Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 180 Suppl 2: S1-S22, 2023 10.
Article in English | MEDLINE | ID: mdl-38123153

ABSTRACT

The Concise Guide to PHARMACOLOGY 2023/24 is the sixth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of approximately 1800 drug targets, and about 6000 interactions with about 3900 ligands. There is an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes almost 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.16176. In addition to this overview, in which are identified 'Other protein targets' which fall outside of the subsequent categorisation, there are six areas of focus: G protein-coupled receptors, ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2023, and supersedes data presented in the 2021/22, 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.


Subject(s)
Databases, Pharmaceutical , Pharmacology , Humans , Databases, Factual , Ion Channels , Ligands , Receptors, Cytoplasmic and Nuclear
2.
Cells ; 11(13)2022 06 30.
Article in English | MEDLINE | ID: mdl-35805174

ABSTRACT

Neuroinflammation is a hallmark of many neurodegenerative diseases (NDs) and plays a fundamental role in mediating the onset and progression of disease. Microglia, which function as first-line immune guardians of the central nervous system (CNS), are the central drivers of neuroinflammation. Numerous human postmortem studies and in vivo imaging analyses have shown chronically activated microglia in patients with various acute and chronic neuropathological diseases. While microglial activation is a common feature of many NDs, the exact role of microglia in various pathological states is complex and often contradictory. However, there is a consensus that microglia play a biphasic role in pathological conditions, with detrimental and protective phenotypes, and the overall response of microglia and the activation of different phenotypes depends on the nature and duration of the inflammatory insult, as well as the stage of disease development. This review provides a comprehensive overview of current research on the various microglia phenotypes and inflammatory responses in health, aging, and NDs, with a special emphasis on the heterogeneous phenotypic response of microglia in acute and chronic diseases such as hemorrhagic stroke (HS), Alzheimer's disease (AD), and Parkinson's disease (PD). The primary focus is translational research in preclinical animal models and bulk/single-cell transcriptome studies in human postmortem samples. Additionally, this review covers key microglial receptors and signaling pathways that are potential therapeutic targets to regulate microglial inflammatory responses during aging and in NDs. Additionally, age-, sex-, and species-specific microglial differences will be briefly reviewed.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Animals , Central Nervous System/pathology , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , Phenotype
3.
Mol Pain ; 18: 17448069221107781, 2022 04.
Article in English | MEDLINE | ID: mdl-35647699

ABSTRACT

Spinal neuroinflammation plays a critical role in the genesis of neuropathic pain. Accumulating data suggest that abscisic acid (ABA), a phytohormone, regulates inflammatory processes in mammals. In this study, we found that reduction of the LANCL2 receptor protein but not the agonist ABA in the spinal cord is associated with the genesis of neuropathic pain. Systemic or intrathecal administration of ABA ameliorates the development and pre-existence of mechanical allodynia and heat hyperalgesia in animals with partial sciatic nerve ligation (pSNL). LANCL2 is expressed only in microglia in the spinal dorsal horn. Pre-emptive treatment with ABA attenuates activation of microglia and astrocytes, ERK activity, and TNFα protein abundance in the dorsal horn in rats with pSNL. These are accompanied by restoration of spinal LANCL2 protein abundance. Spinal knockdown of LANCL2 gene with siRNA recapitulates the behavioral and spinal molecular changes induced by pSNL. Activation of spinal toll-like receptor 4 (TLR4) with lipopolysaccharide leads to activation of microglia, and over production of TNFα, which are concurrently accompanied by suppression of protein levels of LANCL2 and peroxisome proliferator activated-receptor γ. These changes are ameliorated when ABA is added with LPS. The anti-inflammatory effects induced by ABA do not requires Gi protein activity. Our study reveals that the ABA/LANCL2 system is a powerful endogenous system regulating spinal neuroinflammation and nociceptive processing, suggesting the potential utility of ABA as the management of neuropathic pain.


Subject(s)
Abscisic Acid , Neuralgia , Abscisic Acid/metabolism , Abscisic Acid/pharmacology , Animals , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Lipopolysaccharides/pharmacology , Mammals , Membrane Proteins/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Plant Growth Regulators/metabolism , Plant Growth Regulators/pharmacology , Rats , Spinal Cord/metabolism , Spinal Cord Dorsal Horn/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Front Oncol ; 12: 1116014, 2022.
Article in English | MEDLINE | ID: mdl-36776369

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive form of malignant glioma. The GBM tumor microenvironment (TME) is a complex ecosystem of heterogeneous cells and signaling factors. Glioma associated macrophages and microglia (GAMs) constitute a significant portion of the TME, suggesting that their functional attributes play a crucial role in cancer homeostasis. In GBM, an elevated GAM population is associated with poor prognosis and therapeutic resistance. Neoplastic cells recruit these myeloid populations through release of chemoattractant factors and dysregulate their induction of inflammatory programs. GAMs become protumoral advocates through production a variety of cytokines, inflammatory mediators, and growth factors that can drive cancer proliferation, invasion, immune evasion, and angiogenesis. Among these inflammatory factors, cyclooxygenase-2 (COX-2) and its downstream product, prostaglandin E2 (PGE2), are highly enriched in GBM and their overexpression is positively correlated with poor prognosis in patients. Both tumor cells and GAMs have the ability to signal through the COX-2 PGE2 axis and respond in an autocrine/paracrine manner. In the GBM TME, enhanced signaling through the COX-2/PGE2 axis leads to pleotropic effects that impact GAM dynamics and drive tumor progression.

5.
Br J Pharmacol ; 178 Suppl 1: S1-S26, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34529830

ABSTRACT

The Concise Guide to PHARMACOLOGY 2021/22 is the fifth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of nearly 1900 human drug targets with an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes over 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/bph.15537. In addition to this overview, in which are identified 'Other protein targets' which fall outside of the subsequent categorisation, there are six areas of focus: G protein-coupled receptors, ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2021, and supersedes data presented in the 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.


Subject(s)
Databases, Pharmaceutical , Pharmacology , Humans , Ion Channels , Ligands , Protein Transport , Receptors, Cytoplasmic and Nuclear
6.
Cell Signal ; 83: 109974, 2021 07.
Article in English | MEDLINE | ID: mdl-33705894

ABSTRACT

Chronic activation of microglia is a driving factor in the progression of neuroinflammatory diseases, and mechanisms that regulate microglial inflammatory signaling are potential targets for novel therapeutics. Regulator of G protein Signaling 10 is the most abundant RGS protein in microglia, where it suppresses inflammatory gene expression and reduces microglia-mediated neurotoxicity. In particular, microglial RGS10 downregulates the expression of pro-inflammatory mediators including cyclooxygenase 2 (COX-2) following stimulation with lipopolysaccharide (LPS). However, the mechanism by which RGS10 affects inflammatory signaling is unknown and is independent of its canonical G protein targeted mechanism. Here, we sought to identify non-canonical RGS10 interacting partners that mediate its anti-inflammatory mechanism. Through RGS10 co-immunoprecipitation coupled with mass spectrometry, we identified STIM2, an endoplasmic reticulum (ER) localized calcium sensor and a component of the store-operated calcium entry (SOCE) machinery, as a novel RGS10 interacting protein in microglia. Direct immunoprecipitation experiments confirmed RGS10-STIM2 interaction in multiple microglia and macrophage cell lines, as well as in primary cells, with no interaction observed with the homologue STIM1. We further determined that STIM2, Orai channels, and the calcium-dependent phosphatase calcineurin are essential for LPS-induced COX-2 production in microglia, and this pathway is required for the inhibitory effect of RGS10 on COX-2. Additionally, our data demonstrated that RGS10 suppresses SOCE triggered by ER calcium depletion and that ER calcium depletion, which induces SOCE, amplifies pro-inflammatory genes. In addition to COX-2, we also show that RGS10 suppresses the expression of pro-inflammatory cytokines in microglia in response to thrombin and LPS stimulation, and all of these effects require SOCE. Collectively, the physical and functional links between RGS10 and STIM2 suggest a complex regulatory network connecting RGS10, SOCE, and pro-inflammatory gene expression in microglia, with broad implications in the pathogenesis and treatment of chronic neuroinflammation.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Gene Expression Regulation , Microglia/metabolism , RGS Proteins/metabolism , Stromal Interaction Molecule 2/metabolism , Animals , Calcium Channels/genetics , Inflammation/genetics , Inflammation/metabolism , Mice , RAW 264.7 Cells , RGS Proteins/genetics , Stromal Interaction Molecule 2/genetics
7.
Glia ; 67(3): 482-497, 2019 03.
Article in English | MEDLINE | ID: mdl-30578561

ABSTRACT

Patients receiving paclitaxel for cancer treatment often develop an acute pain syndrome (paclitaxel-associated acute pain syndrome, P-APS), which occurs immediately after paclitaxel treatment. Mechanisms underlying P-APS remain largely unknown. We recently reported that rodents receiving paclitaxel develop acute pain and activation of spinal microglial toll like receptor 4 (TLR4) by paclitaxel penetrating into the spinal cord is a critical event in the genesis of P-APS. Our current study dissected cellular and molecular mechanisms underlying the P-APS. We demonstrated that bath-perfusion of paclitaxel, at a concentration similar to that found in the cerebral spinal fluid in animals receiving i.v. paclitaxel (2 mg/kg), resulted in increased calcium activity in microglia instantly, and in astrocytes with 6 min delay. TLR4 activation in microglia by paclitaxel caused microglia to rapidly release interleukin-1ß (IL-1ß) but not tumor necrosis factor α, IL-6, or interferon-γ. IL-1ß release from microglia depended on capthepsin B. IL-1ß acted on astrocytes, leading to elevated calcium activity and suppressed glutamate uptake. IL-1ß also acted on neurons to increase presynaptic glutamate release and postsynaptic AMPA receptor activity in the spinal dorsal horn. Knockout of IL-1 receptors prevented the development of acute pain induced by paclitaxel in mice. Our study indicates that IL-1ß is a crucial molecule used by microglia to alter functions in astrocytes and neurons upon activation of TLR4 in the genesis of P-APS, and targeting the signaling pathways regulating the production and function of IL-1ß from microglia is a potential avenue for the development of analgesics for the treatment of P-APS.


Subject(s)
Antineoplastic Agents/adverse effects , Glutamic Acid/metabolism , Interleukin-1beta/metabolism , Microglia/metabolism , Paclitaxel/adverse effects , Pain/metabolism , Spinal Cord Dorsal Horn/metabolism , Animals , Calcium/metabolism , Excitatory Postsynaptic Potentials/physiology , Male , Mice , Mice, Knockout , Miniature Postsynaptic Potentials/physiology , Pain/chemically induced , Pain Measurement , Rats
8.
Mol Pharmacol ; 94(4): 1103-1113, 2018 10.
Article in English | MEDLINE | ID: mdl-30049816

ABSTRACT

The small regulator of G protein signaling protein RGS10 is a key regulator of neuroinflammation and ovarian cancer cell survival; however, the mechanism for RGS10 function in these cells is unknown and has not been linked to specific G protein pathways. RGS10 is highly enriched in microglia, and loss of RGS10 expression in microglia amplifies production of the inflammatory cytokine tumor necrosis factor α (TNFα) and enhances microglia-induced neurotoxicity. RGS10 also regulates cell survival and chemoresistance of ovarian cancer cells. Cyclooxygenase-2 (COX-2)-mediated production of prostaglandins such as prostaglandin E2 (PGE2) is a key factor in both neuroinflammation and cancer chemoresistance, suggesting it may be involved in RGS10 function in both cell types, but a connection between RGS10 and COX-2 has not been reported. To address these questions, we completed a mechanistic study to characterize RGS10 regulation of TNFα and COX-2 and to determine if these effects are mediated through a G protein-dependent mechanism. Our data show for the first time that loss of RGS10 expression significantly elevates stimulated COX-2 expression and PGE2 production in microglia. Furthermore, the elevated inflammatory signaling resulting from RGS10 loss was not affected by Gαi inhibition, and a RGS10 mutant that is unable to bind activated G proteins was as effective as wild type in inhibiting TNFα expression. Similarly, suppression of RGS10 in ovarian cancer cells enhanced TNFα and COX-2 expression, and this effect did not require Gi activity. Together, our data strongly indicate that RGS10 inhibits COX-2 expression by a G protein-independent mechanism to regulate inflammatory signaling in microglia and ovarian cancer cells.


Subject(s)
Cyclooxygenase 2/metabolism , GTP-Binding Proteins/metabolism , RGS Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Cell Survival/physiology , Cytokines/metabolism , Dinoprostone/metabolism , Female , Humans , Inflammation/metabolism , Mice , Microglia/metabolism , Ovarian Neoplasms/metabolism , Signal Transduction/physiology
9.
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
10.
Cell Signal ; 42: 77-87, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29042285

ABSTRACT

Regulators of G protein signaling (RGS) are a family of proteins classically known to accelerate the intrinsic GTPase activity of G proteins, which results in accelerated inactivation of heterotrimeric G proteins and inhibition of G protein coupled receptor signaling. RGS proteins play major roles in essential cellular processes, and dysregulation of RGS protein expression is implicated in multiple diseases, including cancer, cardiovascular and neurodegenerative diseases. The expression of RGS proteins is highly dynamic and is regulated by epigenetic, transcriptional and post-translational mechanisms. This review summarizes studies that report dysregulation of RGS protein expression in disease states, and presents examples of drugs that regulate RGS protein expression. Additionally, this review discusses, in detail, the transcriptional and post-transcriptional mechanisms regulating RGS protein expression, and further assesses the therapeutic potential of targeting these mechanisms. Understanding the molecular mechanisms controlling the expression of RGS proteins is essential for the development of therapeutics that indirectly modulate G protein signaling by regulating expression of RGS proteins.


Subject(s)
Cardiovascular Diseases/drug therapy , Drugs, Investigational/therapeutic use , Epigenesis, Genetic , Neoplasms/drug therapy , Neurodegenerative Diseases/drug therapy , Protein Processing, Post-Translational , RGS Proteins/genetics , Animals , Azacitidine/therapeutic use , Benzodiazepines/therapeutic use , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Hydroxamic Acids/therapeutic use , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Olanzapine , RGS Proteins/agonists , RGS Proteins/antagonists & inhibitors , RGS Proteins/metabolism , Signal Transduction , Vorinostat
11.
Mol Pharmacol ; 91(3): 197-207, 2017 03.
Article in English | MEDLINE | ID: mdl-28031332

ABSTRACT

RGS10 has emerged as a key regulator of proinflammatory cytokine production in microglia, functioning as an important neuroprotective factor. Although RGS10 is normally expressed in microglia at high levels, expression is silenced in vitro following activation of TLR4 receptor. Given the ability of RGS10 to regulate inflammatory signaling, dynamic regulation of RGS10 levels in microglia may be an important mechanism to tune inflammatory responses. The goals of the current study were to confirm that RGS10 is suppressed in an in vivo inflammatory model of microglial activation and to determine the mechanism for activation-dependent silencing of Rgs10 expression in microglia. We demonstrate that endogenous RGS10 is present in spinal cord microglia, and RGS10 protein levels are suppressed in the spinal cord in a nerve injury-induced neuropathic pain mouse model. We show that the histone deacetylase (HDAC) enzyme inhibitor trichostatin A blocks the ability of lipopolysaccharide (LPS) to suppress Rgs10 transcription in BV-2 and primary microglia, demonstrating that HDAC enzymes are required for LPS silencing of Rgs10 Furthermore, we used chromatin immunoprecipitation to demonstrate that H3 histones at the Rgs10 proximal promoter are deacetylated in BV-2 microglia following LPS activation, and HDAC1 association at the Rgs10 promoter is enhanced following LPS stimulation. Finally, we have shown that sphingosine 1-phosphate, an endogenous microglial signaling mediator that inhibits HDAC activity, enhances basal Rgs10 expression in BV-2 microglia, suggesting that Rgs10 expression is dynamically regulated in microglia in response to multiple signals.


Subject(s)
Gene Silencing , Histone Deacetylases/metabolism , Microglia/metabolism , RGS Proteins/genetics , Transcription, Genetic , Acetylation/drug effects , Animals , Azacitidine/pharmacology , Cell Line , Chemokine CXCL2/metabolism , Disease Models, Animal , Gene Silencing/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Inflammation/pathology , Lipopolysaccharides/pharmacology , Lysophospholipids/pharmacology , Methyltransferases/metabolism , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , RGS Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/metabolism
12.
Neuroscience ; 326: 158-169, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27058143

ABSTRACT

Emerging studies have shown that pharmacological activation of adenosine monophosphate-activated protein kinase (AMPK) produces potent analgesic effects in different animal pain models. Currently, the spinal molecular and synaptic mechanism by which AMPK regulates the pain signaling system remains unclear. To address this issue, we utilized the Cre-LoxP system to conditionally knockout the AMPKα1 gene in the nervous system of mice. We demonstrated that AMPKα1 is imperative for maintaining normal nociception, and mice deficient for AMPKα1 exhibit mechanical allodynia. This is concomitantly associated with increased glutamatergic synaptic activities in neurons located in the superficial spinal dorsal horn, which results from the increased glutamate release from presynaptic terminals and function of ligand-gated glutamate receptors at the postsynaptic neurons. Additionally, AMPKα1 knockout mice have increased activities of extracellular signal-regulated kinases (ERK) and p38 mitogen-activated protein kinases (p38), as well as elevated levels of interleukin-1ß (IL-1ß), reactive oxygen species (ROS), and heme oxygenase 1 (HO-1) in the spinal dorsal horn. Systemic administration of a non-specific ROS scavenger (phenyl-N-tert-butylnitrone, PBN) or a HO-1 activator (Cobalt protoporphyrin IX, CoPP) attenuated allodynia in AMPKα1 knockout mice. Bath-perfusion of the ROS scavenger or HO-1 activator effectively attenuated the increased ROS levels and glutamatergic synaptic activities in the spinal dorsal horn. Our findings suggest that ROS are the key down-stream signaling molecules mediating the behavioral hypersensitivity in AMPKα1 knockout mice. Thus, targeting AMPKα1 may represent an effective approach for the treatment of pathological pain conditions associated with neuroinflammation at the spinal dorsal horn.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glutamic Acid/physiology , Hyperalgesia/metabolism , Nociception/physiology , Presynaptic Terminals/metabolism , Reactive Oxygen Species/metabolism , Spinal Cord Dorsal Horn/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/physiology , Animals , Cyclic N-Oxides/pharmacology , Encephalitis/metabolism , Excitatory Postsynaptic Potentials , Female , Free Radical Scavengers/pharmacology , Heme Oxygenase-1/metabolism , Interleukin-1beta/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Knockout
13.
Future Med Chem ; 7(12): 1483-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26293348

ABSTRACT

More than 30 regulators of G protein signaling (RGS) proteins encompass the RGS protein superfamily of critical regulators essential to cellular homeostasis. There is enormous structural and functional diversity among the RGS superfamily, and as such they serve a wide range of functions in regulating cell biology and physiology. Recent evidence has suggested roles for multiple RGS proteins in cancer initiation and progression, which has prompted research toward the potential modulation of these proteins as a new approach in cancer therapy. This article will discuss basic RGS molecular pharmacology, summarize the cellular functions and epigenetic regulation of RGS10, review ovarian cancer chemotherapy and describe the role of RGS10 in ovarian cancer survival signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovary/drug effects , RGS Proteins/genetics , Animals , Antineoplastic Agents/therapeutic use , Epigenesis, Genetic , Female , GTP-Binding Proteins/metabolism , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , RGS Proteins/analysis , RGS Proteins/metabolism
14.
Cancer Lett ; 359(2): 233-40, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25600708

ABSTRACT

Nucleoside analogs are used as chemotherapeutic options for the treatment of platinum-resistant ovarian cancers. Human concentrative nucleoside transporter 1 (hCNT1) is implicated in sensitizing solid tumors to nucleoside analogs although its role in determining drug efficacy in ovarian cancers remains unclear. Here we examined the functional expression of hCNT1 and compared its contributions toward gemcitabine efficacy in histological subtypes of ovarian cancer. Radioactivity analysis identified hCNT1-mediated (3)H-gemcitabine transport in ovarian cancer cells to be significantly reduced compared with that of normal ovarian surface epithelial cells. Biochemical and immunocytochemical analysis identified that unlike normal ovarian cells which expressed high levels of hCNT1 at the apical cell surface, the transporter was either diminished in expression and/or mislocalized in cell lines of various subtypes of ovarian cancer. Retroviral expression of hCNT1 selectively rescued gemcitabine transport in cell lines representing serous, teratocarcinoma, and endometrioid subtypes, but not clear cell carcinoma (CCC). In addition, exogenous hCNT1 predominantly accumulated in intracytoplasmic vesicles in CCC suggesting defective cellular trafficking of hCNT1 as a contributing factor to transport deficiency. Despite diminution of hCNT1 transport in the majority of ovarian cancers and apparent trafficking defects with CCC, the chemotherapeutic efficacy of gemcitabine was broadly enhanced in all subtypes when delivered via engineered nanoparticles (NPs). Additionally, by bypassing the transport requirement, the delivery of a gemcitabine-cisplatin combination in NP formulation increased their synergistic interactions. These findings uncover hCNT1 as a putative determinant for nucleoside analog chemoresistance in ovarian cancer and may help rationalize drug selection and delivery strategies for various histological subtypes of ovarian cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Membrane Transport Proteins/metabolism , Antimetabolites, Antineoplastic/chemistry , Biological Transport , Cell Line, Tumor , Cisplatin/chemistry , Cisplatin/pharmacology , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Synergism , Female , Humans , Inhibitory Concentration 50 , Nanocapsules/chemistry , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Gemcitabine
15.
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
16.
Neurosignals ; 22(1): 43-51, 2014.
Article in English | MEDLINE | ID: mdl-24903911

ABSTRACT

G protein-coupled receptors (GPCRs) and their ligands are critical regulators of neural progenitor differentiation, and GPCR signaling pathways are regulated by regulator of G protein signaling (RGS) proteins. RGS protein expression is dynamically regulated, and we have recently described the epigenetic regulation of RGS transcript expression. Given the potential of RGS proteins to regulate GPCR signaling and the established role of epigenetic regulation in progenitor differentiation, we explored the impact of epigenetic regulation of RGS transcripts during in vitro differentiation of human neural progenitors. Here, we demonstrate robust upregulation of the RGS transcripts RGS4, RGS5, RGS6, RGS7, and RGS11 during neuronal differentiation, while DNA methyltransferase (DNMT) and histone deacetylase enzyme expression is suppressed during differentiation. Transcripts encoding R7 subfamily RGS proteins and the R7-binding partners R7BP and R9AP showed the greatest upregulation. Further, we showed that direct pharmacological inhibition of DNMT activity enhances expression of RGS2, RGS4, RGS5, RGS6, RGS7, RGS8, RGS9L, RGS10, and RGS14 as well as R7BP and R9AP transcripts in progenitors, consistent with regulation by DNMTs. Our results reveal marked upregulation of RGS expression during neuronal differentiation and suggest that decreased expression of DNMT enzymes during differentiation contributes to upregulation.


Subject(s)
DNA Methylation , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , RGS Proteins/genetics , Cell Differentiation/genetics , Epigenesis, Genetic , Gene Expression , Histone Deacetylases/genetics , Humans , Signal Transduction
17.
PLoS One ; 9(1): e87455, 2014.
Article in English | MEDLINE | ID: mdl-24475290

ABSTRACT

RGS10 is an important regulator of cell survival and chemoresistance in ovarian cancer. We recently showed that RGS10 transcript expression is suppressed during acquired chemoresistance in ovarian cancer. The suppression of RGS10 is due to DNA hypermethylation and histone deacetylation, two important mechanisms that contribute to silencing of tumor suppressor genes during cancer progression. Here, we fully investigate the molecular mechanisms of epigenetic silencing of RGS10 expression in chemoresistant A2780-AD ovarian cancer cells. We identify two important epigenetic regulators, HDAC1 and DNMT1, that exhibit aberrant association with RGS10 promoters in chemoresistant ovarian cancer cells. Knockdown of HDAC1 or DNMT1 expression, and pharmacological inhibition of DNMT or HDAC enzymatic activity, significantly increases RGS10 expression and cisplatin-mediated cell death. Finally, DNMT1 knock down also decreases HDAC1 binding to the RGS10 promoter in chemoresistant cells, suggesting HDAC1 recruitment to RGS10 promoters requires DNMT1 activity. Our results suggest that HDAC1 and DNMT1 contribute to the suppression of RGS10 during acquired chemoresistance and support inhibition of HDAC1 and DNMT1 as an adjuvant therapeutic approach to overcome ovarian cancer chemoresistance.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase 1/antagonists & inhibitors , Ovarian Neoplasms/metabolism , RGS Proteins/metabolism , Chromatin Immunoprecipitation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Primers/genetics , Female , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction
18.
PLoS One ; 8(3): e60185, 2013.
Article in English | MEDLINE | ID: mdl-23533674

ABSTRACT

RGS10 regulates ovarian cancer cell growth and survival, and RGS10 expression is suppressed in cell models of ovarian cancer chemoresistance. However, the mechanisms governing RGS10 expression in ovarian cancer are poorly understood. Here we report RGS10 suppression in primary ovarian cancer and CAOV-3 ovarian cancer cells compared to immortalized ovarian surface epithelial (IOSE) cells, and in A2780-AD chemoresistant cells compared to parental A2780 cells. RGS10-1 and RGS10-2 transcripts are expressed in ovarian cancer cells, but only RGS10-1 is suppressed in A2780-AD and CAOV-3 cells, and the RGS10-1 promoter is uniquely enriched in CpG dinucleotides. Pharmacological inhibition of DNA methyl-transferases (DNMTs) increased RGS10 expression, suggesting potential regulation by DNA methylation. Bisulfite sequencing analysis identified a region of the RGS10-1 promoter with significantly enhanced DNA methylation in chemoresistant A2780-AD cells relative to parental A2780 cells. DNA methylation in CAOV-3 and IOSE cells was similar to A2780 cells. More marked differences were observed in histone acetylation of the RGS10-1 promoter. Acetylated histone H3 associated with the RGS10-1 promoter was significantly lower in A2780-AD cells compared to parental cells, with a corresponding increase in histone deacetylase (HDAC) enzyme association. Similarly, acetylated histone levels at the RGS10-1 promoter were markedly lower in CAOV-3 cells compared to IOSE cells, and HDAC1 binding was doubled in CAOV-3 cells. Finally, we show that pharmacological inhibition of DNMT or HDAC enzymes in chemoresistant A2780-AD cells increases RGS10 expression and enhances cisplatin toxicity. These data suggest that histone de-acetylation and DNA methylation correlate with RGS10 suppression and chemoresistance in ovarian cancer. Markers for loss of RGS10 expression may identify cancer cells with unique response to therapeutics.


Subject(s)
DNA Methylation/genetics , Histones/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RGS Proteins/genetics , Acetylation , Cell Line, Tumor , Female , Humans
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...