Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299224

ABSTRACT

Inflammation is important for the initiation and progression of breast cancer. We have previously reported that in monocytes, estrogen regulates TLR4/NFκB-mediated inflammation via the interaction of the Erα isoform ERα36 with GPER1. We therefore investigated whether a similar mechanism is present in breast cancer epithelial cells, and the effect of ERα36 expression on the classic 66 kD ERα isoform (ERα66) functions. We report that estrogen inhibits LPS-induced NFκB activity and the expression of downstream molecules TNFα and IL-6. In the absence of ERα66, ERα36 and GPER1 are both indispensable for this effect. In the presence of ERα66, ERα36 or GPER1 knock-down partially inhibits NFκB-mediated inflammation. In both cases, ERα36 overexpression enhances the inhibitory effect of estrogen on inflammation. We also verify that ERα36 and GPER1 physically interact, especially after LPS treatment, and that GPER1 interacts directly with NFκB. When both ERα66 and ERα36 are expressed, the latter acts as an inhibitor of ERα66 via its binding to estrogen response elements. We also report that the activation of ERα36 leads to the inhibition of breast cancer cell proliferation. Our data support that ERα36 is an inhibitory estrogen receptor that, in collaboration with GPER1, inhibits NFκB-mediated inflammation and ERα66 actions in breast cancer cells.


Subject(s)
Estrogen Receptor alpha/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Breast Neoplasms , Cell Line, Tumor , Epithelial Cells/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Estrogens/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inflammation/metabolism , Interleukin-6/metabolism , MCF-7 Cells , Monocytes/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Estrogen/physiology , Receptors, G-Protein-Coupled/physiology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 891-899, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29287776

ABSTRACT

BACKGROUND & AIMS: TGFß superfamily member Activin-A is a multifunctional hormone/cytokine expressed in multiple tissues and cells, where it regulates cellular differentiation, proliferation, inflammation and tissue architecture. High activin-A levels have been reported in alcoholic cirrhosis and non-alcoholic steatohepatitis (NASH). Our aim was to identify the cell types involved in the fibrotic processes induced by activin-A in liver and verify the liver diseases that this molecule can be found increased. METHODS: We studied the effect of activin-A on mouse primary Kupffer cells (KCs) and Hepatic Stellate cells (HSCs) and the levels of activin-A and its inhibitor follistatin in the serum of patients from a large panel of liver diseases. RESULTS: Activin-A is expressed by mouse hepatocytes, HSCs and Liver Sinusoid Endothelial cells but not KCs. Each cell type expresses different activin receptor combinations. HSCs are unresponsive to activin-A due to downregulation/desensitization of type-II activin receptors, while KCs respond by increasing the expression/production of TNFα και TGFß1. In the presence of KCs or conditioned medium from activin-A treated KCs, HSCs switch to a profibrogenic phenotype, including increased collagen and αSMA expression and migratory capacity. Incubation of activin-A treated KC conditioned medium with antibodies against TNFα and TGFß1 partially blocks its capacity to activate HSCs. Only patients with alcoholic liver diseases and NASH cirrhosis have significantly higher activin-A levels and activin-A/follistatin ratio. CONCLUSIONS: Activin-A may induce fibrosis in NASH and alcoholic cirrhosis via activation of KCs to express pro-inflammatory molecules that promote HSC-dependent fibrogenesis and could be a target for future anti-fibrotic therapies.


Subject(s)
Activins/physiology , Hepatic Stellate Cells/metabolism , Kupffer Cells/metabolism , Liver/pathology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Activins/genetics , Activins/metabolism , Aged , Animals , Case-Control Studies , Fibrosis/genetics , Fibrosis/metabolism , Humans , Kupffer Cells/pathology , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred BALB C , Middle Aged , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
3.
Neural Plast ; 2016: 8373020, 2016.
Article in English | MEDLINE | ID: mdl-26881135

ABSTRACT

The functional significance of cannabinoids in ocular physiology and disease has been reported some decades ago. In the early 1970s, subjects who smoked Cannabis sativa developed lower intraocular pressure (IOP). This led to the isolation of phytocannabinoids from this plant and the study of their therapeutic effects in glaucoma. The main treatment of this disease to date involves the administration of drugs mediating either the decrease of aqueous humour synthesis or the increase of its outflow and thus reduces IOP. However, the reduction of IOP is not sufficient to prevent visual field loss. Retinal diseases, such as glaucoma and diabetic retinopathy, have been defined as neurodegenerative diseases and characterized by ischemia-induced excitotoxicity and loss of retinal neurons. Therefore, new therapeutic strategies must be applied in order to target retinal cell death, reduction of visual acuity, and blindness. The aim of the present review is to address the neuroprotective and therapeutic potential of cannabinoids in retinal disease.


Subject(s)
Cannabinoids/therapeutic use , Retinal Diseases/drug therapy , Animals , Cannabinoids/administration & dosage , Cannabinoids/metabolism , Disease Models, Animal , Endocannabinoids/metabolism , Glaucoma/metabolism , Humans , Intraocular Pressure/drug effects , Neuroprotective Agents , Receptors, Cannabinoid/metabolism , Retina/drug effects , Retina/metabolism , Retinal Diseases/metabolism , Retinal Ganglion Cells/metabolism
4.
J Leukoc Biol ; 99(2): 333-47, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26394816

ABSTRACT

Estrogens are known modulators of monocyte/macrophage functions; however, the underlying mechanism has not been clearly defined. Recently, a number of estrogen receptor molecules and splice variants were identified that exert different and sometimes opposing actions. We assessed the expression of estrogen receptors and explored their role in mediating estrogenic anti-inflammatory effects on human primary monocytes. We report that the only estrogen receptors expressed are estrogen receptor-α 36-kDa splice variant and G-protein coupled receptor 30/G-protein estrogen receptor 1, in a sex-independent manner. 17-ß-Estradiol inhibits the LPS-induced IL-6 inflammatory response, resulting in inhibition of NF-κB transcriptional activity. This is achieved via a direct physical interaction of ligand-activated estrogen receptor-α 36-kDa splice variant with the p65 component of NF-κB in the nucleus. G-protein coupled receptor 30/G-protein estrogen receptor 1, which also physically interacts with estrogen receptor-α 36-kDa splice variant, acts a coregulator in this process, because its inhibition blocks the effect of estrogens on IL-6 expression. However, its activation does not mimic the effect of estrogens, on neither IL-6 nor NF-κB activity. Finally, we show that the estrogen receptor profile observed in monocytes is not modified during their differentiation to macrophages or dendritic cells in vitro and is shared in vivo by macrophages present in atherosclerotic plaques. These results position estrogen receptor-α 36-kDa splice variant and G-protein coupled receptor 30 as important players and potential therapeutic targets in monocyte/macrophage-dependent inflammatory processes.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/physiology , Monocytes/drug effects , Receptor Cross-Talk/physiology , Receptors, Estrogen/physiology , Receptors, G-Protein-Coupled/physiology , Aged , Cell Line, Tumor , Cell Nucleus/metabolism , Coronary Artery Disease/pathology , Dendritic Cells/metabolism , Estradiol/analogs & derivatives , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/genetics , Female , Foam Cells/metabolism , Foam Cells/pathology , Fulvestrant , Genes, Reporter , Humans , Inflammation , Interleukin-6/biosynthesis , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/pathology , Male , Middle Aged , Monocytes/metabolism , Myelopoiesis , Protein Interaction Mapping , Protein Isoforms/chemistry , Protein Isoforms/drug effects , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA Interference , RNA, Small Interfering/genetics , Receptors, Estrogen/chemistry , Receptors, G-Protein-Coupled/chemistry , Transcription Factor RelA/metabolism , Transcription, Genetic/drug effects
5.
J Neurosci ; 35(34): 11960-75, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311777

ABSTRACT

Within the hippocampus, the major somatostatin (SRIF) receptor subtype, the sst2A receptor, is localized at postsynaptic sites of the principal neurons where it modulates neuronal activity. Following agonist exposure, this receptor rapidly internalizes and recycles slowly through the trans-Golgi network. In epilepsy, a high and chronic release of somatostatin occurs, which provokes, in both rat and human tissue, a decrease in the density of this inhibitory receptor at the cell surface. The insulin-regulated aminopeptidase (IRAP) is involved in vesicular trafficking and shares common regional distribution with the sst2A receptor. In addition, IRAP ligands display anticonvulsive properties. We therefore sought to assess by in vitro and in vivo experiments in hippocampal rat tissue whether IRAP ligands could regulate the trafficking of the sst2A receptor and, consequently, modulate limbic seizures. Using pharmacological and cell biological approaches, we demonstrate that IRAP ligands accelerate the recycling of the sst2A receptor that has internalized in neurons in vitro or in vivo. Most importantly, because IRAP ligands increase the density of this inhibitory receptor at the plasma membrane, they also potentiate the neuropeptide SRIF inhibitory effects on seizure activity. Our results further demonstrate that IRAP is a therapeutic target for the treatment of limbic seizures and possibly for other neurological conditions in which downregulation of G-protein-coupled receptors occurs. SIGNIFICANCE STATEMENT: The somatostatin type 2A receptor (sst2A) is localized on principal hippocampal neurons and displays anticonvulsant properties. Following agonist exposure, however, this receptor rapidly internalizes and recycles slowly. The insulin-regulated aminopeptidase (IRAP) is involved in vesicular trafficking and shares common regional distribution with the sst2A receptor. We therefore assessed by in vitro and in vivo experiments whether IRAP could regulate the trafficking of this receptor. We demonstrate that IRAP ligands accelerate sst2A recycling in hippocampal neurons. Because IRAP ligands increase the density of sst2A receptors at the plasma membrane, they also potentiate the effects of this inhibitory receptor on seizure activity. Our results further demonstrate that IRAP is a therapeutic target for the treatment of limbic seizures.


Subject(s)
Cystinyl Aminopeptidase/metabolism , Hippocampus/metabolism , Receptors, Somatostatin/metabolism , Seizures/metabolism , Seizures/prevention & control , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Limbic System/metabolism , Male , Mice , Protein Transport/physiology , Rats , Rats, Wistar
6.
Regul Pept ; 188: 5-12, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24291170

ABSTRACT

BACKGROUND AND AIMS: Somatostatin and its analogs may influence hepatic fibrosis interfering through several mechanisms. The aim of this study was to investigate the effect of octreotide on cytokine activated hepatic stellate cells (HSC). METHODS: Primary HSCs were isolated from rats and were cultured on plastic for activation. Expression of somatostatin receptors (SSTR) was investigated in cultured HSCs by immunofluorescence and western blot. The effect of octreotide on cellular proliferation was studied with the MTT assay and western blot for α1-procollagen (α1-PROC) production in TNFα, TGF-ß1 or PDGF treated HSCs. Phosphotyrosine (PTP) and phosphoserine-phosphothreonine (STP) phosphatases inhibition was performed with sodium orthovanadate and okadaic acid respectively. RESULTS: Activated HSC express SSTR subtypes 1, 2A, 2B, 3 and 4 and their expression is enhanced by further HSC activation. Octreotide did not have an effect on HSC proliferation but inhibited plastic induced α1-PROC production. Interestingly, it enhanced PDGF-induced HSC proliferation but inhibited PDGF and TGFß1 dependent expression of α1-PROC, while an opposite effect was observed in TNFα-induced cell proliferation and collagen production. PTP inhibition reversed the inhibitory effect of octreotide on α1-PROC, but potentiated its effect on PDGF and TGFß1 dependent α1-PROC production. Finally, STP inhibition profoundly inhibited α1-PROC expression in all cases suggesting that both STP and PTP phosphatases are important regulators of pro-fibrotic mechanisms. CONCLUSIONS: The net effect of octreotide on HSCs and therefore liver fibrosis is subject to the cytokine microenvironment of these cells. This effect is modulated by PTPs and STPs inhibition. Especially in the case of STPs their profibrotic effects could be an interesting new therapeutic target in liver fibrosis.


Subject(s)
Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Octreotide/pharmacology , Platelet-Derived Growth Factor/physiology , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Proliferation , Cells, Cultured , Collagen/biosynthesis , Hepatic Stellate Cells/metabolism , Liver/pathology , Male , Phosphoproteins/metabolism , Protein Processing, Post-Translational , Protein Tyrosine Phosphatases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/metabolism
7.
Neuroreport ; 21(4): 254-8, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20134357

ABSTRACT

Somatostatin-14 (SRIF-14) exerts anticonvulsive effects in several rat seizure models, generally attributed to sst(2) receptor activation. Whereas sst(1) immunoreactivity has been localized to both polymorphic interneurons and principal cells in the rat hippocampus, its potential role as an inhibitory autoreceptor or as a receptor involved in mediating anticonvulsive actions remains unknown. We showed that intrahippocampal administration of the sst(1) antagonist SRA880 (1 microM) induced a robust increase in hippocampal SST-14 levels without affecting gamma-aminobutyric acid levels in conscious rats, indicating that the sst(1) receptor acts as an inhibitory autoreceptor. SRA880 did not affect seizure severity and did not reverse the anticonvulsive action of SRIF-14 (1 microM) against pilocarpine-induced seizures, suggesting that hippocampal sst(1) receptors are not involved in the anticonvulsive effects of SRIF-14.


Subject(s)
Autoreceptors/metabolism , Hippocampus/metabolism , Piperazines/pharmacology , Quinolines/pharmacology , Receptors, Somatostatin/metabolism , Seizures/metabolism , Somatostatin/metabolism , Animals , Chromatography, Liquid , Hippocampus/chemistry , Hippocampus/drug effects , Male , Microdialysis/methods , Microinjections/methods , Muscarinic Agonists , Pilocarpine , Piperazines/administration & dosage , Quinolines/administration & dosage , Rats , Rats, Wistar , Receptors, Somatostatin/agonists , Seizures/chemically induced , Somatostatin/drug effects , gamma-Aminobutyric Acid/metabolism
8.
Neuropharmacology ; 58(1): 297-303, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19576912

ABSTRACT

In a recent study, we employed an in vivo model of retinal excitotoxicity to investigate the neuroprotective effect of somatostatinergic agents. Intravitreal administration of somatostatin and sst(2) selective agonists protected the retina from (RS)-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid hydrobromide (AMPA) induced excitotoxicity. The sst(1) and sst(4) selective ligands had no effect (Kiagiadaki and Thermos, 2008). The presence of sst(5) receptors in rat retina was only recently reported (Ke and Zhong, 2007). Synthetic agonists that activate sst(2) receptors also bind with high affinity to the sst(5) subtype. In the present study the putative neuroprotective effects of sst(5) receptor activation were investigated. Adult female and male Sprague-Dawley (250-350g) rats were employed. Groups of animals received intravitreally PBS (50mM) or AMPA (42 nmol/eye) alone or in combination with L-817,818 (sst(5), 10(-5), 10(-4)M). To exclude neuroprotective effects via the activation of sst(2) receptors, L-817,818 (10(-4)M) was coinjected with the sst(2) antagonist CYN-154806 (10(-4)M). Immunohistochemistry (IHC) studies using the anti-retinal marker choline acetyltransferase (ChAT) and TUNEL staining were employed to examine retinal cell loss and protection. IHC and Western blot analysis were also employed to assess whether the sst(5) receptors are viable in the AMPA treated tissue as compared to control retina. sst(5) receptors were not affected by AMPA. L-817,818 protected the retina from the AMPA insult in the dose of 10(-4)M, while CYN-154806 (10(-4)M) had no effect on the sst(5) neuroprotection. TUNEL staining confirmed the AMPA-induced retinal toxicity and the L-817,818 neuroprotection. These results demonstrate for the first time that sst(5) receptors are functional in the retina, and that sst(5) analogs administered intravitreally protect the retina from excitotoxicity. Further studies are essential to ascertain the therapeutic relevance of these results.


Subject(s)
Excitatory Amino Acid Agonists/adverse effects , Neurotoxicity Syndromes/etiology , Receptors, Somatostatin/metabolism , Retina/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/adverse effects , Amides/pharmacology , Amides/therapeutic use , Animals , Cell Death/drug effects , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , In Situ Nick-End Labeling/methods , Male , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Retina/drug effects
9.
Invest Ophthalmol Vis Sci ; 49(7): 3080-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18390640

ABSTRACT

PURPOSE: The aim of the present study was to use an in vivo model of retinal excitotoxicity to investigate the neuroprotective effect of somatostatin (SRIF)-ergic agents. METHODS: Adult Sprague-Dawley rats (weight range, 250-300 g) intravitreally received (RS)-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid hydrobromide (AMPA; 21, 42, 84 nmol/eye) or PBS (50 mM). Time-dependent responses were examined in animals that received AMPA (42 nmol/eye). Animals received AMPA (42 nmol) alone or in combination with SRIF (10(-5), 10(-4) M) or the sst-selective ligands lanreotide (sst(2),10(-5),10(-4) M), L-779976 (sst(2,)10(-6),10(-5), 10(-4) M), L-797591 (sst(1),10(-4) M), and L-803087 (sst(4),10(-4) M). Immunohistochemistry and TUNEL studies were used to examine retinal cell loss and protection. Immunochemistry, Western blot analysis, and radioimmunoassay assessed the viability of sst(2A) receptors and SRIF levels, respectively, in control and AMPA-treated tissue. RESULTS: AMPA (42 nmol) treatment resulted in total and major loss of ChAT and bNOS immunoreactivity, respectively, 24 hours after its administration. This loss was sustained up to 30 days for ChAT- and 8 days for bNOS-expressing amacrine cells. SRIF and the sst(2) receptors were not affected by AMPA. SRIF and the sst(2) analogs protected the retina from the AMPA insult in a dose-dependent manner, whereas activation of the sst(1) and sst(4) subtypes had no effect. TUNEL staining confirmed AMPA-induced retinal ischemia and L-779976 neuroprotection. CONCLUSIONS: These results demonstrate for the first time that SRIF and the sst(2) analogs, administered intravitreally, protect the retina from excitotoxicity. Further studies are essential to ascertain the therapeutic relevance of these results.


Subject(s)
Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Retina/drug effects , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Amacrine Cells/enzymology , Amides/pharmacology , Animals , Choline O-Acetyltransferase/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Immunohistochemistry , Indoles/pharmacology , Ischemia/chemically induced , Male , Neuroprotective Agents/administration & dosage , Nitric Oxide Synthase Type I/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/agonists , Retina/enzymology , Retinal Vessels , Somatostatin/administration & dosage , Time Factors , Vitreous Body , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
10.
Invest Ophthalmol Vis Sci ; 49(1): 342-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18172111

ABSTRACT

PURPOSE: To investigate whether nitric oxide (NO) and/or cGMP protects the retina from chemical ischemia and underlie somatostatin's neuroprotective effects. METHODS: Eyecups of female Sprague-Dawley rats were incubated with PBS or the chemical ischemia mixture [iodoacetic acid (5 mM)/sodium cyanate (25 mM)] in the absence or presence of (1) arginine (0.05-2.0 mM), the substrate of nitric oxide synthase (NOS); (2) the NO donors sodium nitroprusside (SNP; 0.25-4.0 mM), 3-morpholinosydnonimine (SIN-1; 0.1, 0.3, 1.0 mM), SIN-1 (0.1 mM)/L-cysteine (5 mM, peroxynitrite scavenger), and NONOate (1, 5, 10 microM, slow NO releaser); (3) 8-Br-cGMP (0.1, 0.5, 1.0 mM); (4) BIM23014 (sst(2) receptor agonist; 1 microM), alone or in the presence of (5) the NOS inhibitor N(gamma)-monomethyl-L-arginine (NMMA; 0.5 mM); or (6) the guanylyl cyclase inhibitors 1H-[1,2,4]oxadiazolol [4,3-a]quinoxalin-1-one (ODQ;100 microM) and NS2028 (50 microM) for 60 minutes, at 5%CO(2)/air in 37 degrees C. The effect of SIN-1 (0.1, 0.3, 1.0, or 3.0 mM) on the retina was also examined. Subsequently, the eyecups were fixed and sectioned for choline acetyltransferase (ChAT) immunoreactivity and TUNEL staining. RESULTS: Arginine and SNP had no effect on the chemical ischemia-induced toxicity. SIN-1, NONOate, and 8-Br-cGMP produced a concentration-dependent protective effect, as shown by ChAT immunoreactivity. TUNEL staining also confirmed the neuroprotective effect of these agents. L-cysteine partially reduced the SIN-1-induced protective effect. SIN-1 alone was toxic only at the highest concentration used (3 mM). NMMA, ODQ, and NS2028 reversed the protective effect of BIM23014. CONCLUSIONS: The results suggest that a NO/peroxynitrite/cGMP mechanism may be important in the protection of the retina from ischemic insult. Furthermore, the NO/sGC/cGMP pathway is involved in the neuroprotective effects of sst(2) ligands against retinal ischemia.


Subject(s)
Cyclic GMP/physiology , Ischemia/prevention & control , Nitric Oxide/physiology , Peptides, Cyclic/pharmacology , Retinal Diseases/prevention & control , Retinal Vessels/drug effects , Somatostatin/analogs & derivatives , Animals , Choline O-Acetyltransferase/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Fluorescent Antibody Technique, Indirect , In Situ Nick-End Labeling , Ischemia/chemically induced , Ischemia/metabolism , Ischemia/pathology , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Rats , Rats, Sprague-Dawley , Retinal Diseases/chemically induced , Retinal Diseases/metabolism , Retinal Diseases/pathology , Somatostatin/pharmacology , Spermine/analogs & derivatives , Spermine/pharmacology
11.
Exp Eye Res ; 86(1): 18-24, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17961553

ABSTRACT

Somatostatin (SRIF) influences the release of two important neuromodulators of retinal circuitry, dopamine (DA) and nitric oxide (NO). The aim of the present study was to examine whether DA and NO modulate SRIF release in rat retina, and the mechanisms involved in their actions. Retinas of adult female Sprague--Dawley rats (250--300 g) were mechanically detached from the eyecup and ex vivo experiments were performed. Retinal explants were incubated in the presence of dopaminergic [DA (10 microM, 100 microM and 200 microM), apomorphine (nonselective D1/D2 agonist, 0.50 mM, 1.0 microM and 10 microM), A68930 (D1 selective agonist, 0.50 microM, 1.0 microM and 10 microM), quinpirole (D2 selective agonist, 0.50 microM, 1.0 microM and 10 microM), SCH 23390 (D1 selective antagonist, 250 nM and 500 nM) and sulpiride (D2 selective antagonist, 100 microM and 200 microM)], and nitrinergic agents [arginine (62.5 microM--5mM), SIN-1 (50 microM, 100 microM and 500 microM) and 8-Br-cGMP (50 microM, 250 microM and 500 microM)]. SRIF levels were quantified using radioimmunoassay (RIA). Dopamine had no effect on SRIF levels. Apomorphine produced a concentration dependent decrease and increase in SRIF levels, suggestive of pre- and postsynaptic effects. A68930 (10 microM) and SCH 23390 (250 nM and 500 nM) mimicked and reversed apomorphine's postsynaptic actions, respectively. Quinpirole had no effect, but blockade of D2 autoreceptors by sulpiride (200 microM) afforded an increase in SRIF levels. Arginine and SIN-1 increased, and 8-Br-cGMP attenuated SRIF levels. These results show that dopamine D1 receptors, and NO/peroxynitrite agents modulate SRIF release in the retina suggesting that the triad SRIF--DA--NO have reciprocal interactions via which they regulate retinal circuitry and vision transduction.


Subject(s)
Nitric Oxide/physiology , Receptors, Dopamine D1/metabolism , Retina/metabolism , Somatostatin/metabolism , Animals , Apomorphine/pharmacology , Arginine/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dopamine/pharmacology , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , Female , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/physiology , Retina/drug effects , Tissue Culture Techniques
12.
Regul Pept ; 133(1-3): 41-6, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16280179

ABSTRACT

The present study investigated the effect of somatostatin in the regulation of cGMP levels in rat retina and the mechanisms involved in this process. Isolated rat retinas were treated alone or in the presence of somatostatin (0.01-10 microM), BIM23014 (sst2 agonist, 0.01-10 microM), L-796,778 (sst3 agonist, 10 microM), somatostatin (0.1 microM) in combination with CYN154806 (sst2 antagonist, 1 microM), N(G)-methyl-L-arginine acetate salt (NMMA, inhibitor of the nitric oxide synthase (NOS), 250 microM), orthovanadate (inhibitor of tyrosine phosphatase, SHP-1, 1 microM), and arginine alone (250 microM). cGMP levels were quantified by ELISA. Immunohistochemistry studies were performed for the detection of cGMP and nNOS, while Western blot analysis was employed for the detection of SHP-1. Somatostatin increased cGMP levels in a concentration-dependent manner. This increase was inhibited by CYN154806. BIM23014 increased cGMP levels only at the concentration of 10 microM, while L-796,778 had no effect. NMMA blocked completely the somatostatin stimulated increase of cGMP levels and nNOS was detected in rat retina. cGMP immunoreactivity was observed primarily in bipolar cells only of nitroprusside-treated retinas. SHP-1 inhibition by orthovanadate reduced the somatostatin effect in a statistically significant manner. These results suggest that a SRIF/SHP-1/NO/cGMP mechanism underlies the actions of somatostatin in the retina and in its influence of retinal circuitry.


Subject(s)
Cyclic GMP/metabolism , Rats/metabolism , Receptors, Somatostatin/physiology , Retina/metabolism , Animals , Dose-Response Relationship, Drug , Female , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/metabolism , Somatostatin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...