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











Publication year range
1.
J Pept Sci ; 20(9): 736-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24931620

ABSTRACT

The solid-phase synthesis, structural characterization, and biological evaluation of a small library of cancer-targeting peptides have been determined in HepG2 hepatoblastoma cells. These peptides are based on the highly specific Pep42 motif, which has been shown to target the glucose-regulated protein 78 receptors overexpressed and exclusively localized on the cell surface of tumors. In this study, Pep42 was designed to contain varying lengths (3-12) of poly(arginine) sequences to assess their influence on peptide structure and biology. Peptides were effectively synthesized by 9-fluorenylmethoxycarbonyl-based solid-phase peptide synthesis, in which the use of a poly(ethylene glycol) resin provided good yields (14-46%) and crude purities >95% as analyzed by liquid chromatography-mass spectrometry. Peptide structure and biophysical properties were investigated using circular dichroism spectroscopy. Interestingly, peptides displayed secondary structures that were contingent on solvent and length of the poly(arginine) sequences. Peptides exhibited helical and turn conformations, while retaining significant thermal stability. Structure-activity relationship studies conducted by flow cytometry and confocal microscopy revealed that the poly(arginine) derived Pep42 sequences maintained glucose-regulated protein 78 binding on HepG2 cells while exhibiting cell translocation activity that was contingent on the length of the poly(arginine) strand. In single dose (0.15 mM) and dose-response (0-1.5 mM) cell viability assays, peptides were found to be nontoxic in human HepG2 liver cancer cells, illustrating their potential as safe cancer-targeting delivery agents.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cell-Penetrating Peptides , Drug Delivery Systems/methods , Liver Neoplasms/drug therapy , Peptides , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasm Proteins/metabolism , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Structure-Activity Relationship
2.
Bioorg Med Chem Lett ; 23(19): 5270-4, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23988358

ABSTRACT

Linear, branch and hyperbranch siRNAs were effectively prepared for down-regulating GRP78 expression and inducing cell death in HepG2 liver cancer cells. Branch and hyperbranch GRP78 siRNAs were synthesized by automated solid-phase synthesis in good yields (44-78%) and isolated in excellent purities (>99%) following HPLC purification. Moreover, siRNAs adopted stable intramolecular hybrids as discerned by native PAGE and thermal denaturation studies. These sequences also exhibited the pre-requisite A-type helical trajectory for triggering RNAi activity as determined by CD spectroscopy. Biological studies confirmed potent suppression of GRP78 expression (50-60%) while compromising cancer cell viability by ~20%. Thus, branch and hyperbranch siRNAs may serve as potent siRNA candidates in cancer gene therapy applications.


Subject(s)
RNA Interference , RNA, Small Interfering/chemical synthesis , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Hep G2 Cells , Humans , Microscopy, Confocal , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Solid-Phase Synthesis Techniques
3.
Mol Cell Endocrinol ; 286(1-2): 169-79, 2008 May 14.
Article in English | MEDLINE | ID: mdl-18375050

ABSTRACT

Somatostatin (SST) regulates multiple biological processes via five genetically distinct, G-protein coupled receptors. Clinical interest in therapy for neuroendocrine and metabolic disorders has resulted in the development of new tools for exploring the function of somatostatin receptors (SSTRs). The development of highly SSTR-selective agonists and antagonists, animal models with the deletion of individual SSTRs, as well as SSTR-specific antibodies have all been utilized in delineating SSTR functions. In the pancreas, SST is a potent regulator of insulin and glucagon secretion. Indeed, the inappropriate regulation of pancreatic A- and B-cell function in metabolic diseases provides an impetus to evaluate the SSTRs as therapeutic targets. By combining the results obtained from molecular biology, pharmacology and immunochemical studies the current review provides a summary of important recent developments which have extended our knowledge of SST actions in the endocrine pancreas.


Subject(s)
Islets of Langerhans/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Humans , Mice , Mice, Knockout , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Rats , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors
4.
Endocrinology ; 146(11): 4975-84, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16081634

ABSTRACT

The secretion of GH by somatotropes is inhibited by somatostatin (SRIF) through five specific membrane receptors (SSTRs). SRIF increases both transient outward (IA) and delayed rectifying (IK) K+ currents. We aim to clarify the subtype(s) of SSTRs involved in K+ current enhancement in GH3 somatotrope cells using specific SSTR subtype agonists. Expression of all five SSTRs was confirmed in GH3 cells by RT-PCR. Nystatin-perforated patch clamp was used to record voltage-gated K+ currents. We first established the presence of IA and IK type K+ currents in GH3 cells using different holding potentials (-40 or -70 mV) and specific blockers (4-aminopirimidine and tetraethylammonium chloride). SRIF (200 nM) increased the amplitude of both IA and IK in a fully reversible manner. Various concentrations of each specific SRTR agonist were tested on K+ currents to find the maximal effective concentration. Activation of SSTR2 and SSTR4 by their respective agonists, L-779,976 and L-803,087 (10 nM), increased K+ current amplitude without preference to IA or IK, and abolished any further increase by SRIF. Activation of SSTR1 and SSTR5 by their respective agonists, L-797,591 or L-817,818 (10 nM), increased K+ current amplitude, but SRIF evoked a further increase. The SSTR3 agonist L-797,778 (10 nM) did not affect the K+ currents or the response to SRIF. These results indicate that SSTR1, -2, -4, and -5 may all be involved in the enhancement of K+ currents by SRIF but that only the activation of SSTR2 or -4 results in the full activation of K+ current caused by SRIF.


Subject(s)
Growth Hormone/metabolism , Hormones/pharmacology , Pituitary Gland, Anterior/metabolism , Potassium Channels, Voltage-Gated/drug effects , Potassium Channels, Voltage-Gated/physiology , Somatostatin/pharmacology , Animals , Cell Line , Pituitary Gland, Anterior/cytology , Protein Isoforms/agonists , Protein Isoforms/metabolism , Rats , Receptors, Somatostatin/agonists , Receptors, Somatostatin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
5.
Curr Vasc Pharmacol ; 3(2): 125-31, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15853632

ABSTRACT

Somatostatin (somatotropin release inhibitory factor; SRIF) peptides are widely distributed throughout the mammalian body and act through a family of genetically distinct, guanine nucleotide regulatory protein coupled (G-protein-coupled), cell surface receptors (sst(1-5)). Compelling evidence shows that SRIF and SRIF peptidyl analogs modulate vascular function, with actions upon smooth muscle and endothelium. SRIF receptors are known to exist in the carotid endothelium, a principal target for the pro-inflammatory cascade that accompanies coronary artery disease. SRIF-14 and SRIF analogs are anti-inflammatory but the molecular mechanism involved remains unclear. Since crucial steps in the endothelial inflammation response include endothelial activation by cytokines, adhesion molecule expression and cell-monocyte interactions, peptide agents that inhibit these steps might provide a novel strategy for reducing vascular inflammation. SRIF, acting through its cognate receptors, modulates a variety of intracellular effectors that are linked to inflammation including phosphotyrosine phosphatases, the extracellular regulated protein kinase 1 and 2 (ERK1/2) cascade, adenylyl cyclase and endothelial nitric oxide synthase. Directly or indirectly, SRIF also functions to inhibit endothelial cell proliferation and induce apoptosis. A detailed understanding of SRIF actions could provide a rational basis for using SRIF ligands in controlling vascular inflammation and inhibiting cytokine signaling, critical events in atherogenesis.


Subject(s)
Hormone Antagonists/pharmacology , Somatostatin/physiology , Animals , Humans , Receptors, Somatostatin/drug effects
6.
Biochem Biophys Res Commun ; 319(4): 1222-7, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15194497

ABSTRACT

Somatostatin (somatotropin release inhibitory factor; SRIF) is an endogenous peptide produced at sites of inflammation, making the SRIF a candidate in regulating vascular inflammation. We have used primary human coronary artery endothelial cells (hCAEC) as a model to study SRIF's vascular actions. RT-PCR analysis of hCAEC total mRNA demonstrated the presence of the sst(4) receptor subtype, providing a target for SRIF intracellular signaling. Western blotting with phospho-specific ERK1/2 antibodies showed that SRIF-14 acutely inhibited basal phosphorylation of the extracellular regulated kinases (ERK1/2) by 80%. In addition, SRIF-14 treated hCAEC cell lysates showed a 2.6-fold increase in phosphatase activity, which was inhibited by sodium vanadate. Furthermore, SRIF-14 appeared to be anti-inflammatory in hCAEC as IL-1beta-induced adhesion molecule expression was reduced by 50%. Together, these results show that the coronary artery endothelium is a direct target of SRIF action.


Subject(s)
Carotid Arteries/anatomy & histology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Signal Transduction/physiology , Somatostatin/metabolism , Cells, Cultured , Endothelial Cells/cytology , Humans , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1/metabolism , Membrane Proteins , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Isoforms/metabolism , Receptors, Somatostatin/metabolism , Vanadates/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
7.
Protein Pept Lett ; 11(2): 141-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15078202

ABSTRACT

Nonpeptidyl agonists for the somatostatin (SRIF) receptor family have been developed. We have studied the desensitization effects for two of these agonists upon SRIF receptor function in AtT-20 cells, a neuroendocrine tumor cell, which endogenously expresses two distinct SRIF receptor, subtypes. We observe that SRIF and the nonpeptidyl, subtype selective agonists, differentially regulate SRIF receptor subtypes in the AtT-20 cell.


Subject(s)
Receptors, Somatostatin/metabolism , Signal Transduction , Somatostatin/metabolism , Animals , Cell Line, Tumor , Colforsin/pharmacology , Cyclic AMP/metabolism , Mice , Receptors, Somatostatin/agonists , Signal Transduction/drug effects , Somatostatin/analogs & derivatives
8.
Biochem Biophys Res Commun ; 314(2): 501-4, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14733934

ABSTRACT

The effects of a clinically useful cardiovascular agent, dipyridamole, were examined in a rodent tissue culture model of neuroprotection. Dipyridamole effectively protected rat embryonic day 18 (E18) cortical neurons from either 48 h trophic deprivation or 48 h exposure to the glutathione synthesis inhibitor, L-buthionine (R,S) sulfoximine. The neuron sparing actions of dipyridamole were time- and concentration-dependent and mimicked the actions of exogenously applied glutathione. These results demonstrate that dipyridamole protects primary neuronal cultures against either trophic or chemically mediated insults, and suggest that dipyridamole has a potent antioxidant ability that compensates for glutathione depletion in neuronal cultures.


Subject(s)
Dipyridamole/pharmacology , Neurons/cytology , Animals , Antioxidants/pharmacology , Buthionine Sulfoximine/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Glutathione/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidation-Reduction , Phosphodiesterase Inhibitors/pharmacology , Rats , Time Factors
9.
J Pharmacol Exp Ther ; 306(2): 447-54, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12754318

ABSTRACT

A sensitive quantitative-competitive reverse transcriptase-polymerase chain reaction method was developed to measure micro-opioid receptor (MOR) mRNA expression in SHSY-5Y neuroblastoma cells. Differentiation of SHSY-5Y cells with either retinoic acid (RA) or 12-o-tetradecanoyl-phorbol-13-acetate (TPA) significantly increased MOR mRNA levels. Morphine treatment (10 microM) for 24 h decreased MOR mRNA levels in control, as well as RA- and TPA-differentiated cells. In contrast, chronic exposure to the opioid peptides endomorphin-1 or endomorphin-2 significantly increased MOR mRNA levels in undifferentiated and RA-differentiated cells. An opioid antagonist, naloxone, reversed the morphine and endomorphin-1 and -2 effects on MOR mRNA levels in undifferentiated SHSY-5Y cells, but naloxone had differential reversing effects on the agonists' regulation of MOR mRNA in RA- or TPA-differentiated cells. To investigate whether the changes in MOR mRNA expression paralleled changes in MOR receptor function, intracellular cAMP accumulation in SHSY-5Y cells was measured. After chronic treatment with morphine, forskolin-induced cAMP levels in SHSY-5Y cells were significantly higher than those of untreated control cells. In contrast, forskolin-induced cAMP accumulation levels were lower in cells treated with endomorphin-1 or -2 than in untreated control cells. Together, our studies indicate that the opioid alkaloid morphine and the opioid peptides endomorphin-1 and -2 differentially regulate MOR mRNA expression and MOR function in SHSY-5Y cells.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Oligopeptides/pharmacology , Receptors, Opioid, mu/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Drug Interactions , Humans , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neuroblastoma/pathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Opioid, mu/genetics , Tumor Cells, Cultured
10.
Mol Endocrinol ; 17(1): 93-106, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12511609

ABSTRACT

Somatostatin (SRIF) regulates pancreatic insulin and glucagon secretion. In the present study we describe the generation of SRIF receptor subtype 5 knockout (sst(5) KO) mice to examine the role of SRIF receptor subtypes (sst) in regulating insulin secretion and glucose homeostasis. Mice deficient in sst(5) were viable, fertile, appeared healthy, and displayed no obvious phenotypic abnormalities. Pancreatic islets isolated from sst(5) KO mice displayed increased total insulin content as compared with islets obtained from wild-type (WT) mice. Somatostatin-28 (SRIF-28) and the sst(5)/sst(1)-selective agonist compound 5/1 potently inhibited glucose-stimulated insulin secretion from WT islets. SRIF-28 inhibited insulin secretion from sst(5) KO islets with 16-fold less potency while the maximal effect of compound 5/1 was markedly diminished when compared with its effects in WT islets. sst(5) KO mice exhibited decreased blood glucose and plasma insulin levels and increased leptin and glucagon concentrations compared with WT mice. Furthermore, sst(5) KO mice displayed decreased susceptibility to high fat diet-induced insulin resistance. The results of these studies suggest sst(5) mediates SRIF inhibition of pancreatic insulin secretion and contributes to the regulation of glucose homeostasis and insulin sensitivity. Our findings suggest a potential beneficial role of sst(5) antagonists for alleviating metabolic abnormalities associated with obesity and insulin resistance.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Receptors, Somatostatin/physiology , Animals , CHO Cells , Cloning, Molecular/methods , Cricetinae , Female , Gene Targeting/methods , Homeostasis/physiology , Insulin Resistance/physiology , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout , Organ Culture Techniques , Receptors, Somatostatin/agonists , Receptors, Somatostatin/deficiency , Receptors, Somatostatin/genetics , Somatostatin/metabolism , Somatostatin-28 , Transfection
11.
Neuroendocrinology ; 75(6): 339-46, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12065886

ABSTRACT

Somatostatin (SRIH) regulates pituitary adrenocorticotropin (ACTH) secretion by interacting with a family of homologous G protein-coupled membrane receptors. The SRIH receptor subtypes (sst(1)-sst(5)) that control ACTH release remain unknown. Using novel, subtype-selective SRIH analogs, we have identified the SRIH receptor subtypes involved in regulating ACTH release from AtT-20 cells, a model for cell line pituitary corticotropes. Radioligand-binding studies with (125)I-SRIH-14 and (125)I-SRIH-28 showed that SRIH-14 and SRIH-28 recognized specific, high-affinity and saturable membrane-binding sites. Nonpeptidyl agonists with selectivity for the sst(2) (L-779,976; compound 2) or sst(1)/sst(5)) (L-817,818; compound 5) receptor subtypes potently displaced (125)I-SRIH-28 from AtT-20 cell membranes, while agonists selective for the sst(1) (L-779,591; compound 1), sst(3) (L-796,778; compound 3) or sst(4) (L-803,087; compound 4) subtypes were inactive. Tyr(11)-SRIH-14, compound 2 (sst(2)) or compound 5 (sst(5)) inhibited forskolin and corticotropin-releasing hormone (CRH)-induced increases in intracellular cAMP. Furthermore, the sst(2) and sst(5) agonists potently inhibited CRH-induced ACTH release from AtT-20 cells. These results provide the first evidence that sst(2) and sst(5) receptor subtypes, but not sst(1), sst(3) or sst(4), inhibit cAMP accumulation and regulate ACTH secretion in the AtT-20 cell model of the rodent corticotrope.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Fungal Proteins/physiology , GTPase-Activating Proteins , Pituitary Gland/metabolism , Receptors, Somatostatin/physiology , Saccharomyces cerevisiae Proteins , Binding, Competitive , Cell Line , Cyclic AMP/metabolism , Fungal Proteins/metabolism , Iodine Radioisotopes , Pituitary Gland/drug effects , Radioligand Assay , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Somatostatin-28
SELECTION OF CITATIONS
SEARCH DETAIL