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.
Methods Mol Biol ; 1697: 141-151, 2018.
Article in English | MEDLINE | ID: mdl-28361481

ABSTRACT

Sphingosine-1-phosphate (S1P) and its receptors are important in nervous system development. Reliable in vitro human model systems are needed to further define specific roles for S1P signaling in neural development. We have described S1P-regulated signaling, survival, and differentiation in a human embryonic stem cell-derived neuroepithelial progenitor cell line (hNP1) that expresses functional S1P receptors. These cells can be further differentiated to a neuronal cell type and therefore represent a good model system to study the role of S1P signaling in human neural development. The following sections describe in detail the culture and differentiation of hNP1 cells and two assays to measure S1P signaling in these cells.


Subject(s)
Cell Culture Techniques/methods , Human Embryonic Stem Cells/cytology , Lysophospholipids/metabolism , Neurons/cytology , Sphingosine/analogs & derivatives , Cell Differentiation , Cell Survival , Cells, Cultured , Human Embryonic Stem Cells/metabolism , Humans , Neurogenesis , Neurons/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/metabolism
2.
ASN Neuro ; 6(6)2014.
Article in English | MEDLINE | ID: mdl-25424429

ABSTRACT

The bioactive lysophospholipids lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P) have diverse effects on the developing nervous system and neural progenitors, but the molecular basis for their pleiotropic effects is poorly understood. We previously defined LPA and S1P signaling in proliferating human neural progenitor (hNP) cells, and the current study investigates their role in neuronal differentiation of these cells. Differentiation in the presence of LPA or S1P significantly enhanced cell survival and decreased expression of neuronal markers. Further, the LPA receptor antagonist Ki16425 fully blocked the effects of LPA, and differentiation in the presence of Ki16425 dramatically enhanced neurite length. LPA and S1P robustly activated Erk, but surprisingly both strongly suppressed Akt activation. Ki16425 and pertussis toxin blocked LPA activation of Erk but not LPA inhibition of Akt, suggesting distinct receptor and G-protein subtypes mediate these effects. Finally, we explored cross talk between lysophospholipid signaling and the cytokine leukemia inhibitory factor (LIF). LPA/S1P effects on neuronal differentiation were amplified in the presence of LIF. Similarly, the ability of LPA/S1P to regulate Erk and Akt was impacted by the presence of LIF; LIF enhanced the inhibitory effect of LPA/S1P on Akt phosphorylation, while LIF blunted the activation of Erk by LPA/S1P. Taken together, our results suggest that LPA and S1P enhance survival and inhibit neuronal differentiation of hNP cells, and LPA1 is critical for the effect of LPA. The pleiotropic effects of LPA may reflect differences in receptor subtype expression or cross talk with LIF receptor signaling.


Subject(s)
Adult Stem Cells/drug effects , Cell Differentiation/drug effects , Leukemia Inhibitory Factor/pharmacology , Lysophospholipids/pharmacology , MAP Kinase Kinase Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sphingosine/analogs & derivatives , Cell Line, Transformed , Dose-Response Relationship, Drug , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Isoxazoles/pharmacology , MAP Kinase Kinase Kinase 3/genetics , Propionates/pharmacology , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/metabolism , Sphingosine/pharmacology
3.
Methods Mol Biol ; 874: 193-200, 2012.
Article in English | MEDLINE | ID: mdl-22528449

ABSTRACT

Sphingosine-1-phosphate (S1P) and its receptors are important in nervous system development. Reliable in vitro human model systems are needed to further define specific roles for S1P signaling in neural development. We have recently reported that human embryonic stem cell-derived neuroepithelial progenitor cells (hES-NEP) express functional S1P receptors. These cells can be further differentiated to a neuronal cell type, and therefore represent a good model system to study the role of S1P signaling in human neural development. The following sections describe in detail the culture of hES-NEP cells and two assays to measure S1P signaling in these cells.


Subject(s)
Lysophospholipids/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Adenylyl Cyclases/metabolism , Cells, Cultured , Humans , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Sphingosine/metabolism
4.
Neuropharmacology ; 62(2): 988-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22016110

ABSTRACT

In vivo and in vitro studies suggest a crucial role for Sphingosine 1-phosphate (S1P) and its receptors in the development of the nervous system. Dihydrosphingosine 1-phosphate (dhS1P), a reduced form of S1P, is an agonist at S1P receptors, but the pharmacology and physiology of dhS1P has not been widely studied. The mycotoxin fumonisin B1 (FB(1)) is a potent inhibitor of ceramide synthases and causes selective accumulation of dihydrosphingosine and dhS1P. Recent studies suggest that maternal exposure to FB(1) correlates with the development of neural tube defects (NTDs) in which the neural epithelial progenitor cell layers of the developing brain fail to fuse. We hypothesize that the altered balance of S1P and dhS1P in neural epithelial cells contributes to the developmental effects of FB(1). The goal of this work was first to define the effect of FB(1) exposure on levels of sphingosine and dh-sphingosine and their receptor-active 1-phosphate metabolites in human embryonic stem cell-derived neural epithelial progenitor (hES-NEP) cells; and second, to define the relative activity of dhS1P and S1P in hES-NEP cells. We found that dhS1P is a more potent stimulator of inhibition of cAMP and Smad phosphorylation than is S1P in neural progenitors, and this difference in apparent potency may be due, in part, to more persistent presence of extracellular dhS1P applied to human neural progenitors rather than a higher activity at S1P receptors. This study establishes hES-NEP cells as a useful human in vitro model system to study the mechanism of FB(1) toxicity and the molecular pharmacology of sphingolipid signaling. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.


Subject(s)
Lysophospholipids/metabolism , Neural Stem Cells/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fumonisins/pharmacology , Humans , Neural Stem Cells/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Smad Proteins/metabolism , Sphingosine/metabolism
5.
Pharmacol Ther ; 129(3): 290-306, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21073897

ABSTRACT

Stem cell-based therapeutics have the potential to effectively treat many terminal and debilitating human diseases, but the mechanisms by which their growth and differentiation are regulated are incompletely defined. Recent data from multiple systems suggest major roles for G protein coupled receptor (GPCR) pathways in regulating stem cell function in vivo and in vitro. The goal of this review is to illustrate common ground between the growing field of stem cell therapeutics and the long-established field of G protein coupled receptor signaling. Herein, we briefly introduce basic stem cell biology and discuss how several conserved pathways regulate pluripotency and differentiation in mouse and human stem cells. We further discuss general mechanisms by which GPCR signaling may impact these pluripotency and differentiation pathways, and summarize specific examples of receptors from each of the major GPCR subfamilies that have been shown to regulate stem cell function. Finally, we discuss possible therapeutic implications of GPCR regulation of stem cell function.


Subject(s)
Cell Differentiation/physiology , Pluripotent Stem Cells/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Humans , Pluripotent Stem Cells/cytology , Receptors, G-Protein-Coupled/metabolism
6.
Mol Cancer ; 9: 289, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-21044322

ABSTRACT

BACKGROUND: A critical therapeutic challenge in epithelial ovarian carcinoma is the development of chemoresistance among tumor cells following exposure to first line chemotherapeutics. The molecular and genetic changes that drive the development of chemoresistance are unknown, and this lack of mechanistic insight is a major obstacle in preventing and predicting the occurrence of refractory disease. We have recently shown that Regulators of G-protein Signaling (RGS) proteins negatively regulate signaling by lysophosphatidic acid (LPA), a growth factor elevated in malignant ascites fluid that triggers oncogenic growth and survival signaling in ovarian cancer cells. The goal of this study was to determine the role of RGS protein expression in ovarian cancer chemoresistance. RESULTS: In this study, we find that RGS2, RGS5, RGS10 and RGS17 transcripts are expressed at significantly lower levels in cells resistant to chemotherapy compared with parental, chemo-sensitive cells in gene expression datasets of multiple models of chemoresistance. Further, exposure of SKOV-3 cells to cytotoxic chemotherapy causes acute, persistent downregulation of RGS10 and RGS17 transcript expression. Direct inhibition of RGS10 or RGS17 expression using siRNA knock-down significantly reduces chemotherapy-induced cell toxicity. The effects of cisplatin, vincristine, and docetaxel are inhibited following RGS10 and RGS17 knock-down in cell viability assays and phosphatidyl serine externalization assays in SKOV-3 cells and MDR-HeyA8 cells. We further show that AKT activation is higher following RGS10 knock-down and RGS 10 and RGS17 overexpression blocked LPA mediated activation of AKT, suggesting that RGS proteins may blunt AKT survival pathways. CONCLUSIONS: Taken together, our data suggest that chemotherapy exposure triggers loss of RGS10 and RGS17 expression in ovarian cancer cells, and that loss of expression contributes to the development of chemoresistance, possibly through amplification of endogenous AKT signals. Our results establish RGS10 and RGS17 as novel regulators of cell survival and chemoresistance in ovarian cancer cells and suggest that their reduced expression may be diagnostic of chemoresistance.


Subject(s)
Ovarian Neoplasms/metabolism , RGS Proteins/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Computational Biology , Docetaxel , Drug Resistance, Neoplasm/drug effects , Female , Humans , Lysophospholipids/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Polymerase Chain Reaction , RGS Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Taxoids/pharmacology , Vincristine/pharmacology
7.
BMC Neurosci ; 9: 118, 2008 Dec 11.
Article in English | MEDLINE | ID: mdl-19077254

ABSTRACT

BACKGROUND: Lysophospholipids regulate the morphology and growth of neurons, neural cell lines, and neural progenitors. A stable human neural progenitor cell line is not currently available in which to study the role of lysophospholipids in human neural development. We recently established a stable, adherent human embryonic stem cell-derived neuroepithelial (hES-NEP) cell line which recapitulates morphological and phenotypic features of neural progenitor cells isolated from fetal tissue. The goal of this study was to determine if hES-NEP cells express functional lysophospholipid receptors, and if activation of these receptors mediates cellular responses critical for neural development. RESULTS: Our results demonstrate that Lysophosphatidic Acid (LPA) and Sphingosine-1-phosphate (S1P) receptors are functionally expressed in hES-NEP cells and are coupled to multiple cellular signaling pathways. We have shown that transcript levels for S1P1 receptor increased significantly in the transition from embryonic stem cell to hES-NEP. hES-NEP cells express LPA and S1P receptors coupled to G i/o G-proteins that inhibit adenylyl cyclase and to G q-like phospholipase C activity. LPA and S1P also induce p44/42 ERK MAP kinase phosphorylation in these cells and stimulate cell proliferation via G i/o coupled receptors in an Epidermal Growth Factor Receptor (EGFR)- and ERK-dependent pathway. In contrast, LPA and S1P stimulate transient cell rounding and aggregation that is independent of EGFR and ERK, but dependent on the Rho effector p160 ROCK. CONCLUSION: Thus, lysophospholipids regulate neural progenitor growth and morphology through distinct mechanisms. These findings establish human ES cell-derived NEP cells as a model system for studying the role of lysophospholipids in neural progenitors.


Subject(s)
Cell Line , Embryonic Stem Cells/metabolism , Neuroepithelial Cells/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysosphingolipid/metabolism , Cell Proliferation , Embryonic Stem Cells/cytology , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Proteins/metabolism , Humans , Microscopy, Video , Neuroepithelial Cells/cytology , Neuroepithelial Cells/physiology , RNA, Messenger/metabolism , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/physiology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...