Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Mol Ther ; 24(9): 1697-706, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27357628

ABSTRACT

Loss of p53 suppressor function, through mutations or inactivation of the p53 pathway, occurs in most human cancers. SGT-53 is a liposomal nanocomplex designed for systemic, tumor-targeting delivery of the wt p53 gene. In this nanodelivery system, an anti-transferrin receptor single-chain antibody fragment serves as the targeting moiety. In an initial phase 1 trial in patients with advanced solid tumors, SGT-53 demonstrated tumor-specific targeting, was shown to be well tolerated, and was associated with an antitumor effect in several patients. Our preclinical studies have also demonstrated enhanced antitumor activity with the combination of SGT-53 and docetaxel. Thus, this dose-escalation trial was undertaken to assess the combination of SGT-53 and docetaxel for safety and potential efficacy in 14 advanced cancer patients. Results reveal that the combination of SGT-53 (maximum dose, 3.6 mg DNA/infusion) and docetaxel (75 mg/m(2)/infusion) was well tolerated. Moreover, clinical activity involving 12 evaluable patients was observed. Three of these patients achieved RECIST-verified partial responses with tumor reductions of -47%, -51%, and -79%. Two others had stable disease with significant shrinkage (-25% and -16%). These results support phase 2 testing of SGT-53 in combination with docetaxel.


Subject(s)
Genes, p53 , Liposomes , Neoplasms/drug therapy , Neoplasms/genetics , Taxoids/administration & dosage , Adult , Aged , Cohort Studies , Combined Modality Therapy , Docetaxel , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged , Nanoparticles , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/diagnosis , Retreatment , Tomography, X-Ray Computed , Treatment Outcome
2.
Mol Cell Endocrinol ; 314(2): 158-63, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-19486924

ABSTRACT

The pulsatile secretion of GnRH from normal and immortalized hypothalamic GnRH neurons is highly calcium-dependent and is stimulated by cAMP. It is also influenced by agonist activation of the endogenous GnRH receptor (GnRH-R), which couples to multiple G proteins. This autocrine mechanism could serve as a timer to determine the frequency of pulsatile GnRH release by regulating Ca(2+)- and cAMP-dependent signaling and GnRH neuronal firing. The firing of individual and/or bursts of action potentials (APs) in spontaneously active GnRH neurons is followed by afterhyperpolarization (AHP) that lasts from several milliseconds to several seconds. GnRH-induced activation of GnRH neurons causes a significant increase in medium AHP that is partially sensitive to apamin. GnRH-induced modulation of Ca(2+) influx and the consequent changes in AHP current suggest that the GnRH receptors expressed in hypothalamic GnRH neurons are important modulators of their neuronal excitability. The coexistence of multiple regulatory mechanisms could provide a high degree of redundancy in the maintenance of this crucial component of the reproductive process. It is also conceivable that this multifactorial system could reflect the gradation from simple to more complex neuroendocrine control systems for regulating hypothalamo-pituitary function and gonadal activity during the evolution of the GnRH pulse generator.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Ion Channels/physiology , Receptors, G-Protein-Coupled/physiology , Second Messenger Systems/physiology , Action Potentials/physiology , Animals , Calcium/physiology , Cell Line , Humans , Neurons/physiology , Receptors, Serotonin/physiology
3.
Trends Endocrinol Metab ; 20(8): 402-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19740674

ABSTRACT

Pulsatile secretion of gonadotropin-releasing hormone (GnRH) release is an intrinsic property of hypothalamic GnRH neurons. Pulse generation has been attributed to multiple specific mechanisms, including spontaneous electrical activity of GnRH neurons, calcium and cAMP signaling, a GnRH receptor autocrine regulatory component, a GnRH concentration-dependent switch in GnRH receptor (GnRH-R) coupling to specific G proteins, the expression of G protein-coupled receptors (GPCRs) and steroid receptors, and homologous and heterologous interactions between cell membrane receptors expressed in GnRH neurons. The coexistence of multiple regulatory mechanisms for pulsatile GnRH secretion provides a high degree of redundancy in maintaining this crucial component of the mammalian reproductive process. These studies provide insights into the basic cellular and molecular mechanisms involved in GnRH neuronal function.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Animals , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypothalamus/metabolism , Models, Biological , Pituitary Gland/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/physiology
4.
Mol Endocrinol ; 22(10): 2250-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18701637

ABSTRACT

Estradiol (E(2)) acts as a potent feedback molecule between the ovary and hypothalamic GnRH neurons, and exerts both positive and negative regulatory actions on GnRH synthesis and secretion. However, the extent to which these actions are mediated by estrogen receptors (ERs) expressed in GnRH neurons has been controversial. In this study, Single-cell RT-PCR revealed the expression of both ERalpha and ERbeta isoforms in cultured fetal and adult rat hypothalamic GnRH neurons. Both ERalpha and ERbeta or individual ERs were expressed in 94% of cultured fetal GnRH neurons. In adult female rats at diestrus, 68% of GnRH neurons expressed ERs, followed by 54% in estrus and 19% in proestrus. Expression of individual ERs was found in 24% of adult male GnRH neurons. ERalpha exerted marked G(i)-mediated inhibitory effects on spontaneous action potential (AP) firing, cAMP production, and pulsatile GnRH secretion, indicating its capacity for negative regulation of GnRH neuronal function. In contrast, increased E(2) concentration and ERbeta agonists increase the rate of AP firing, GnRH secretion, and cAMP production, consistent with ERbeta-dependent positive regulation of GnRH secretion. Consonant with the coupling of ERalpha to pertussis toxin-sensitive G(i/o) proteins, E(2) also activates G protein-activated inwardly rectifying potassium channels, decreasing membrane excitability and slowing the firing of spontaneous APs in hypothalamic GnRH neurons. These findings demonstrate that the dual actions of E(2) on GnRH neuronal membrane excitability, cAMP production, and GnRH secretion are mediated by the dose-dependent activation of ERalpha and ERbeta expressed in hypothalamic GnRH neurons.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Animals , Cells, Cultured , Cyclic AMP/metabolism , Estrogen Receptor Modulators/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrogens/metabolism , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gonadotropin-Releasing Hormone/genetics , Hypothalamus/cytology , Male , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Pregnancy , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley
5.
Int J Biochem Cell Biol ; 40(11): 2627-37, 2008.
Article in English | MEDLINE | ID: mdl-18573679

ABSTRACT

The functional activity of G protein-coupled receptors can be modified by their ability to form oligomeric complexes with G protein-coupled receptors from other receptor families. Emerging evidence suggests that the appetite-regulating hormone ghrelin is a directly acting vasodilator peptide with anti-inflammatory activity, therefore, we have examined the ability of ghrelin receptors to oligomerize with members of the prostanoid receptor family which are also involved in modulating vascular activity and inflammatory responses. Using the techniques of bioluminescence resonance energy transfer and co-immunoprecipitation, we detected the ability of ghrelin receptors to hetero-oligomerize with prostaglandin E2 receptor subtype EP3-I, prostacyclin receptors, and thromboxane A2 (TPalpha) receptors, when transiently over-expressed in human embryonic kidney 293 cells. These results suggest that hetero-oligomeric interactions between ghrelin receptors and prostanoid receptors are likely to be of biological relevance. Co-transfection of cells with ghrelin receptor and prostanoid receptors significantly decreased ghrelin receptor expression and attenuated its constitutive activation of phospholipase C without changing its affinity for ghrelin. We also observed an increase in the proportion of ghrelin receptors localized intracellularly in the presence of prostanoid receptors. Taken together, these results suggest that the increased expression of prostanoid receptors in conditions of vascular inflammation, such as in atherosclerotic plaques, could influence those cellular responses dependent on the constitutive activation of ghrelin receptors.


Subject(s)
Protein Isoforms/metabolism , Receptors, Ghrelin/metabolism , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Cell Line , Enzyme Activation , Humans , Protein Isoforms/genetics , Receptors, Epoprostenol , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Ghrelin/genetics , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP3 Subtype , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Type C Phospholipases/metabolism
6.
Mol Endocrinol ; 21(12): 3062-70, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17698953

ABSTRACT

The G protein-coupled receptor 54 (GPR54) and its endogenous ligand, kisspeptin, are essential for activation and regulation of the hypothalamic-pituitary-gonadal axis. Analysis of RNA extracts from individually identified hypothalamic GnRH neurons with primers for GnRH, kisspeptin-1, and GPR54 revealed expression of all three gene products. Also, constitutive and GnRH agonist-induced bioluminescence resonance energy transfer between Renilla luciferase-tagged GnRH receptor and GPR54 tagged with green fluorescent protein, expressed in human embryonic kidney 293 cells, revealed heterooligomerization of the two receptors. Whole cell patch-clamp recordings from identified GnRH neurons showed initial depolarizing effects of kisspeptin on membrane potential, followed by increased action potential firing. In perifusion studies, treatment of GT1-7 neuronal cells with kisspeptin-10 increased GnRH peak amplitude and duration. The production and secretion of kisspeptin in cultured hypothalamic neurons and GT1-7 cells were detected by a specific RIA and was significantly reduced by treatment with GnRH. The expression of kisspeptin and GPR54 mRNAs in identified hypothalamic GnRH neurons, as well as kisspeptin secretion, indicate that kisspeptins may act as paracrine and/or autocrine regulators of the GnRH neuron. Stimulation of GnRH secretion by kisspeptin and the opposing effects of GnRH on kisspeptin secretion indicate that GnRH receptor/GnRH and GPR54/kisspeptin autoregulatory systems are integrated by negative feedback to regulate GnRH and kisspeptin secretion from GnRH neurons.


Subject(s)
Gene Expression Regulation , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cells, Cultured , Electrophysiology , Female , Fluorescence Resonance Energy Transfer , Gonadotropin-Releasing Hormone/genetics , Humans , Kisspeptins , Mice , Patch-Clamp Techniques , Proteins/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1 , Tumor Suppressor Proteins/genetics
7.
Cell Signal ; 19(5): 1011-22, 2007 May.
Article in English | MEDLINE | ID: mdl-17229547

ABSTRACT

The dimerization properties of the ghrelin receptor (GRLN-R) and its non-signalling, naturally occurring, truncated splice variant (GHS-R1b) have been investigated in human embryonic kidney 293 cells heterologously expressing these proteins. Using the techniques of bioluminescence resonance energy transfer and co-immunoprecipitation, we detected the formation of GRLN-R homodimers and GRLN-R/GHS-R1b heterodimers, but ghrelin-induced conformational changes were only detected in the GRLN-R homodimers. When the expression of GHS-R1b exceeded that of GRLN-R, there was a decrease in the cell surface expression of GRLN-R with a consequent decrease in constitutive activation of phosphatidylinositol-specific phospholipase C (PI-PLC). Furthermore, there was no change in ghrelin affinity, and the efficacy of cell signalling as measured by stimulation of PI-PLC and extracellular signal-regulated kinase 1/2 was unchanged. Cellular localization studies suggest that GRLN-R is normally distributed between the plasma membrane and cytosolic fractions, but in the presence of GHS-R1b, GRLN-R is localized to the nucleus. Therefore, we propose that the decrease in GRLN-R constitutive signalling was due to translocation of GRLN-R to the nucleus due to the formation of GRLN-R/GHS-R1b heterodimers. Therefore, GHS-R1b appears to act as a dominant-negative mutant of the full-length GRLN-R.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Alternative Splicing , Cell Line , Dimerization , Fluorescence Resonance Energy Transfer/methods , Gene Expression , Humans , Immunoprecipitation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Ghrelin , Transfection
8.
Int J Biochem Cell Biol ; 39(4): 752-64, 2007.
Article in English | MEDLINE | ID: mdl-17169600

ABSTRACT

In addition to regulating growth hormone release from the pituitary, ghrelin receptors also influence cell proliferation and apoptosis. By studying mitogen-activated protein kinase activity in human embryonic kidney 293 cells over-expressing ghrelin receptors, we aimed to identify the specific cell signalling pathways used by ghrelin receptors, and to determine if the truncated ghrelin receptor polypeptide had any influence on the functional activity of ghrelin receptors. We found that ghrelin activated extracellular signal-regulated kinases 1/2 with an EC50 value of 10 nM, and that this response was inhibited by the ghrelin receptor antagonists D-Lys3-GHRP-6 and [D-Arg1,D-Phe5,D-Trp(7,9),Leu11]-substance P. Ghrelin had little or no effect on the activity of c-Jun N-terminal kinase, p38 kinase or Akt. Ghrelin appeared to activate extracellular signal-regulated kinases 1/2 through a calcium-independent novel protein kinase C isoform which may utilize diacylglycerol derived from hydrolysis of phosphatidylcholine rather than from phosphatidylinositol. Ghrelin-stimulated extracellular signal-regulated kinases 1/2 activity was independent of transactivation of epidermal growth factor receptors, and even when ghrelin receptor internalization was blocked by concanavalin A or a beta-arrestin mutant, there was no decrease in phosphorylated extracellular signal-regulated kinases 1/2, suggesting this is a G protein-dependent process. The truncated ghrelin receptor polypeptide had no effect on ghrelin receptor signalling to extracellular signal-regulated kinases 1/2, but decreased the constitutive activation of phosphatidylinositol-specific phospholipase C by ghrelin receptors. In conclusion, our results suggest that any up-regulation of the truncated ghrelin receptor polypeptide might preferentially attenuate functional activity dependent on the constitutive activation of ghrelin receptors, while leaving ghrelin-dependent signalling unaffected.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Peptide Hormones/pharmacology , Phosphatidylinositol Diacylglycerol-Lyase/metabolism , Receptors, G-Protein-Coupled/metabolism , Blotting, Western , Cell Line , Concanavalin A/pharmacology , Dose-Response Relationship, Drug , Endocytosis/drug effects , Enzyme Activation/drug effects , Gene Expression , Ghrelin , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Oligopeptides/pharmacology , Phosphoinositide Phospholipase C , Phosphorylation/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, Ghrelin , Substance P/analogs & derivatives , Substance P/pharmacology , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Cell Mol Immunol ; 2(3): 181-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16212885

ABSTRACT

Coumarins belong to a diverse group of naturally occurring non-nutrient phytochemicals known as benzo-alpha-pyrones. In this study, esculetin, a 6,7-dihydroxy derivative of coumarin with pleiotropic biological activities, was found to have no significant cytotoxic effect on normal murine macrophages, but it could increase the in vivo migration of the thioglycollate-elicited macrophages in a dose-dependent manner. Moreover, esculetin significantly increased the endocytic activity, and augmented the nitric oxide production and iNOS gene expression in LPS-treated macrophages. In addition, in vivo administration of esculetin into mice was shown to increase the mitogenesis of splenic lymphocytes towards Con A and LPS stimulations, and induced the LAK activity of splenic lymphocytes. Collectively, our results indicate that esculetin could exert immunomodulatory effects on murine macrophages and lymphocytes, both in vitro and in vivo, and this might be one of the possible mechanisms by which coumarins can exert their chemopreventive and anti-tumor activities in vivo.


Subject(s)
Lymphocytes/drug effects , Lymphocytes/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Umbelliferones/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation , Concanavalin A/pharmacology , Coumarins/pharmacology , Endocytosis/drug effects , Gene Expression Regulation/drug effects , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/drug effects , Spleen/metabolism , Umbelliferones/chemistry
10.
FEBS Lett ; 577(1-2): 147-53, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15527776

ABSTRACT

We have recently cloned the full-length cDNAs of the two growth hormone secretagogue receptor (GHSR) subtypes from a teleost species, the black seabream (Acanthopagrus schlegeli) [Mol. Cell. Endocrinol. 214 (2004) 81], namely sbGHSR-1a and sbGHSR-1b. Functional expression of these two receptor constructs in human embryonic kidney 293 (HEK293) cells indicated that stimulation of sbGHSR-1a by growth hormone secretagogues (GHS) could evoke increases in intracellular Ca2+ concentration ([Ca2+]i), whereas sbGHSR-1b appeared to play an inhibitory role on the signal transduction activity of sbGHSR-1a. In the present study, we have further investigated the signal transduction mechanism of sbGHSR-1a. The peptide GHS GHRP-6 and the non-peptide GHS L163,540 were able to trigger a receptor specific and phospholipase C (PLC)-dependent elevation of [Ca2+]i in HEK293 cells stably expressing sbGHSR-1a. This GHS-induced calcium mobilization was also dependent on protein kinase C activated L-type calcium channel opening. It was found that sbGHSR-1a could function in an agonist-independent manner as it exhibited a high basal activity of inositol phosphate production in the absence of GHS, indicating that the fish receptor is constitutively active. In addition, the extracellular signal-regulated kinases 1 and 2 (ERK1/2) were found to be activated upon stimulation of sbGHSR-1a by GHRP-6. This observation provides direct evidence in the coupling of sbGHSR-1a to ERK1/2 activation. Neither Gs nor Gi proteins are coupled to the receptor, as GHS did not induce cAMP production nor inhibit forskolin-stimulated cAMP accumulation in the sbGHSR-1a bearing cells. Furthermore, the ability of the GHSR antagonist D-Lys3-GHRP-6 to inhibit basal PLC and basal ERK1/2 activity suggests that this compound is an inverse agonist. In summary, the sbGHSR-1a appears to couple through the G(q/11)-mediated pathway to activate PLC, resulting in increased IP3 production and Ca2+ mobilization from both intracellular and extracellular stores. Moreover, sbGHSR-1a may trigger multiple signal transduction cascades to exert its physiological functions.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Animals , Calcium/metabolism , Cell Line , Cyclic AMP/biosynthesis , Humans , Inositol Phosphates/metabolism , Protein Kinase C/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Ghrelin , Sea Bream , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...