Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 159
Filter
1.
Regul Pept ; 120(1-3): 133-40, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15177931

ABSTRACT

Processing of prohormones to generate active products typically occurs at basic residues via cleavage by proprotein convertases. A less common type of cleavage is mediated at hydrophobic (L, V, F, N) or small amino acid (A, T, S) residues. Efforts to identify the proteinases responsible for processing precursors at their hydrophobic amino acids has led to the recent cloning of a new type-1 membrane-bound subtilase called SKI-1. The NH2-terminal region of prosomatostatin, previously shown to contain a sorting signal for the regulated secretory pathways, is processed to generate PSST[1-10]. The exact cleavage mechanism is unknown, but has been assumed to involve monobasic processing at Lys13 followed by carboxypeptidase trimming. We found that K13A mutation did not block PSST[1-10] production. Since the prosomatostatin sequence R8-Q9-F10-L11 \ qualifies as a potential SKI-1 substrate, using a vaccinia virus expression system along with HPLC and radioimmunoassays, we observed that overexpression of recombinant SKI-1 in COS-1 and HEK-293 cells significantly increased the production of PSST[1-10]. Additionally, in CHO cells lacking SKI-1, there was a significant reduction in PSST[1-10] production which could be increased upon SKI-1 stimulation. Mutagenesis studies showed that efficient processing of PSST to PSST[1-10] required the RXRXXL motif. However, this NH2-terminal cleavage was not a prerequisite for the formation of SST-14 and SST-28.


Subject(s)
Proprotein Convertases/pharmacology , Protein Precursors/metabolism , Protein Processing, Post-Translational , Serine Endopeptidases/pharmacology , Somatostatin/metabolism , Amino Acid Motifs , Animals , CHO Cells , COS Cells , Chromatography, High Pressure Liquid , Cricetinae , Humans , Kidney/metabolism , Mutagenesis , Mutation/genetics , Protein Precursors/genetics , Protein Structure, Tertiary , Radioimmunoassay , Somatostatin/genetics , Vaccinia virus/genetics
2.
Neuroscience ; 125(2): 441-8, 2004.
Article in English | MEDLINE | ID: mdl-15062986

ABSTRACT

The actions of somatostatin (SST) in the nervous system are mediated by specific high affinity SST receptors (SSTR1-5). However, the role of this hormone and the distribution of its receptor subtypes have not yet been defined in neural structures of the human fetus. We have analyzed four neural tissues (CNS, hypothalamus, pituitary and spinal cord) from early to midgestation for the expression of five human SSTR mRNAs, using a reverse transcription-polymerase chain reaction and Southern blot approach. These fetal neural tissues all express mRNA for multiple SSTR subtypes from as early as 16 weeks of fetal life but the developmental patterns of expression vary considerably. Transcripts for SSTR1 and SSTR2A are the most widely distributed, being expressed in all four neural tissues. SSTR2A is often the earliest transcript to be detected (7.5 weeks in CNS). SSTR3 mRNA is confined to the pituitary, hypothalamus, and spinal cord. SSTR4 is expressed in fetal brain, hypothalamus and spinal cord but not pituitary. SSTR5 mRNA is detectable in the pituitary and spinal cord by 14-16 weeks of fetal life. This mapping of SSTR mRNA expression patterns in human fetal neural tissues is an important first step toward our goal of determining the role of SST in the nervous system during early stages in human development.


Subject(s)
Gene Expression Regulation, Developmental , Hypothalamus/metabolism , Pituitary Gland/metabolism , Receptors, Somatostatin/metabolism , Spinal Cord/metabolism , Aborted Fetus , Blotting, Southern/methods , Central Nervous System/anatomy & histology , Central Nervous System/metabolism , Gestational Age , Humans , Hypothalamus/embryology , Pituitary Gland/embryology , RNA, Messenger/biosynthesis , Receptors, Somatostatin/classification , Receptors, Somatostatin/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Spinal Cord/embryology
3.
Gastroenterology ; 121(4): 915-30, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11606505

ABSTRACT

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) are considered therapeutic targets to decrease portal hypertension. To elucidate some of the hemodynamic effects of somatostatin (SST) on portal pressure, the presence and function of SST receptors (SSTRs) on HSCs were investigated. METHODS: SSTR messenger RNA expression, and SSTR presence was investigated using reverse-transcription polymerase chain reaction, real-time quantitative polymerase chain reaction, Western blotting, and immunohistochemistry. The function of SSTRs was studied by examining the effects of SST and specific SSTR agonists on endothelin-1(ET-1)-induced HSC contraction. RESULTS: Specific amplicons for SSTR subtypes 1, 2, and 3 were demonstrated in rat liver and in activated HSCs. The presence of SSTR subtypes 1, 2, and 3 was confirmed by Western blotting. With immunohistochemistry, a strong staining of HSCs was obtained for SSTR subtypes 1, 2, and 3 in CCl4-treated rats, but not in normal rat liver. Incubation of HSCs on collagen gels with buffer, 10(-8) mol/L SST, and 2 x 10(-8) mol/L ET-1 resulted in collagen surface area decreases of 5.5% +/- 3.3%, 6.8% +/- 4.4%, and 49.8% +/- 8.3%, respectively. Relative contraction of gels preincubated with 10(-8) mol/L SST followed by 2 x 10(-8) mol/L ET-1 or vice versa as compared with maximal contraction (100%) with 2 x 10(-8) mol/L ET-1 were 72.6% +/- 17.9% and 76.2% +/- 12.6%, respectively (P < 0.05). SSTR agonist 1, but not SSTR agonist 2 or 3, was able to counteract the contractile effect of ET-1. CONCLUSIONA: Activated rat HSCs bear SSTR subtypes 1, 2, and 3. SST causes significant partial inhibition of ET-1-induced contraction of activated HSCs, mainly by stimulation of SSTR subtype 1.


Subject(s)
Endothelin-1/pharmacology , Gene Expression Regulation/physiology , Liver/cytology , Receptors, Somatostatin/genetics , Somatostatin/pharmacology , Animals , Cell Culture Techniques/methods , Cell Size/drug effects , Cells, Cultured , DNA Primers , Endothelin-1/antagonists & inhibitors , Gene Expression Regulation/drug effects , Immunohistochemistry , Liver/drug effects , Male , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Rats , Rats, Wistar , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
4.
J Clin Invest ; 107(12): 1571-80, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11413165

ABSTRACT

Pulsatile growth hormone (GH) secretion differs between males and females and regulates the sex-specific expression of cytochrome P450s in liver. Sex steroids influence the secretory dynamics of GH, but the neuroendocrine mechanisms have not been conclusively established. Because periventricular hypothalamic somatostatin (SST) expression is greater in males than in females, we generated knockout (Smst(-/-)) mice to investigate whether SST peptides are necessary for sexually differentiated GH secretion and action. Despite marked increases in nadir and median plasma GH levels in both sexes of Smst(-/-) compared with Smst(+/+) mice, the mutant mice had growth curves identical to their sibling controls and retained a normal sexual dimorphism in weight and length. In contrast, the liver of male Smst(-/-) mice was feminized, resulting in an identical profile of GH-regulated hepatic mRNAs between male and female mutants. Male Smst(-/-) mice show higher expression of two SST receptors in the hypothalamus and pituitary than do females. These data indicate that SST is required to masculinize the ultradian GH rhythm by suppressing interpulse GH levels. In the absence of SST, male and female mice exhibit similarly altered plasma GH profiles that eliminate sexually dimorphic liver function but do not affect dimorphic growth.


Subject(s)
Growth Hormone/physiology , Liver/metabolism , Sex Characteristics , Somatostatin/physiology , Animals , Body Weight , Female , Genotype , Growth Hormone/metabolism , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Pituitary Gland/metabolism , RNA, Messenger/biosynthesis , Receptors, Somatostatin/biosynthesis , Receptors, Somatostatin/genetics , Recombination, Genetic , Somatostatin/genetics , Transcription, Genetic
5.
Clin Endocrinol (Oxf) ; 54(5): 641-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11380495

ABSTRACT

BACKGROUND: We have analysed the distribution of the five somatostatin receptors (sst1-5) by immunohistochemistry in a large retrospective series of 51 medullary carcinoma of the thyroid (MCT) specimens and correlated the pattern of sst expression with expression of somatostatin (SRIF) peptide, tumour pathology and clinical outcome. MEASUREMENTS: Immunohistochemistry was performed with rabbit polyclonal antipeptide antibodies directed against the extracellular domains or cytoplasmic tail of human (h) sst1-5. SRIF immunoreactivity was investigated in parallel paraffin sections. RESULTS: Eighty-five percent of the tumours were positive for one or more sst, localized to both tumour cells as well as surrounding peritumoural structures, especially blood vessels. Forty-nine percent of the tumours were positive for sst1, 43% for sst2, 47% for sst3, 4% for sst4, and 57% for sst5. Fifty-one percent of tumours expressed one or two sst subtypes; 33% were positive for three or more sst isoforms. All five sst receptors were detected in only two cases. Tumours expressing octreotide sensitive subtypes (sst2,3,5) accounted for 75% of the series. 50% of the tumours co-expressed SRIF suggesting tumour cell regulation by endogenous SRIF via paracrine/autocrine circuits. There was no correlation between sst1-5 expression and age, sex, tumour size or stage, histological type or clinical outcome. Simultaneous analysis of primary tumour and lymph node metastases revealed a similar pattern of sst immunoreactivity indicating that sst expression is not modified in the course of disease progression. CONCLUSIONS: With the exception of sst4, medullary carcinoma of the thyroid display a rich but heterogeneous expression of sst subtypes. Immunohistochemical typing of sst receptor expression using specific antireceptor antibodies represents an ideal approach for characterizing sst subtype expression in medullary carcinoma of the thyroid for optimizing receptor targeted diagnosis and therapy with somatostatin analogs.


Subject(s)
Carcinoma, Medullary/chemistry , Receptors, Somatostatin/analysis , Thyroid Neoplasms/chemistry , Adult , Aged , Aged, 80 and over , Carcinoma, Medullary/pathology , Chi-Square Distribution , Female , Humans , Immunohistochemistry , Male , Middle Aged , Retrospective Studies , Somatostatin/analysis , Thyroid Gland/chemistry , Thyroid Neoplasms/pathology , Treatment Outcome
6.
J Biol Chem ; 276(28): 26308-16, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11309402

ABSTRACT

Mammalian prosomatostatin (PSST) contains the bioactive peptides SST-14 and SST-28 at the COOH-terminal end of the molecule and a putative sorting signal in the propeptide segment for targeting the precursor to the regulated secretory pathway. The NH(2)-terminal segment of PSST consists of an amphipathic alpha-helix, which has been totally conserved throughout vertebrate evolution. We have analyzed the PSST-(3--15) region for sorting function by alanine scanning and deletional mutagenesis. Mutants created were stably expressed in AtT-20 cells. Regulated secretion was studied by analyzing basal and stimulated release of SST-14 LI and by immunocytochemistry for staining of SST-14 LI in punctate granules. Deletion of the PSST-(3--15) segment blocked regulated secretion and rerouted PSST for constitutive secretion as unprocessed precursor. Alanine scanning mutagenesis identified the region Pro(5)--Gln(12) as being important in precursor targeting, with Leu(7) and Leu(11) being critical. Molecular modeling demonstrated that these two residues are located in close proximity on a hydrophobic surface of the alpha-helix. Disruption of the alpha-helix did not impair the ability of PSST to be processed at the COOH terminus to SST-14 and SST-28. Processing, however, was shifted to the early compartments of the secretory pathway rather than storage granules and was relatively inefficient.


Subject(s)
Protein Precursors/genetics , Protein Precursors/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , Fluorescent Antibody Technique , Mice , Molecular Sequence Data , Protein Folding , Protein Transport
7.
Clin Invest Med ; 23(3): 172-87, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10911548

ABSTRACT

Acromegaly is a chronic, debilitating condition caused by excessive secretion of growth hormone (GH). In the majority of cases the condition results from benign pituitary adenomas or, rarely, from ectopic production of GH-releasing hormone. Regardless of the cause, excess GH results in physical disfigurement associated with arthropathy, diabetes, hypertension, cardiac dysfunction, obstructive sleep apnea and colonic neoplasia. The death rate for acromegalic patients is 2 to 3 times higher than that of the general population, but with appropriate reduction of GH hypersecretion it tends to shift into the normal range. Treatment is thus aimed at normalizing GH secretion; eradicating or stabilizing the pituitary tumour while preserving normal pituitary function, and managing the associated complications. The treatment modalities available to achieve these objectives include transsphenoidal surgery, pharmacotherapy and radiation, or various combinations of these. This review provides an update on our current understanding of the pathophysiology of GH hypersecretion in acromegaly, the newly defined diagnostic criteria and the end point for a cure for acromegaly, and on new developments in drug treatment with the advent of slow-release forms of somatostatin analogues and the longer-acting dopamine receptor agonists, as well as in the area of radiotherapy. Its main purpose is to guide any physician involved in the diagnosis and management of patients with acromegaly.


Subject(s)
Acromegaly/diagnosis , Acromegaly/therapy , Practice Guidelines as Topic , Acromegaly/mortality , Acromegaly/surgery , Canada , Health Care Costs , Human Growth Hormone/antagonists & inhibitors , Human Growth Hormone/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Pituitary Gland/metabolism , Pituitary Gland/pathology , Pituitary Gland/physiopathology , Pituitary Gland/radiation effects , Radiotherapy , Receptors, Somatotropin/antagonists & inhibitors , Receptors, Somatotropin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Somatostatin/therapeutic use
8.
J Biol Chem ; 275(13): 9244-50, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10734062

ABSTRACT

Activation of initiator and effector caspases, mitochondrial changes involving a reduction in its membrane potential and release of cytochrome c (cyt c) into the cytosol, are characteristic features of apoptosis. These changes are associated with cell acidification in some models of apoptosis. The hierarchical relationship between these events has, however, not been deciphered. We have shown that somatostatin (SST), acting via the Src homology 2 bearing tyrosine phosphatase SHP-1, exerts cytotoxic action in MCF-7 cells, and triggers cell acidification and apoptosis. We investigated the temporal sequence of apoptotic events linking caspase activation, acidification, and mitochondrial dysfunction in this system and report here that (i) SHP-1-mediated caspase-8 activation is required for SST-induced decrease in pH(i). (ii) Effector caspases are induced only when there is concomitant acidification. (iii) Decrease in pH(i) is necessary to induce reduction in mitochondrial membrane potential, cyt c release and caspase-9 activation and (iv) depletion of ATP ablates SST-induced cyt c release and caspase-9 activation, but not its ability to induce effector caspases and apoptosis. These data reveal that SHP-1-/caspase-8-mediated acidification occurs at a site other than the mitochondrion and that SST-induced apoptosis is not dependent on disruption of mitochondrial function and caspase-9 activation.


Subject(s)
Acids/metabolism , Apoptosis/drug effects , Caspases/metabolism , Mitochondria/metabolism , Somatostatin/pharmacology , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Apoptotic Protease-Activating Factor 1 , Blotting, Western , Caspase 8 , Caspase 9 , Cytochrome c Group/metabolism , Enzyme Activation , Humans , Hydrogen-Ion Concentration , Mitochondria/enzymology , Proteins/metabolism , Tumor Cells, Cultured
9.
J Biol Chem ; 275(11): 7862-9, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10713101

ABSTRACT

The existence of receptor dimers has been proposed for several G protein-coupled receptors. However, the question of whether G protein-coupled receptor dimers are necessary for activating or modulating normal receptor function is unclear. We address this question with somatostatin receptors (SSTRs) of which there are five distinct subtypes. By using transfected mutant and wild type receptors, as well as endogenous receptors, we provide pharmacological, biochemical, and physical evidence, based on fluorescence resonance energy transfer analysis, that activation by ligand induces SSTR dimerization, both homo- and heterodimerization with other members of the SSTR family, and that dimerization alters the functional properties of the receptor such as ligand binding affinity and agonist-induced receptor internalization and up-regulation. Double label confocal fluorescence microscopy showed that when SSTR1 and SSTR5 subtypes were coexpressed in Chinese hamster ovary-K1 cells and treated with agonist they underwent internalization and were colocalized in cytoplasmic vesicles. SSTR5 formed heterodimers with SSTR1 but not with SSTR4 suggesting that heterodimerization is a specific process that is restricted to some but not all receptor subtype combinations. Direct protein interaction between different members of the SSTR subfamily defines a new level of molecular cross-talk between subtypes of the SSTR and possibly related receptor families.


Subject(s)
Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Dimerization , Energy Transfer , Fluorescence , Humans , Membrane Proteins , Peptide Fragments/metabolism , Protein Conformation , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/classification , Receptors, Somatostatin/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/classification , Recombinant Proteins/metabolism , Somatostatin/analogs & derivatives
10.
Science ; 288(5463): 154-7, 2000 Apr 07.
Article in English | MEDLINE | ID: mdl-10753124

ABSTRACT

Somatostatin and dopamine are two major neurotransmitter systems that share a number of structural and functional characteristics. Somatostatin receptors and dopamine receptors are colocalized in neuronal subgroups, and somatostatin is involved in modulating dopamine-mediated control of motor activity. However, the molecular basis for such interaction between the two systems is unclear. Here, we show that dopamine receptor D2R and somatostatin receptor SSTR5 interact physically through hetero-oligomerization to create a novel receptor with enhanced functional activity. Our results provide evidence that receptors from different G protein (heterotrimeric guanine nucleotide binding protein)-coupled receptor families interact through oligomerization. Such direct intramembrane association defines a new level of molecular crosstalk between related G protein-coupled receptor subfamilies.


Subject(s)
Receptor Cross-Talk , Receptors, Dopamine D2/metabolism , Receptors, Somatostatin/metabolism , Animals , CHO Cells , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Colforsin/pharmacology , Corpus Striatum/metabolism , Cricetinae , Cyclic AMP/metabolism , Dimerization , Dopamine D2 Receptor Antagonists , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Ligands , Male , Neurons/metabolism , Pyramidal Cells/metabolism , Quinpirole/pharmacology , Rats , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/genetics , Somatostatin/metabolism , Somatostatin/pharmacology , Spiperone/pharmacology , Sulpiride/pharmacology , Transfection
11.
Front Neuroendocrinol ; 20(3): 157-98, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10433861

ABSTRACT

Somatostatin (SST), a regulatory peptide, is produced by neuroendocrine, inflammatory, and immune cells in response to ions, nutrients, neuropeptides, neurotransmitters, thyroid and steroid hormones, growth factors, and cytokines. The peptide is released in large amounts from storage pools of secretory cells, or in small amounts from activated immune and inflammatory cells, and acts as an endogenous inhibitory regulator of the secretory and proliferative responses of target cells that are widely distributed in the brain and periphery. These actions are mediated by a family of seven transmembrane (TM) domain G-protein-coupled receptors that comprise five distinct subtypes (termed SSTR1-5) that are endoded by separate genes segregated on different chromosomes. The five receptor subtypes bind the natural SST peptides, SST-14 and SST-28, with low nanomolar affinity. Short synthetic octapeptide and hexapeptide analogs bind well to only three of the subtypes, 2, 3, and 5. Selective nonpeptide agonists with nanomolar affinity have been developed for four of the subtypes (SSTR1, 2, 3, and 4) and putative peptide antagonists for SSTR2 and SSTR5 have been identified. The ligand binding domain for SST ligands is made up of residues in TMs III-VII with a potential contribution by the second extracellular loop. SSTRs are widely expressed in many tissues, frequently as multiple subtypes that coexist in the same cell. The five receptors share common signaling pathways such as the inhibition of adenylyl cyclase, activation of phosphotyrosine phosphatase (PTP), and modulation of mitogen-activated protein kinase (MAPK) through G-protein-dependent mechanisms. Some of the subtypes are also coupled to inward rectifying K(+) channels (SSTR2, 3, 4, 5), to voltage-dependent Ca(2+) channels (SSTR1, 2), a Na(+)/H(+) exchanger (SSTR1), AMPA/kainate glutamate channels (SSTR1, 2), phospholipase C (SSTR2, 5), and phospholipase A(2) (SSTR4). SSTRs block cell secretion by inhibiting intracellular cAMP and Ca(2+) and by a receptor-linked distal effect on exocytosis. Four of the receptors (SSTR1, 2, 4, and 5) induce cell cycle arrest via PTP-dependent modulation of MAPK, associated with induction of the retinoblastoma tumor suppressor protein and p21. In contrast, SSTR3 uniquely triggers PTP-dependent apoptosis accompanied by activation of p53 and the pro-apoptotic protein Bax. SSTR1, 2, 3, and 5 display acute desensitization of adenylyl cyclase coupling. Four of the subtypes (SSTR2, 3, 4, and 5) undergo rapid agonist-dependent endocytosis. SSTR1 fails to be internalized but is instead upregulated at the membrane in response to continued agonist exposure. Among the wide spectrum of SST effects, several biological responses have been identified that display absolute or relative subtype selectivity. These include GH secretion (SSTR2 and 5), insulin secretion (SSTR5), glucagon secretion (SSTR2), and immune responses (SSTR2).


Subject(s)
Receptors, Somatostatin/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Amino Acid Sequence , Animals , Cell Division , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Molecular Sequence Data , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics , Receptors, Somatostatin/immunology , Signal Transduction , Somatostatin/agonists , Somatostatin/antagonists & inhibitors
12.
Eur J Immunol ; 29(8): 2454-63, 1999 08.
Article in English | MEDLINE | ID: mdl-10458759

ABSTRACT

Macrophages secrete the immunoregulatory peptide somatostatin (SOM) that inhibits IFN-gamma release by splenocytes and granuloma cells of schistosome-infected mice. In this report we demonstrate that granuloma cells express mRNA for the SOM receptor SSTR2 but not the other four SSTR subtypes. Blocking SSTR2 activity with anti-SSTR2 antiserum prevents SOM inhibition of T cell IFN-gamma production. This demonstrates that SOM regulates T cell function via SSTR2. Two isoforms of SSTR2 exist due to alternative RNA splicing. We developed sensitive and specific competitive PCR assays to quantify total SSTR2, SSTR2A and SSTR2B mRNA levels. The SSTR2A isoform accounts for 99% of inflammatory cell SSTR2 mRNA and does not appear to be regulated at the transcripitonal level. B cells and macrophage cell lines also express SSTR2 mRNA which raises the possibility that SOM influences T cell IFN-gamma release by regulating accessory cell function. We show that SOM acts directly on T cells to inhibit TCR-stimulated IFN-gamma release. Thus, SOM may directly regulate T cell IFN-gamma release at inflammatory sites.


Subject(s)
Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/metabolism , Receptors, Somatostatin/metabolism , T-Lymphocytes/immunology , Animals , Antibodies , B-Lymphocytes/metabolism , Base Sequence , Cell Line , Cytokines/pharmacology , DNA Primers/genetics , Female , Gene Expression , In Vitro Techniques , Inflammation/genetics , Inflammation Mediators/pharmacology , Macrophages/metabolism , Mice , Mice, Inbred CBA , Octreotide/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Somatostatin/classification , Receptors, Somatostatin/genetics , Somatostatin/pharmacology
13.
J Biol Chem ; 274(35): 24550-8, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10455118

ABSTRACT

We have previously reported that the human somatostatin receptor type 1 (hSSTR1) stably expressed in Chinese hamster ovary-K1 cells does not internalize but instead up-regulates at the membrane during continued agonist treatment (1 microM somatostatin (SST)-14 x 22 h). Here we have investigated the molecular basis of hSSTR1 up-regulation. hSSTR1 was up-regulated by SST in a time-, temperature-, and dose-dependent manner to saturable levels, in intact cells but not in membrane preparations. Although hSSTR1 was acutely desensitized to adenylyl cyclase coupling after 1 h SST-14 treatment, continued agonist exposure (22 h) restored functional effector coupling. Up-regulation was unaffected by cycloheximide but blocked by okadaic acid. Confocal fluorescence immunocytochemistry of intact and permeabilized cells showed progressive, time-dependent increase in surface hSSTR1 labeling, associated with depletion of intracellular SSTR1 immunofluorescent vesicles. To investigate the structural domains of hSSTR1 responsible for up-regulation, we constructed C-tail deletion (Delta) mutants and chimeric hSSTR1-hSSTR5 receptors. Human SSTR5 was chosen because it internalizes readily, displays potent C-tail internalization signals, and does not up-regulate. Like wild type hSSTR1, Delta C-tail hSSTR1 did not internalize and additionally lost the ability to up-regulate. Swapping the C-tail of hSSTR1 with that of hSSTR5 induced internalization (27%) but not up-regulation. Substitution of hSSTR5 C-tail with that of hSSTR1 converted the chimeric receptor to one resembling wild type hSSTR1 (poor internalization, 71% up-regulation). These results show that ligand-induced up-regulation of hSSTR1 occurs by a temperature-dependent active process of receptor recruitment from a pre-existing cytoplasmic pool to the plasma membrane. It does not require new protein synthesis or signal transduction, is sensitive to dephosphorylation events, and critically dependent on molecular signals in the receptor C-tail.


Subject(s)
Receptors, Somatostatin/agonists , Adenylate Cyclase Toxin , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cycloheximide/pharmacology , Endocytosis , Fluoroimmunoassay , Humans , Molecular Sequence Data , Mutation , Okadaic Acid/pharmacology , Receptors, Somatostatin/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Up-Regulation , Virulence Factors, Bordetella/pharmacology
14.
J Neurochem ; 73(3): 1273-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10461921

ABSTRACT

Preprocortistatin (PPCST) has been recently identified as a novel somatostatin (SST)-related gene expressed only in brain. PPCST shares 11 of 14 residues with SST-14 at its C-terminal segment, where it features Lys-Lys and Lys-Arg basic sites for cleavage to putative cortistatin (CST)-14 and CST-29 peptides, respectively. Although synthetic replicates of the two putative CST peptides interact with SST receptors, they also display novel effects suggesting independent biological functions. Nothing is currently known about the naturally occurring mature cleavage products of PPCST posttranslational processing. Here we have cloned rat PPCST cDNA, stably expressed it in AtT-20 pituitary cells, and characterized the cellular and releasable products of PPCST processing by HPLC and radioimmunoassay using a SST-14 antibody that recognizes synthetic CST-14 and CST-29. Transfected cells released 120 +/- 21 pg of total CST-LI per plate basally, with an increase to 204 +/- 33 pg per plate with forskolin stimulation (p < 0.05). HPLC chromatograms of cell extracts revealed three peaks corresponding to CST-14, CST-29, and unprocessed PPCST (ratio, 41:55:4.5). CST was released preferentially as CST-14 (63-70%) compared with CST-29 (30-37%) under basal and forskolin-stimulated conditions. These studies demonstrate efficient processing of PPCST to both CST-14 and CST-29 through putative cleavage at both C-terminal dibasic sites of PPCST. Although the two peptides are synthesized approximately equally, CST-14 is released preferentially via the regulated secretory pathway.


Subject(s)
Protein Precursors/biosynthesis , Animals , Cell Line , Chromatography, High Pressure Liquid , Colforsin/pharmacology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Mice , Pituitary Gland/cytology , Pituitary Gland/metabolism , Protein Precursors/genetics , Radioimmunoassay , Rats , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin/biosynthesis , Transfection
15.
Proc Natl Acad Sci U S A ; 96(4): 1657-62, 1999 Feb 16.
Article in English | MEDLINE | ID: mdl-9990080

ABSTRACT

Niemann-Pick type C disease (NP-C) is an inherited neurovisceral lipid storage disorder characterized by progressive neurodegeneration. Most cases of NP-C result from inactivating mutations of NPC1, a recently identified member of a family of genes encoding membrane-bound proteins containing putative sterol sensing domains. By using a specific antipeptide antibody to human NPC1, we have here investigated the cellular and subcellular localization and regulation of NPC1. By light and electron microscopic immunocytochemistry of monkey brain, NPC1 was expressed predominantly in perisynaptic astrocytic glial processes. At a subcellular level, NPC1 localized to vesicles with the morphological characteristics of lysosomes and to sites near the plasma membrane. Analysis of the temporal and spatial pattern of neurodegeneration in the NP-C mouse, a spontaneous mutant model of human NP-C, by amino-cupric-silver staining, showed that the terminal fields of axons and dendrites are the earliest sites of degeneration that occur well before the appearance of a neurological phenotype. Western blots of cultured human fibroblasts and monkey brain homogenates revealed NPC1 as a 165-kDa protein. NPC1 levels in cultured fibroblasts were unchanged by incubation with low density lipoproteins or oxysterols but were increased 2- to 3-fold by the drugs progesterone and U-18666A, which block cholesterol transport out of lysosomes, and by the lysosomotropic agent NH4Cl. These studies show that NPC1 in brain is predominantly a glial protein present in astrocytic processes closely associated with nerve terminals, the earliest site of degeneration in NP-C. Given the vesicular localization of NPC1 and its proposed role in mediating retroendocytic trafficking of cholesterol and other lysosomal cargo, these results suggest that disruption of NPC1-mediated vesicular trafficking in astrocytes may be linked to neuronal degeneration in NP-C.


Subject(s)
Astrocytes/metabolism , Brain/pathology , Carrier Proteins , Membrane Glycoproteins , Niemann-Pick Diseases/pathology , Proteins/analysis , Amino Acid Sequence , Animals , Antibodies , Astrocytes/pathology , Axons/pathology , Axons/ultrastructure , Brain/cytology , Brain/metabolism , CHO Cells , Cells, Cultured , Cricetinae , Dendrites/pathology , Dendrites/ultrastructure , Female , Fibroblasts , Humans , Immunohistochemistry , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Macaca fascicularis , Male , Mice , Microscopy, Immunoelectron , Molecular Sequence Data , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/cytology , Neurons/pathology , Niemann-Pick C1 Protein , Niemann-Pick Diseases/metabolism , Proteins/genetics , Proteins/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Transfection
16.
Mol Endocrinol ; 13(1): 82-90, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892014

ABSTRACT

Ligand-activated somatostatin receptors (SSTRs) initiate cytotoxic or cytostatic antiproliferative signals. We have previously shown that cytotoxicity leading to apoptosis was signaled solely via human (h) SSTR subtype 3, whereas the other four hSSTR subtypes initiated a cytostatic response that led to growth inhibition. In the present study we characterized the antiproliferative signaling mediated by hSSTR subtypes 1, 2, 4, and 5 in CHO-K1 cells. We report here that cytostatic signaling via these subtypes results in induction of the retinoblastoma protein Rb and G1 cell cycle arrest. Immunoblot analysis revealed an increase in hypophosphorylated form of Rb in agonist-treated cells. The relative efficacy of these receptors to initiate cytostatic signaling was hSSTR5 > hSSTR2 > hSSTR4 approximately = hSSTR1. Cytostatic signaling via hSSTR5 also induced a marginal increase in cyclin-dependent kinase inhibitor p21. hSSTR5-initiated cytostatic signaling was G protein dependent and protein tyrosine phosphatase (PTP) mediated. Octreotide treatment induced a translocation of cytosolic PTP to the membrane, whereas it did not stimulate PTP activity when added directly to the cell membranes. C-tail truncation mutants of hSSTR5 displayed progressive loss of antiproliferative signaling proportional to the length of deletion, as reflected by the marked decrease in the effects of octreotide on membrane translocation of cytosolic PTP, and induction of Rb and G1 arrest. These data demonstrate that the C-terminal domain of hSSTR5 is required for cytostatic signaling that is PTP dependent and leads to induction of hypophosphorylated Rb and G1 arrest.


Subject(s)
G1 Phase/physiology , Receptors, Somatostatin/metabolism , Retinoblastoma Protein/metabolism , Animals , CHO Cells/drug effects , CHO Cells/metabolism , Cricetinae , Cytosol/metabolism , G1 Phase/drug effects , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Hormones/pharmacology , Humans , Membrane Proteins , Mutation , Octreotide/pharmacology , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/drug effects , Protein Tyrosine Phosphatases/metabolism , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/genetics , Retinoblastoma Protein/genetics , Somatostatin/metabolism , Somatostatin/pharmacology , Virulence Factors, Bordetella/pharmacology
17.
Diabetes ; 48(1): 77-85, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892225

ABSTRACT

We have developed a panel of rabbit polyclonal antipeptide antibodies against the five human somatostatin receptor subtypes (hSSTR1-5) and used them to analyze the pattern of expression of hSSTR1-5 in normal human islet cells by quantitative double-label confocal fluorescence immunocytochemistry. All five hSSTR subtypes were variably expressed in islets. The number of SSTR immunopositive cells showed a rank order of SSTR1 > SSTR5 > SSTR2 > SSTR3 > SSTR4. SSTR1 was strongly colocalized with insulin in all beta-cells. SSTR5 was also an abundant isotype, being colocalized in 87% of beta-cells. SSTR2 was found in 46% of beta-cells, whereas SSTR3 and SSTR4 were relatively poorly expressed. SSTR2 was strongly colocalized with glucagon in 89% of alpha-cells, whereas SSTR5 and SSTR1 colocalized with glucagon in 35 and 26% of alpha-cells, respectively. SSTR3 was detected in occasional alpha-cells, and SSTR4 was absent. SSTR5 was preferentially expressed in 75% of SST-positive cells and was the principal delta-cell SSTR subtype, whereas SSTR1-3 were colocalized in only a few delta-cells, and SSTR4 was absent. These studies reveal predominant expression of SSTR1, SSTR2, and SSTR5 in human islets. Beta-cells, alpha-cells, and delta-cells each express multiple SSTR isoforms, beta-cells being rich in SSTR1 and SSTR5, alpha-cells in SSTR2, and delta-cells in SSTR5. Although there is no absolute specificity of any SSTR for an islet cell type, SSTR1 is beta-cell selective, and SSTR2 is alpha-cell selective. SSTR5 is well expressed in beta-cells and delta-cells and moderately well expressed in alpha-cells, and thereby it lacks the islet cell selectivity displayed by SSTR1 and SSTR2. Subtype-selective SSTR expression in islet cells could be the basis for preferential insulin suppression by SSTR1-specific ligands and of glucagon inhibition by SSTR2-selective compounds.


Subject(s)
Islets of Langerhans/metabolism , Receptors, Somatostatin/metabolism , Animals , Cell Line , Glucagon/metabolism , Humans , Immunohistochemistry , Insulin/metabolism , Islets of Langerhans/cytology , Isomerism , Rabbits , Somatostatin/metabolism , Substrate Specificity , Tissue Distribution
18.
J Biol Chem ; 273(33): 21416-22, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9694905

ABSTRACT

We have investigated the role of the cytoplasmic tail (C-tail) of the human somatostatin receptor type 5 (hSSTR5) in regulating receptor coupling to adenylyl cyclase (AC) and in mediating agonist-dependent desensitization and internalization responses. Mutant receptors with progressive C-tail truncation (Delta347, Delta338, Delta328, Delta318), Cys320 --> Ala substitution (to block palmitoylation), or Tyr304 --> Ala substitution of a putative NPXXY internalization motif were stably expressed in Chinese hamster ovary K1 cells. Except for the Tyr304 --> Ala mutant, which showed no binding, all other mutant receptors exhibited binding characteristics (Kd and Bmax) and G protein coupling comparable with wild type (wt) hSSTR5. The C-tail truncation mutants displayed progressive reduction in coupling to AC, with the Delta318 mutant showing complete loss of effector coupling. Agonist pretreatment of wt hSSTR5 led to uncoupling of AC inhibition, whereas the desensitization response of the C-tail deletion mutants was variably impaired. Compared with internalization (66% at 60 min) of wt hSSTR5, truncation of the C-tail to 318, 328, and 338 residues reduced receptor internalization to 46, 46, and 23%, respectively, whereas truncation to 347 residues slightly improved internalization (72%). Mutation of Cys320 --> Ala induced a reduction in AC coupling, desensitization, and internalization. These studies show that the C-tail of hSSTR5 serves a multifunctional role in mediating effector coupling, desensitization, and internalization. Whereas coupling to AC is dependent on the length of the C-tail, desensitization and internalization require specific structural domains. Furthermore, internalization is regulated through both positive and negative molecular signals in the C-tail and can be dissociated from the signaling and acute desensitization responses of the receptor.


Subject(s)
Adenylyl Cyclases/metabolism , Cytoplasm/metabolism , Endocytosis , Receptors, Somatostatin/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , GTP-Binding Proteins/metabolism , Humans , Iodine Radioisotopes , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Receptors, Somatostatin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
19.
Gene ; 214(1-2): 45-9, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9651476

ABSTRACT

The human somatostatin receptor subtype 5 (hSSTR5) gene has previously been cloned and localized to chromosome 16 p13.3. This region is evolutionarily conserved in all vertebrate genomes from the puffer fish (Fugu rubripes) to human, and also contains loci for genes associated with two common multisystemic disorders, adult polycystic kidney disease (PKD1) and tuberous sclerosis (TSC2). Analysis of the 5' flanking region of the hSSTR5 gene has revealed consensus sequences for a number of transcription factors as well as Alu-like repeat elements. In the present study, genomic DNA from 53 unrelated individuals was analysed by PCR and Southern blots probed with radiolabeled fragments generated from different segments of the hSSTR5 gene. We have identified two restriction fragment length polymorphisms (RFLP) with high heterozygosity values at the 5' flanking region of the hSSTR5 gene. These RFLP markers will be useful for determining the allelic loss of genetic material from this region. The observed polymorphism in the promoter region may affect the function of the hSSTR5 gene.


Subject(s)
Polymorphism, Restriction Fragment Length , Receptors, Somatostatin/genetics , Adult , Animals , Base Sequence , Chromosomes, Human, Pair 16/genetics , DNA/genetics , DNA/isolation & purification , DNA Primers/genetics , Evolution, Molecular , Gene Expression , Genes, Regulator , Humans , Loss of Heterozygosity , Middle Aged , Polymerase Chain Reaction
20.
Int J Cancer ; 76(5): 620-7, 1998 May 29.
Article in English | MEDLINE | ID: mdl-9610716

ABSTRACT

Expression of mRNA for the 5 somatostatin receptors (sst1-5) was characterized by Northern blot and RT-PCR analysis in 20 meningioma and 9 glioma samples. sst1 mRNA was detectable by Northern blots of poly-A+ RNA in meningiomas but not gliomas. In contrast, sst2 mRNA was readily detected by Northern blots of total RNA as a major 2.3 kb transcript and 2 minor 4.3 kb and 8 kb transcripts in all meningiomas and 6 out of 9 gliomas. Quantitation of the 2.3 kb sst2 mRNA showed that 15 out of 20 tumors expressed 1.3- to 33-fold higher levels than control normal human brain. Mean sst2 mRNA for the 20 meningioma samples was 978% that of normal brain. Three gliomas showed 7- to 14-fold higher sst2 mRNA than normal brain whereas the remaining samples displayed very low or undetectable levels. Immunocytochemistry of meningioma and glioma samples, with a sst2-specific antibody revealed immunoreactivity in tumor cells and peritumoral tissue, with prominent expression in blood vessels. mRNA for sst3,4,5 could not be detected by Northern blots in any of the tumors. RT-PCR analysis of meningiomas and gliomas revealed the following percent of tumors positive for a given sst mRNA: sst1 (86%), sst2 (100%), sst3 (60%), sst4 (58%), and sst5 (67%); 85% of tumors expressed 3 of the 5 subtypes. No correlation was found between the pattern of expression of sst mRNA and tumor type, location, and histology for either the meningiomas or gliomas. Our results show that meningiomas and gliomas are all positive for at least one sst subtype, the majority expressing multiple subtypes. sst2 is the most abundant isoform with a rich expression in both tumor and peritumoral tissue especially blood vessels.


Subject(s)
Brain Neoplasms/ultrastructure , Glioma/ultrastructure , Meningioma/ultrastructure , Receptors, Somatostatin/biosynthesis , Receptors, Somatostatin/classification , Adult , Aged , Aged, 80 and over , Antibody Specificity , Blotting, Northern , Brain Neoplasms/metabolism , Glioma/metabolism , Humans , Immunohistochemistry , Meningioma/metabolism , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, Somatostatin/immunology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...