesis via modulation of fatty acid and lipid synthesis. AKR1B10 inhibition results in apoptosis of tumor cells whose lipids, especially phospholipids, were decreased by over 50%, suggesting involvement of phospholipids like sphingosine-1-phosphate (S1P) in AKR1B10's oncogenic function. Using a co-culture system, we found that co-culture of QSG-7701 (human hepatocyte) with HepG2 (hepatoma cell line) increases QSG-7701's proliferation, in which AKR1B10-S1P signaling plays a pivotal role. Consistent with previous findings, AKR1B10 mRNA and protein levels were higher in primary hepatocellular carcinoma (PHC) tissues than in peri-tumor tissues. Interestingly, the level of S1P was also higher in PHC tissues than in peri-tumor tissues. After analyzing the correlation between AKR1B10 mRNA expression in PHC tissues and the clinical data, we found that AKR1B10 mRNA expression was associated with serum alpha-fetoprotein (AFP), tumor-node-metastasis (TNM) stage, and lymph node metastasis, but not with other clinicopathologic variables. A higher AKR1B10 mRNA expression level is related to a shorter DFS (disease free survival) and OS (overall survival), serving as an independent predictor of DFS and OS in PHC patients with surgical resection.

    Topics: Aldehyde Reductase; Aldo-Keto Reductases; Cell Line, Tumor; Cell Proliferation; Coculture Techniques; Hepatocytes; Humans; Liver Neoplasms; Lysophospholipids; Sphingosine

2016
FTY720P inhibits hepatic Na(+)-K(+) ATPase via S1PR2 and PGE2.
    Biochemistry and cell biology = Biochimie et biologie cellulaire, 2016, Volume: 94, Issue:4

    Sphingosine-1-phosphate (S1P) was found previously to inhibit Na(+)-K(+) ATPase in HepG2 cells. Whether fingolimod (FTY720), a S1P receptor (S1PR) agonist, similarly inhibits the ATPase is a question that needs to be addressed. The aim of this work was to study the effect of FTY720P, the active form of the drug, on the activity of Na(+)-K(+) ATPase in HepG2 cells and determine its mechanism of action. The activity of the ATPase was assayed by measuring the amount of inorganic phosphate liberated in the presence and the absence of ouabain. FTY720-P (7.5 nmol/L, 15 min) significantly reduced the activity of the ATPase. This effect disappeared completely in the presence of JTE-013, which is a specific blocker of sphingosine-1-phosphate receptor 2 (S1PR2), as well as in the presence of calphostin and indomethacin, which are inhibitors of protein kinase C (PKC) and COX-2, respectively. The effect of FTY720P was mimicked by prostaglandin E2 (PGE2) and PMA, but abrogated by NF-κB inhibition. When NF-κB was inhibited, the effect of exogenous PGE2 still appeared, but that of PMA did not manifest, suggesting that NF-κB is upstream of PGE2 and downstream of PKC. It was concluded that FTY720P activates via S1PR2, PKC, and NF-κB. The latter induces PGE2 generation and inhibits Na(+)-K(+) ATPase.

    Topics: Blotting, Western; Carcinoma, Hepatocellular; Dinoprostone; Humans; Liver Neoplasms; Lysophospholipids; NF-kappa B; Organophosphates; Protein Kinase C; Receptors, Lysosphingolipid; Sodium-Potassium-Exchanging ATPase; Sphingosine; Sphingosine-1-Phosphate Receptors; Tumor Cells, Cultured

2016
Sphingosine-1-phosphate induced epithelial-mesenchymal transition of hepatocellular carcinoma via an MMP-7/ syndecan-1/TGF-β autocrine loop.
    Oncotarget, 2016, Sep-27, Volume: 7, Issue:39

    Sphingosine-1-phosphate (S1P) induces epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma (HCC). However, its underlying mechanism remains largely unknown. In the present study, we investigated the correlation between S1P and syndecan-1 in HCC, the molecular mechanism involved, as well as their roles in EMT of HCC. Results revealed a high serum S1P level presents in patients with HCC, which positively correlated with the serum syndecan-1 level. A significant inverse correlation existed between S1P1 and syndecan-1 in HCC tissues. S1P elicits activation of the PI3K/AKT signaling pathways via S1P1, which triggers HPSE, leading to increases in expression and activity of MMP-7 and leading to shedding and suppression of syndecan-1. The loss of syndecan-1 causes an increase in TGF-β1 production. The limited chronic increase in TGF-β1 can convert HCC cells into a mesenchymal phenotype via establishing an MMP-7/Syndecan-1/TGF-β autocrine loop. Finally, TGF-β1 and syndecan-1 are essential for S1P-induced epithelial to mesenchymal transition. Taken together, our study demonstrates that S1P induces advanced tumor phenotypes of HCC via establishing an MMP-7/syndecan-1/TGF-β1 autocrine loop, and implicates targetable S1P1-PI3K/AKT-HPSE-MMP-7 signaling axe in HCC metastasis.

    Topics: Aged; Carcinoma, Hepatocellular; Cell Line, Tumor; Enzyme Inhibitors; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Hep G2 Cells; Humans; Liver Neoplasms; Lysophospholipids; Male; Matrix Metalloproteinase 7; Middle Aged; Phenotype; Signal Transduction; Sphingosine; Syndecan-1; Transforming Growth Factor beta1

2016
Immunohistochemical detection of sphingosine-1-phosphate and sphingosine kinase-1 in human tissue samples.
    Methods in molecular biology (Clifton, N.J.), 2012, Volume: 874

    Sphingosine-1-phosphate (S1P) and the enzyme primarily responsible for its production, sphingosine kinase-1 (SphK-1), are thought to be dysregulated in multiple human diseases including cancer, multiple sclerosis (MS), diabetes, neurological diseases, fibrosis, and certain pathologies associated with impaired angiogenesis such as, age-related macular degeneration (AMD). Antibody-based techniques to identify and localize S1P and SphK-1 within cells and tissue specimens represent powerful tools not only to understand the biological role of these molecules but also to validate these unique in-class targets in multiple state diseases. Consequently, the potential applications of these molecules for therapy and diagnostic purposes are currently under investigation. Here, we describe two staining procedures for identification of S1P and SphK-1 in human frozen tissue samples and the challenges encountered in the process of localization in tissue samples of lipid molecules, such as S1P.

    Topics: Carcinoma, Hepatocellular; Humans; Immunohistochemistry; Liver Neoplasms; Lysophospholipids; Phosphotransferases (Alcohol Group Acceptor); Sphingosine

2012
Antitumor activity of sphingosine kinase 2 inhibitor ABC294640 and sorafenib in hepatocellular carcinoma xenografts.
    Cancer biology & therapy, 2011, Mar-01, Volume: 11, Issue:5

    The balance between the pro-apoptotic lipids ceramide and sphingosine and the pro-survival lipid sphingosine 1-phosphate (S1P) is termed the "sphingosine rheostat". Two isozymes, sphingosine kinase 1 and 2 (SK1 and SK2), are responsible for phosphorylation of pro-apoptotic sphingosine to form pro-survival S1P. We have previously reported the antitumor properties of an SK2 selective inhibitor, ABC294640, alone or in combination with the multikinase inhibitor sorafenib in mouse models of kidney carcinoma and pancreatic adenocarcinoma. Here we evaluated the combined antitumor effects of the aforementioned drug combination in two mouse models of hepatocellular carcinoma. Although combining the SK2 inhibitor, ABC294640, and sorafenib in vitro only afforded additive drug-drug effects, their combined antitumor properties in the mouse model bearing HepG2 cells mirrored effects previously observed in animals bearing kidney carcinoma and pancreatic adenocarcinoma cells. Combining ABC294640 and sorafenib led to a decrease in the levels of phosphorylated ERK in SK-HEP-1 cells, indicating that the antitumor effect of this drug combination is likely mediated through a suppression of the MAPK pathway in hepatocellular models. We also measured levels of S1P in the plasma of mice treated with two different doses of ABC294640 and sorafenib. We found decreases in the levels of S1P in plasma of mice treated daily with 100 mg/kg of ABC294640 for 5 weeks, and this decrease was not affected by co-administration of sorafenib. Taken together, these data support combining ABC294640 and sorafenib in clinical trials in HCC patients. Furthermore, monitoring levels of S1P may provide a pharmacodynamic marker of ABC294640 activity.

    Topics: Adamantane; Animals; Antineoplastic Agents; Apoptosis; Benzenesulfonates; Blotting, Western; Carcinoma, Hepatocellular; Flow Cytometry; Hep G2 Cells; Humans; Liver Neoplasms; Lysophospholipids; Mice; Mitogen-Activated Protein Kinases; Niacinamide; Phenylurea Compounds; Phosphotransferases (Alcohol Group Acceptor); Pyridines; Sorafenib; Sphingosine; Xenograft Model Antitumor Assays

2011
Induction of apoptosis by sphingosine-1-phosphate in human hepatoma cells is associated with enhanced expression of bax gene product.
    Biochemical and biophysical research communications, 1996, Dec-04, Volume: 229, Issue:1

    Sphingolipid metabolites are important regulators of cell growth and differentiation. Recent studies have suggested that sphingosine-1-phosphate, a biologically active sphingolipid metabolite, acts as a crucial messenger in apoptosis. In the present work, we examined the expression levels of the members of the bcl-2-related gene family to determine their roles in sphingosine-1-phosphate-induced apoptosis is human hepatoma cells. Our results indicate that sphinogosine-1-phosphate-induced apoptosis is associated with enhanced expression of Bax protein. Moreover, the regulation of bax gene expression by sphingosine-1-phosphate is independent of the p53 tumor suppressor.

    Topics: Apoptosis; bcl-2-Associated X Protein; bcl-X Protein; Carcinoma, Hepatocellular; DNA Fragmentation; Dose-Response Relationship, Drug; Gene Expression; Humans; Liver; Liver Neoplasms; Lysophospholipids; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-bcl-2; Sphingolipids; Sphingosine; Time Factors; Tumor Cells, Cultured

1996