Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 12(1): 27, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33414460

ABSTRACT

Senescence is an antiproliferative mechanism that can suppress tumor development and can be induced by oncogenes such as genes of the Ras family. Although studies have implicated bioactive sphingolipids (SL) in senescence, the specific mechanisms remain unclear. Here, using MCF10A mammary epithelial cells, we demonstrate that oncogenic K-Ras (Kirsten rat sarcoma viral oncogene homolog) is sufficient to induce cell transformation as well as cell senescence-as revealed by increases in the percentage of cells in the G1 phase of the cell cycle, p21WAF1/Cip1/CDKN1A (p21) expression, and senescence-associated ß-galactosidase activity (SA-ß-gal). Furthermore, oncogenic K-Ras altered SL metabolism, with an increase of long-chain (LC) C18, C20 ceramides (Cer), and very-long-chain (VLC) C22:1, C24 Cer, and an increase of sphingosine kinase 1 (SK1) expression. Since Cer and sphingosine-1-phosphate have been shown to exert opposite effects on cellular senescence, we hypothesized that targeting SK1 could enhance oncogenic K-Ras-induced senescence. Indeed, SK1 downregulation or inhibition enhanced p21 expression and SA-ß-gal in cells expressing oncogenic K-Ras and impeded cell growth. Moreover, SK1 knockdown further increased LC and VLC Cer species (C18, C20, C22:1, C24, C24:1, C26:1), especially the ones increased by oncogenic K-Ras. Fumonisin B1 (FB1), an inhibitor of ceramide synthases (CerS), reduced p21 expression induced by oncogenic K-Ras both with and without SK1 knockdown. Functionally, FB1 reversed the growth defect induced by oncogenic K-Ras, confirming the importance of Cer generation in the senescent phenotype. More specifically, downregulation of CerS2 by siRNA blocked the increase of VLC Cer (C24, C24:1, and C26:1) induced by SK1 knockdown and phenocopied the effects of FB1 on p21 expression. Taken together, these data show that targeting SK1 is a potential therapeutic strategy in cancer, enhancing oncogene-induced senescence through an increase of VLC Cer downstream of CerS2.


Subject(s)
Cellular Senescence , Ceramides/metabolism , Genes, ras , Membrane Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/physiology , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line , Humans
2.
Cureus ; 12(7): e8976, 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32775058

ABSTRACT

Amyloidosis can affect multiple organs, and involvement of the heart is the most common cause of death. Signs and symptoms vary depending upon the organ system affected by amyloid. Liver involvement is often seen, but symptoms are usually mild and nonspecific in isolated hepatic amyloidosis. None of the laboratory markers and imaging is characteristic of this condition; therefore, diagnosis is often delayed. Tissue biopsy is required for definitive diagnosis. Herein, we report a case where the patient's symptoms had been attributed to alcohol-related cirrhosis; however, further workup ultimately led to a diagnosis of systemic amyloidosis with multi-organ involvement.

3.
Clin Nucl Med ; 44(2): 150-152, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30516688

ABSTRACT

A 34-year-old man with end-stage renal failure status post rejection of a deceased donor kidney transplant presented with bone pain in the setting of elevated serum parathyroid hormone and calcium levels. A Tc-MIBI SPECT/CT was performed before planned subtotal parathyroidectomy. SPECT/CT imaging revealed a 1.9-cm anterior mediastinal lesion with radiotracer uptake on both the immediate and delayed images. Surgical pathology of the lesion showed a benign thymic cyst with no parathyroid component.


Subject(s)
Mediastinal Cyst/metabolism , Technetium Tc 99m Sestamibi/metabolism , Adult , Biological Transport , Humans , Male , Mediastinal Cyst/diagnostic imaging , Mediastinal Cyst/surgery , Parathyroidectomy , Single Photon Emission Computed Tomography Computed Tomography
4.
J Biol Chem ; 287(38): 31794-803, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22833671

ABSTRACT

Sphingosine kinase 1 (SK1) is an important enzyme involved in the production of the bioactive lipid sphingosine 1-phosphate (S1P). SK1 is overexpressed in many forms of cancer, however, the contribution of SK1 to cancer progression is still unclear. One of the best characterized mutations found in several forms of human cancer is an activating point mutation in the Ras oncogene, which disrupts its GTPase activity and leads to stimulation of the MEK/ERK pathway. Because SK1 activity and subcellular localization have been shown to be regulated by ERK, we wished to investigate the effect of oncogenic Ras, a potent activator of the Raf/MEK/ERK pathway, on the activity of SK1 and sphingolipid metabolism. Using HEK293T cells transiently transfected with the K-RasG12V oncogene and both wild type and Sphk1(-/-) mouse embryonic fibroblasts stably infected with retroviral K-RasG12V, we found that K-RasG12V increases the production of S1P and decreases the production of ceramide in a SK1-dependent manner. In addition, we found that expression of the K-RasG12V oncogene leads to plasma membrane localization of SK1 and a reduction in cytosolic levels of SK1. This effect is likely mediated by the Raf/MEK/ERK pathway as constitutively active B-Raf or MEK1 are able to activate SK1, but constitutively active Akt1 is not. We believe this research has important implications for how sphingolipids may be contributing to oncogenic transformation and provide some of the first evidence for oncogenes inducing specific changes in sphingolipid metabolism through SK1 regulation.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins/metabolism , Sphingolipids/chemistry , ras Proteins/metabolism , Animals , Cell Membrane/metabolism , Cell Transformation, Neoplastic , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phosphorylation , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Subcellular Fractions/metabolism , ras Proteins/physiology
5.
Crit Rev Biochem Mol Biol ; 46(4): 342-51, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21787121

ABSTRACT

For several decades, lipid biologists have investigated how sphingolipids contribute to physiology, cell biology, and cell fate. Foremost among these discoveries is the finding that the bioactive sphingolipids ceramide, sphingosine, and sphingosine-1-phosphate (S1P) have diverse and often opposing effects on cell fate. Interestingly, these bioactive sphingolipids can be interconverted by just a few enzymatic reactions. Therefore, much attention has been paid to the enzymes which govern these reactions with a disproportionate amount of focus on the enzyme sphingosine kinase 1 (SK1). Several studies have found that tissue expression of SK1 correlates with cancer stage, chemotherapy response, and tumor aggressiveness. In addition, overexpression of SK1 in multiple cancer cell lines increases their resistance to chemotherapy, promotes proliferation, allows for anchorage independent growth, and increases local angiogenesis. Inhibition of SK1 using either pharmacological inhibitors or by crossing SK1 null mice has shown promise in many xenograft models of cancer, as well as several genetic and chemically induced mouse models of carcinogenesis. Here, we review the majority of the evidence that suggests SK1 is a promising target for the prevention and/or treatment of various cancers. Also, we strongly advocate for further research into basic mechanisms of bioactive sphingolipid signaling, and an increased focus on the efficacy of SK inhibitors in non-xenograft models of cancer progression.


Subject(s)
Neoplasms/therapy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Sphingolipids/metabolism , Animals , Cell Proliferation , Ceramides/metabolism , Clinical Trials as Topic , Humans , Lysophospholipids/metabolism , Mice , Neoplasms/enzymology , Neoplasms, Experimental , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/therapy , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism
6.
Blood ; 117(22): 5941-52, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21527515

ABSTRACT

The mechanisms by which sphingosine kinase-1 (SK-1)/sphingosine 1-phosphate (S1P) activation contributes to imatinib resistance in chronic myeloid leukemia (CML) are unknown. We show herein that increased SK-1/S1P enhances Bcr-Abl1 protein stability, through inhibition of its proteasomal degradation in imatinib-resistant K562/IMA-3 and LAMA-4/IMA human CML cells. In fact, Bcr-Abl1 stability was enhanced by ectopic SK-1 expression. Conversely, siRNA-mediated SK-1 knockdown in K562/IMA-3 cells, or its genetic loss in SK-1(-/-) MEFs, significantly reduced Bcr-Abl1 stability. Regulation of Bcr-Abl1 by SK-1/S1P was dependent on S1P receptor 2 (S1P2) signaling, which prevented Bcr-Abl1 dephosphorylation, and degradation via inhibition of PP2A. Molecular or pharmacologic interference with SK-1/S1P2 restored PP2A-dependent Bcr-Abl1 dephosphorylation, and enhanced imatinib- or nilotinib-induced growth inhibition in primary CD34(+) mononuclear cells obtained from chronic phase and blast crisis CML patients, K562/IMA-3 or LAMA4/IMA cells, and 32Dcl3 murine progenitor cells, expressing the wild-type or mutant (Y253H or T315I) Bcr-Abl1 in situ. Accordingly, impaired SK-1/S1P2 signaling enhanced the growth-inhibitory effects of nilotinib against 32D/T315I-Bcr-Abl1-derived mouse allografts. Since SK-1/S1P/S1P2 signaling regulates Bcr-Abl1 stability via modulation of PP2A, inhibition of SK-1/S1P2 axis represents a novel approach to target wild-type- or mutant-Bcr-Abl1 thereby overcoming drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Phosphatase 2/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides , Cell Line, Tumor , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, SCID , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Piperazines/administration & dosage , Protein Phosphatase 2/genetics , Pyrimidines/administration & dosage , RNA, Small Interfering/genetics , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine/metabolism , Ubiquitination
7.
J Biol Chem ; 286(15): 13292-303, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21335555

ABSTRACT

Acid sphingomyelinase (aSMase) generates the bioactive lipid ceramide (Cer) from hydrolysis of sphingomyelin (SM). However, its precise roles in regulating specific sphingolipid-mediated biological processes remain ill defined. Interestingly, the aSMase gene gives rise to two distinct enzymes, lysosomal sphingomyelinase (L-SMase) and secretory sphingomyelinase (S-SMase) via alternative trafficking of a shared protein precursor. Previously, our laboratory identified Ser(508) as a crucial residue for the constitutive and regulated secretion of S-SMase in response to inflammatory cytokines, and demonstrated a role for S-SMase in formation of select cellular Cer species (Jenkins, R. W., Canals, D., Idkowiak-Baldys, J., Simbari, F., Roddy, P., Perry, D. M., Kitatani, K., Luberto, C., and Hannun, Y. A. (2010) J. Biol. Chem. 285, 35706-35718). In the present study using a chemokine/cytokine screen, we identified the chemokine CCL5 (formerly known as RANTES) as a candidate-specific downstream target for aSMase. Regulation of CCL5 by aSMase was subsequently validated using both loss-of-function and gain-of-function models indicating that aSMase is both necessary and sufficient for CCL5 production. Interestingly, cells deficient in acid ceramidase (aCDase) also exhibited defects in CCL5 induction, whereas cells deficient in sphingosine kinase-1 and -2 exhibited higher levels of CCL5, suggesting that sphingosine and not sphingosine 1-phosphate (S1P) is responsible for the positive signal to CCL5. Consistent with this, co-expression of aSMase and aCDase was sufficient to strongly induce CCL5. Taken together, these data identify a novel role for aSMase (particularly S-SMase) in chemokine elaboration by pro-inflammatory cytokines and highlight a novel and shared function for aSMase and aCDase.


Subject(s)
Acid Ceramidase/metabolism , Chemokine CCL5/biosynthesis , Signal Transduction/physiology , Sphingomyelin Phosphodiesterase/metabolism , Sphingosine/metabolism , Acid Ceramidase/genetics , Animals , Cell Line, Tumor , Chemokine CCL5/genetics , Farber Lipogranulomatosis/genetics , Farber Lipogranulomatosis/metabolism , Humans , Lysophospholipids/genetics , Lysophospholipids/metabolism , Mice , Mice, Knockout , Sphingomyelin Phosphodiesterase/genetics , Sphingosine/analogs & derivatives , Sphingosine/genetics
8.
Adv Exp Med Biol ; 688: 1-23, 2010.
Article in English | MEDLINE | ID: mdl-20919643

ABSTRACT

Sphingolipids constitute a class of lipids defined by their eighteen carbon amino-alcohol backbones which are synthesized in the ER from nonsphingolipid precursors. Modification of this basic structure is what gives rise to the vast family of sphingolipids that play significant roles in membrane biology and provide many bioactive metabolites that regulate cell function. Despite the diversity of structure and function of sphingolipids, their creation and destruction are governed by common synthetic and catabolic pathways. In this regard, sphingolipid metabolism can be imagined as an array of interconnected networks that diverge from a single common entry point and converge into a single common breakdown pathway. In their simplest forms, sphingosine, phytosphingosine and dihydrosphingosine serve as the backbones upon which further complexity is achieved. For example, phosphorylation of the C1 hydroxyl group yields the final breakdown products and/or the important signaling molecules sphingosine-1-phosphate, phytosphingosine-1-phosphate and dihydrosphingosine-1-phosphate, respectively. On the other hand, acylation of sphingosine, phytosphingosine, or dihydrosphingosine with one of several possible acyl CoA molecules through the action of distinct ceramide synthases produces the molecules defined as ceramide, phytoceramide, or dihydroceramide. Ceramide, due to the differing acyl CoAs that can be used to produce it, is technically a class of molecules rather than a single molecule and therefore may have different biological functions depending on the acyl chain it is composed of. At the apex of complexity is the group of lipids known as glycosphingolipids (GSL) which contain dozens of different sphingolipid species differing by both the order and type of sugar residues attached to their headgroups. Since these molecules are produced from ceramide precursors, they too may have differences in their acyl chain composition, revealing an additional layer of variation. The glycosphingolipids are divided broadly into two categories: glucosphingolipids and galactosphingolipids. The glucosphingolipids depend initially on the enzyme glucosylceramide synthase (GCS) which attaches glucose as the first residue to the C1 hydroxyl position. Galactosphingolipids, on the other hand, are generated from galactosylceramide synthase (GalCerS), an evolutionarily dissimilar enzyme from GCS. Glycosphingolipids are further divided based upon further modification by various glycosyltransferases which increases the potential variation in lipid species by several fold. Far more abundant are the sphingomyelin species which are produced in parallel with glycosphingolipids, however they are defined by a phosphocholine headgroup rather than the addition of sugar residues. Although sphingomyelin species all share a common headgroup, they too are produced from a variety of ceramide species and therefore can have differing acyl chains attached to their C-2 amino groups. Whether or not the differing acyl chain lengths in SMs dictate unique functions or important biophysical distinctions has not yet been established. Understanding the function of all the existing glycosphingolipids and sphingomyelin species will be a major undertaking in the future since the tools to study and measure these species are only beginning to be developed (see Fig 1 for an illustrated depiction of the various sphingolipid structures). The simple sphingolipids serve both as the precursors and the breakdown products of the more complex ones. Importantly, in recent decades, these simple sphingolipids have gained attention for having significant signaling and regulatory roles within cells. In addition, many tools have emerged to measure the levels of simple sphingolipids and therefore have become the focus of even more intense study in recent years. With this thought in mind, this chapter will pay tribute to the complex sphingolipids, but focus on the regulation of simple sphingolipid metabolism.


Subject(s)
Sphingolipids/metabolism , Animals , Biological Transport, Active , Ceramides/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Humans , Molecular Structure , Sphingolipids/biosynthesis , Sphingolipids/chemistry , Sphingomyelins/metabolism
9.
FASEB J ; 24(8): 2727-38, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20335228

ABSTRACT

Sphingosine-1-phosphate (S1P) is an important bioactive sphingolipid involved in angiogenesis and lymphangiogenesis, 2 important processes that influence the growth, survival, and spread of tumors. S1P acts as an extracellular mediator through binding to 5 highly specific S1P receptors, S1P(1-5). Sphingosine kinase-1 (SK1), one of 2 known sphingosine kinase enzymes responsible for S1P production, appears to be overexpressed in many tumors. Although a role for S1P in angiogenesis and lymphangiogenesis has been established, it is unclear whether S1P secreted from cancer cells has a paracrine function in a tumor environment. Here we investigated whether modulation of cellular SK1 could initiate a paracrine angiogenic and lymphangiogenic switch. We found that SK1 overexpression in HEK cells or its down-regulation in glioma or breast cancer cells modulated extracellular S1P levels accordingly, which in turn increased or decreased both migration and tube formation in cocultured vascular or lymphatic endothelial cells. In contrast, down-regulation of sphingosine kinase 2 in both glioma and breast cancer cells had no appreciable effect on cellular or secreted S1P levels. In addition, vascular endothelial growth factors VEGF and VEGF-C down-regulation in cancer cells appeared insufficient to block the angiogenic and lymphangiogenic switch triggered by these cells. Moreover, S1P initiated endothelial cell sprouting in 3-dimensional collagen matrices, which is representative of a multistep angiogenic process. Our data collectively demonstrate for the first time that SK1 plays an essential role in regulating in vitro paracrine angiogenesis and lymphangiogenesis.


Subject(s)
Lymphangiogenesis , Neovascularization, Pathologic , Neovascularization, Physiologic , Paracrine Communication/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Coculture Techniques , Endothelial Cells/cytology , Female , Gene Expression Regulation/physiology , Glioma/blood supply , Glioma/pathology , Humans , Phosphotransferases (Alcohol Group Acceptor)/genetics
10.
Mol Ther ; 17(3): 430-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19107118

ABSTRACT

Radiation resistance in a subset of prostate tumors remains a challenge to prostate cancer radiotherapy. The current study on the effects of radiation on prostate cancer cells reveals that radiation programs an unpredicted resistance mechanism by upregulating acid ceramidase (AC). Irradiated cells demonstrated limited changes of ceramide levels while elevating levels of sphingosine and sphingosine-1-phosphate. By genetically downregulating AC with small interfering RNA (siRNA), we observed radiosensitization of cells using clonogenic and cytotoxicity assays. Conversely, AC overexpression further decreased sensitivity to radiation. We also observed that radiation-induced AC upregulation was sufficient to create cross-resistance to chemotherapy as demonstrated by decreased sensitivity to Taxol and C(6) ceramide compared to controls. Lower levels of caspase 3/7 activity were detected in cells pretreated with radiation, also indicating increased resistance. Finally, utilization of the small molecule AC inhibitor, LCL385, sensitized PPC-1 cells to radiation and significantly decreased tumor xenograft growth. These data suggest a new mechanism of cancer cell resistance to radiation, through upregulation of AC that is, in part, mediated by application of the therapy itself. An improved understanding of radiotherapy and the application of combination therapy achieved in this study offer new opportunities for the modulation of radiation effects in the treatment of cancer.


Subject(s)
Acid Ceramidase/metabolism , Prostatic Neoplasms/enzymology , Radiation-Sensitizing Agents/pharmacology , Up-Regulation/drug effects , Up-Regulation/radiation effects , Acid Ceramidase/antagonists & inhibitors , Acid Ceramidase/genetics , Animals , Cell Line, Tumor , Ceramides/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Male , Mice , Mice, Nude , Myristates/pharmacology , Paclitaxel/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Propanolamines/pharmacology , Prostatic Neoplasms/genetics , RNA, Small Interfering/genetics , Sensitivity and Specificity , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Xenograft Model Antitumor Assays
11.
J Biol Chem ; 283(6): 3365-3375, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18055454

ABSTRACT

Sphingosine 1-phosphate (S1P), a sphingolipid metabolite that plays an important role in the regulation of cell survival, growth, migration, and angiogenesis, acts both inside the cells and as an extracellular mediator through binding to five G protein-coupled receptors (S1P(1-5)). Sphingosine kinase 1 (SK1), the enzyme responsible for S1P production, is overexpressed in many solid tumors, including gliomas. One common feature of these tumors is the presence of "hypoxic regions," characterized by cells expressing high levels of hypoxia-inducible factors HIF-1alpha and HIF-2alpha, two transcription regulators that modulate the levels of proteins with crucial roles in tumor progression. So far, nothing is known about the role and the regulation of SK1 during tumor-induced hypoxia or about SK1 regulation and HIFs. Here we investigated the role of HIF-1alpha and HIF-2alpha in the regulation of SK1 during hypoxic stress in glioma-derived U87MG cells. We report that hypoxia increases SK1 mRNA levels, protein expression, and enzyme activity, followed by intracellular S1P production and S1P release. Interestingly, knockdown of HIF-2alpha by small interfering RNA abolished the induction of SK1 and the production of extracellular S1P after CoCl(2) treatment, whereas HIF-1alpha small interfering RNA resulted in an increase of HIF-2alpha and of SK1 protein levels. Moreover, using chromatin immunoprecipitation analysis, we demonstrate that HIF-2alpha binds the SK1 promoter. Functionally, we demonstrate that conditioned medium from hypoxia-treated tumor cells results in neoangiogenesis in human umbilical vein endothelial cells in a S1P receptor-dependent manner. These studies provide evidence of a link between S1P production as a potent angiogenic agent and the hypoxic phenotype observed in many tumors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Brain Neoplasms/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Hypoxia-Inducible Factor 1/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Promoter Regions, Genetic , Disease Progression , Humans , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transcription, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...