Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 7(4): 916-28, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24301658

ABSTRACT

Foxp3(+) regulatory T cells (Treg) have a central role for keeping the balance between pro- and anti-inflammatory immune responses against chronically encountered antigens at mucosal sites. However, their antigen specificity especially in humans is largely unknown. Here we used a sensitive enrichment technology for antigen-reactive T cells to directly compare the conventional vs. regulatory CD4(+) T-cell response directed against two ubiquitous mucosal fungi, Aspergillus fumigatus and Candida albicans. In healthy humans, fungus-specific CD4(+)CD25(+)CD127(-)Foxp3(+) Treg are strongly expanded in peripheral blood and possess phenotypic, epigenetic and functional features of thymus-derived Treg. Intriguingly, for A. fumigatus, the strong Treg response contrasts with minimal conventional T-cell memory, indicating selective Treg expansion as an effective mechanism to prevent inappropriate immune activation in healthy individuals. By contrast, in subjects with A. fumigatus allergies, specific Th2 cells were strongly expanded despite the presence of specific Treg. Taken together, we demonstrate a largely expanded Treg population specific for mucosal fungi as part of the physiological human T-cell repertoire and identify a unique capacity of A. fumigatus to selectively generate Treg responses as a potentially important mechanism for the prevention of allergic reactions.


Subject(s)
Antigens, Fungal/immunology , Epitopes, T-Lymphocyte/immunology , Fungi/immunology , Immune Tolerance , Mucous Membrane/immunology , Mucous Membrane/microbiology , T-Lymphocytes, Regulatory/immunology , Aspergillus/immunology , Cells, Cultured , Cystic Fibrosis/complications , Cystic Fibrosis/immunology , Humans , Hypersensitivity/etiology , Immunologic Memory , Immunophenotyping , Lymphocyte Count , Phenotype , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
2.
Mol Pharmacol ; 73(2): 292-304, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17981995

ABSTRACT

The somatostatin subtype 2A (sst2A) receptor, a member of the G protein-coupled receptor superfamily, mediates many of the neuroendocrine and neuromodulatory actions of somatostatin, and it represents the primary target for somatostatin analogs used in cancer therapy and tumor localization. Agonist stimulation leads to the rapid phosphorylation, endocytosis, and desensitization of the sst2A receptor; however, little is known about the role of phosphorylation in sst2A regulation. sst2A phosphorylation occurs on serine and threonine residues in the third intracellular loop and carboxyl terminus. Therefore, we generated mutant receptors in which serine (Ser-), threonine (Thr-), or both (Ser-/Thr-) residues in these regions were mutated to alanine. In contrast to the wild-type receptor, somatostatin treatment did not stimulate the phosphorylation of the Ser-/Thr- mutant, and it did not produce desensitization. Furthermore, internalization of the Ser-/Thr- mutant occurred 5 times more slowly than with the wild-type receptor. Mutating only the Ser residues did not inhibit either internalization or desensitization. In contrast, mutating only the Thr residues inhibited receptor endocytosis to the same extent as in the full mutant, but it did not affect receptor desensitization. In both the wild-type and Ser- receptors, agonist binding produced a stable arrestin-receptor complex that was maintained during receptor trafficking, whereas arrestin was not recruited to either the Thr- or the Ser-/Thr- receptors. These results demonstrate that agonist-stimulated receptor phosphorylation is necessary for both desensitization and rapid internalization of the sst2A receptor. However, sst2A receptor internalization and uncoupling can occur independently, involve different receptor phosphorylation sites, and exhibit different requirements for stable arrestin association.


Subject(s)
Arrestin/metabolism , Receptors, Somatostatin/metabolism , Amino Acid Sequence/physiology , Animals , Arrestin/genetics , Binding Sites/physiology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Molecular Sequence Data , Phosphorylation , Protein Binding/physiology , Rats , Receptors, Somatostatin/genetics
3.
Gut ; 53(10): 1431-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15361490

ABSTRACT

BACKGROUND AND AIMS: Many neuroendocrine gastrointestinal tumours express receptors for the regulatory peptide somatostatin. Among the five existing somatostatin receptor (sst) subtypes, sst2A is the most frequently expressed in these tumours. However, little information is available about the cellular location of sst2A in corresponding non-neoplastic epithelial tissues. METHODS: We searched for sst2A immunoreactive cells in non-neoplastic gastrointestinal tissues, and evaluated their number and immunohistochemical characteristics with neuroendocrine markers. RESULTS: The gastric antrum showed numerous sst2A cells, situated in the epithelium, corresponding to gastrin containing neuroendocrine cells, while the gastric corpus was largely devoid of sst2A cells, including enterochromaffin-like cells. The remaining foregut, namely the duodenum and proximal jejunum, also contained a large number of sst2A cells, all being neuroendocrine cells and many of them characterised as gastrin cells. Sst2A cells were also detected in the midgut, in low numbers in the epithelium of the distal jejunum and ileum, but not in the appendix vermiformis, the caecum, or the hindgut, despite the large number of neuroendocrine cells present in this area. In addition, sst2A cells were found in the whole gastrointestinal tract in the myenteric and submucosal plexus. CONCLUSIONS: While sst2A receptors on antral gastrin cells presumably mediate somatostatin inhibition of gastrin secretion, the effects of somatostatin on motility and ion transport in the lower gastrointestinal tract may be mediated by sst2A receptors in the neural plexus. These data provide a molecular basis for the physiological actions of somatostatin in human gastrointestinal tissue.


Subject(s)
Enteroendocrine Cells/metabolism , Receptors, Somatostatin/metabolism , Gastric Mucosa/metabolism , Humans , Immunoenzyme Techniques , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Pyloric Antrum/metabolism , Somatostatin/physiology
4.
Ann N Y Acad Sci ; 1014: 132-6, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15153427

ABSTRACT

Many neuroendocrine gastrointestinal and lung tumors express sst2A somatostatin receptors. Because the cellular location of sst2A in the corresponding non-neoplastic tissue is unknown, we searched for sst2A immuno-reactive cells and characterized their type in these tissues using a highly specific sst2A antibody (R2-88). Epithelial sst2A cells, identified as neuroendocrine, gastrin-producing cells, were found in large numbers in the antrum and the duodenum, but not in the gastric corpus. They were also present in the proximal jejunum, rarely noted in the distal jejunum and ileum, and absent in the large intestine and the appendix vermiformis. Moreover, sst2A cells were found abundantly in the neural plexus. sst2A receptors on antral gastrin cells could mediate somatostatin inhibition on gastrin secretion, whereas those in the neural plexus could mediate somatostatin effects on motility and ion transport in the lower gastrointestinal tract. Rare sst2A cells in bronchi and bronchioles located basally and parabasally in the gastrointestinal epithelium were detected that could represent stem/progenitor cells. It is currently not clear whether and which of the identified sst2A cells are at the origin of sst2A-positive neuroendocrine gut or lung tumors.


Subject(s)
Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Lung Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Respiratory Mucosa/metabolism , Gastrins/metabolism , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology , Lung Neoplasms/pathology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Respiratory Mucosa/pathology
5.
J Biol Chem ; 276(5): 3709-17, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11073961

ABSTRACT

The sst1 somatostatin (SRIF) receptor subtype is widely expressed in the endocrine, gastrointestinal, and neuronal systems as well as in hormone-sensitive tumors, yet little is known about its regulation. Here we investigated the desensitization, internalization, and phosphorylation of sst1 expressed in CHO-K1 cells. Treatment of cells with 100 nm SRIF for 30 min reduced maximal SRIF inhibition of adenylyl cyclase from 40 to 10%. This desensitization was rapid (t(12) < 2 min) and dependent on agonist concentration (EC(50) = 2 nm). However, internalization of receptor-bound ligand occurred slowly (t(12) > 180 min). Incubation of cells with SRIF also caused a rapid (t(12) < 2 min) increase in sst1 receptor phosphorylation in a dose-dependent manner (EC(50) = 1.3 nm), as determined in a mobility shift phosphorylation assay. Receptor phosphorylation was not affected by pertussis toxin, indicating a requirement for receptor occupancy rather than signaling. The protein kinase C activator, phorbol 12-myristate 13-acetate also stimulated sst1 receptor phosphorylation whereas forskolin did not. Both agonist- and phorbol 12-myristate 13-acetate-stimulated receptor phosphorylation occurred mainly on serine. These studies are the first to demonstrate phosphorylation of the sst1 receptor and suggest that phosphorylation mediated uncoupling, rather than sequestration, leads to its desensitization.


Subject(s)
Endocytosis/physiology , Receptors, Somatostatin/metabolism , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Phosphoamino Acids , Phosphorylation , Time Factors
6.
J Clin Endocrinol Metab ; 85(10): 3882-91, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11061553

ABSTRACT

The distribution of the sst2A receptor was investigated, using immunohistochemistry, with the specific antipeptide antibody R2-88, in a total of 120 tumors of the nervous and the neuroendocrine systems, including small-cell lung carcinomas, medulloblastomas, neuroblastomas, pheochromocytomas, and paragangliomas. The great majority of the tumor samples, frozen or formalin-fixed, showed a positive immunohistochemical staining with R2-88, and an excellent correlation with receptor autoradiography using 125I[Tyr3]-octreotide. Whereas small-cell lung carcinomas and medulloblastomas had a predominantly plasma membrane staining, pheochromocytomas and neuroblastomas had variable ratios of cell surface and intracellular staining. Strikingly, a preferentially cytoplasmic staining was seen in tumors with a high level of somatostatin gene expression, whereas a more plasma membranous staining was seen in tumors lacking somatostatin messenger RNA. Specificity of both the plasma membrane and the cytoplasmic staining pattern was confirmed in immunoblots, which showed the immunoreactive receptor migrating as a characteristic 70-kDa broad band. In both immunohistochemical and immunoblotting experiments, staining was abolished by antibody blockade with 100 nM antigen peptide. These results describe, for the first time, the localization of the sst2A receptor protein in human small-cell lung carcinomas, medulloblastomas, neuroblastomas, and paragangliomas. Moreover, it is the first report investigating possible causes for distinct subcellular localizations of sst2A in human tissues. We show that the subcellular distribution of the receptor may be dependent on the surrounding somatostatin concentration, consistent with both the known effect of somatostatin to cause sst2A receptor internalization and an autocrine regulation of tumors by the peptide they produce. Moreover, our demonstration that the sst2A receptor can be identified in this group of tumors using simple immunohistochemical methods in formalin-fixed, paraffin-embedded material opens numerous diagnostic, therapeutic, and prognostic opportunities.


Subject(s)
Nervous System Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Receptors, Somatostatin/metabolism , Subcellular Fractions/metabolism , Autoradiography , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Intracellular Membranes/metabolism , Membranes/metabolism , Octreotide , Somatostatin/biosynthesis
7.
Synapse ; 38(2): 177-86, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11018792

ABSTRACT

Binding of neuropeptides to their receptors usually results in internalization of receptor-ligand complexes. This process serves a crucial role in receptor downregulation, resensitization, and transmembrane signaling. It has mainly been investigated in cells ectopically expressing recombinant receptors. In the present study, we investigated whether rat central neurons and astrocytes naturally expressing somatostatin (SRIF) receptors internalized this neuropeptide. We demonstrated that 29% of cortical and 45% of hippocampal neurons in culture expressed the SRIF receptor sst(2A) and that 40-50% of the neurons internalized fluorescent SRIF. Similarly, an important proportion of astrocytes expressed sst(2A) (up to 60% in cortical cultures) and internalized fluo-SRIF. Competition experiments using the sst(2)/sst(5)-preferring agonist SMS 201-995 (octreotide) showed that a subpopulation of neurons internalized fluo-SRIF via sst(2) and/or sst(5) receptors, but that others also did so via other subtypes. Fluo-SRIF labeling was barely competed for by the sst(1)-selective agonist CH-275, indicating that sst(1) was unlikely to be mediating SRIF internalization in hippocampal and cortical neurons. Given the paucity of sst(5) receptors in cerebral cortex and hippocampus and the poor yield of sst(4) internalization in transfected cells, we conclude that sst(2) and sst(3) subtypes are the most likely to be responsible for SRIF internalization in our culture systems.


Subject(s)
Astrocytes/metabolism , Neurons/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Animals , COS Cells/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Female , Hippocampus/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley
8.
J Neurosci ; 20(16): 5932-9, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10934240

ABSTRACT

To investigate the effects of somatostatin (somatotropin release-inhibiting factor, SRIF) on the regulation of SST(2A) receptors in mammalian brain, we examined how blockade of SRIF release or stimulation by the SRIF analog [d-Trp(8)]-SRIF would affect the expression and cell surface availability of SST(2A) receptors in rat brain slices. First, we measured the intensity of SST(2A) immunoreactivity, using quantitative light microscopic immunocytochemistry, and levels of SST(2A) mRNA, using semiquantitative RT-PCR, under conditions of acute SRIF release blockade. Incubation of slices from the claustrum or basolateral amygdala, two regions previously shown to contain high concentrations of SST(2A) receptors, in Ca(2+)-free Ringer's for 40 min induced a decrease in the intensity of SST(2A) receptor immunoreactivity and concentration of SST(2A) mRNA as compared with control values obtained in Ca(2+)-supplemented Ringer's. These effects were counteracted in a dose-dependent manner by the addition of 10-100 nm [d-Trp(8)]-SRIF to the Ca(2+)-free medium. Furthermore, both of these effects were abolished in the presence of the endocytosis inhibitors phenylarsine oxide or hyperosmolar sucrose, suggesting that they were dependent on receptor internalization. Electron microscopic immunogold labeling confirmed the existence of an agonist-induced internalization of SST(2A) receptors in central neurons. At a high (10 microm), but not at a low (10 nm), concentration of agonist this internalization resulted in a significant decrease in cell surface receptor density, irrespective of the presence of Ca(2+) in the medium. Taken together, these results suggest that ligand-induced endocytosis is responsible for rapid transcriptional (increase in SST(2A) expression) and trafficking (loss of cell surface receptors) events involved in the control of the somatostatinergic signal.


Subject(s)
Brain/drug effects , Neurons/drug effects , Receptors, Cell Surface/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Amygdala/drug effects , Amygdala/metabolism , Amygdala/ultrastructure , Animals , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Basal Ganglia/ultrastructure , Brain/metabolism , Brain/ultrastructure , In Vitro Techniques , Male , Neurons/metabolism , Neurons/ultrastructure , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/drug effects , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/genetics , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
9.
J Clin Endocrinol Metab ; 85(4): 1719-26, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10770220

ABSTRACT

Somatostatin (SS) and SS receptor (SSR) subtypes, code-named sst1-5, are heterogeneously expressed in the normal human thymus. This suggests their involvement in controlling the immune and/or neuroendocrine functions in this organ. Moreover, recently a high in vivo uptake of [111In-DTPA-D-Phe1]octreotide has been reported in patients bearing thymoma. The present study characterizes in vivo and in vitro, functional SS-binding sites in a human thymoma. A high uptake of [111In-DTPA-D-Phe1]octreotide was observed in the chest of a patient with myasthenia gravis due to a cortical thymoma. Specific binding of [125I-Tyr11] SS-14 was found on a membrane preparation of the surgically removed thymoma. Scatchard analysis showed high affinity binding sites (Kd, 47.5 +/- 2.5 pmol/L) with low maximum binding capacity (23.5 +/- 2.5 fmol/mg membrane protein). RT-PCR analysis showed the presence of sst1, sst2A, and a predominant sst3 messenger RNA (mRNA) expression in the tumor tissue. Primary cultured tumor cells expressed sst3 mRNA only. In contrast to the normal thymus, SS mRNA was not expressed. By immunohistochemistry, the tumor cells highly expressed sst3 receptors, weakly expressed sst1 receptors, and showed no immunostaining for sst2A receptors. sst2A immunoreactivity was found in the stromal compartment of the tumor, particularly on the endothelium of small intratumoral blood vessels. In primary cultured tumor cells, both SS and octreotide (10 nmol/L) significantly inhibited [3H]thymidine incorporation by 40.6% and 43.2%, respectively. The following conclusions were reached. 1) As this tumor displayed a high immunoreactivity for sst3 and the cultured tumor cells expressed the sst3 mRNA only, this SSR may be the subtype involved in the inhibition of epithelial tumor cell proliferation by octreotide in vitro. 2) A loss of endogenous SS production in this thymoma might be implicated in the uncontrolled cell growth. 3) In this case, the sst3 may play a role in determining the uptake of [111In-DTPA-D-Phe1]octreotide by in vivo SS receptor scintigraphy.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cell Division/drug effects , Octreotide/pharmacology , Receptors, Somatostatin/analysis , Thymoma/chemistry , Thymus Neoplasms/chemistry , Female , Gene Expression , Humans , Immunohistochemistry , Indium Radioisotopes , Middle Aged , Octreotide/analogs & derivatives , Octreotide/metabolism , Pentetic Acid/analogs & derivatives , Pentetic Acid/metabolism , RNA, Messenger/analysis , Radionuclide Imaging , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin/genetics , Somatostatin/pharmacology , Thymoma/diagnostic imaging , Thymoma/pathology , Thymus Neoplasms/diagnostic imaging , Thymus Neoplasms/pathology , Tumor Cells, Cultured
10.
J Biol Chem ; 275(8): 5591-9, 2000 Feb 25.
Article in English | MEDLINE | ID: mdl-10681540

ABSTRACT

The sst2A receptor is expressed in the endocrine, gastrointestinal, and neuronal systems as well as in many hormone-sensitive tumors. This receptor is rapidly internalized and phosphorylated in growth hormone-R2 pituitary cells following somatostatin binding (Hipkin, R. W., Friedman, J., Clark, R. B., Eppler, C. M., and Schonbrunn, A. (1997) J. Biol. Chem. 272, 13869-13876). The protein kinase C (PKC) activator, phorbol 12-myristate 13-acetate (PMA), also stimulates sst2A phosphorylation. Here we examine the mechanisms and consequences of PMA and agonist-induced sst2A phosphorylation. Like somatostatin, both PMA and bombesin increased sst2A receptor phosphorylation within 2 min. The PKC inhibitor GF109203X blocked PMA- and bombesin- stimulated sst2A phosphorylation, whereas stimulation by the somatostatin analog SMS 201-995 was unaffected. Agonist and PMA each stimulated phosphorylation in two receptor domains, the third intracellular loop and the C-terminal tail. Functionally, PMA dramatically increased the internalization of the sst2A receptor-ligand complex. This PMA stimulation was blocked by GF109203X, whereas basal internalization was unaffected. However, neither basal nor PMA-stimulated internalization was altered by pertussis toxin, whereas both were blocked by hypertonic sucrose. Therefore PKC activation and agonist binding stimulate sst2A phosphorylation by distinct mechanisms, and PKC potentiates internalization of the sst2A receptor via clathrin-coated pits. Thus, hormonal stimulation of PKC-coupled receptors may provide a mechanism for regulating the inhibitory actions of somatostatin in target tissue.


Subject(s)
Protein Kinase C/metabolism , Receptors, Somatostatin/metabolism , Animals , Bombesin/pharmacology , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Peptide Mapping , Phosphorylation/drug effects , Pituitary Gland/enzymology , Protein Structure, Secondary , Rats , Receptors, Somatostatin/agonists , Temperature , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Tumor Cells, Cultured
11.
Endocrinology ; 140(10): 4739-44, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10499533

ABSTRACT

The capacity of the pituitary to suppress hormone secretion in response to somatostatin (SRIF) is markedly age dependent. Immature pituitaries are relatively resistant to SRIF effects, and increasing sensitivity to SRIF with advancing age is believed to cause characteristic developmental changes in pituitary hormone secretion in mammals. However, the cellular mechanism(s) underlying this developmental pattern of response to SRIF are not understood. Because somatostatin receptors (ssts) are critical mediators of SRIF's actions on target tissues, we investigated the expression of sst1, sst2, sst3, sst4, and sst5 messenger RNA (mRNA) in pituitaries of developing and mature rats. Animals were studied at embryonic day 19.5, and at postnatal days 2, 12, 30, 45, 70, and 1 yr; these ages correspond to major changes in circulating GH levels and pituitary responsiveness to SRIF. Pituitary levels of sst2 mRNA increased strikingly and progressively with advancing age after birth (F = 30.92, P < 0.0001). Compared with 2-day-old pituitaries, sst2 mRNA abundance rose 3.25-fold by 12 days of age and 6-fold by 70 days of age. Moreover, Western blot analysis indicated a marked increase in pituitary expression of sst2A protein with advancing age. By contrast, pituitary abundance of sst1, sst3, sst4, and sst5 mRNAs did not differ with age. To assess the role of endogenous SRIF in regulating perinatal sst2 gene expression, we also administered a well-characterized SRIF antiserum (or NSS as controls; 10 microl/10 g) sc daily from postnatal days 2 to 12 of life. Treatment with SRIF antiserum raised GH levels but did not alter pituitary sst2 mRNA abundance, compared with controls. Taken together, these data indicate that 1) the perinatal rat pituitary expresses the same complement of ssts as the adult pituitary; 2) expression of ssts is developmentally regulated in a highly subtype-specific manner; 3) pituitary sst2 mRNA and sst2A protein increase markedly and progressively with advancing age after birth; and 4) the perinatal rise in sst2 mRNA levels is unlikely to be regulated by endogenous SRIF. The finding of subtype-specific, developmentally determined sst expression indicates a novel and potentially fundamental mechanism of sst regulation, and suggests a molecular mechanism underlying developmental maturation in the capacity of the pituitary to respond to SRIF.


Subject(s)
Aging/metabolism , Animals, Newborn/metabolism , Fetus/metabolism , Peptide Fragments/metabolism , Pituitary Gland/metabolism , Receptors, Somatostatin/metabolism , Animals , Animals, Newborn/growth & development , Immune Sera/pharmacology , Immunoblotting , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/chemistry , Somatostatin/immunology
12.
J Clin Endocrinol Metab ; 84(8): 2942-50, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10443702

ABSTRACT

The cellular distribution of the somatostatin sst2A receptor protein was investigated in the lymphatic, smooth muscular, and nervous components of the human gastrointestinal tract using subtype-specific antibody R2-88 for immunohistochemical staining of cryostat and formalin-fixed, paraffin-embedded tissue sections. Germinal centers of intestinal lymphatic follicles were immunostained, exhibiting a predominantly plasma membrane localization of the receptor. Similarly, nerve fibers and cells in the submucosal and myenteric plexus were stained for sst2A. Antibody preabsorption with 100 nmol/L antigen peptide abolished staining in all of these tissues, and immunohistochemical staining correlated with the labeling observed after receptor autoradiography using the sst2-preferring radioligand 125I-[Tyr3]octreotide. Cytoplasmic immunostaining was detected in gastrointestinal smooth muscle cells and was inhibited by antibody pre-absorption with antigen peptide. However, 125I-[Tyr3]octreotide autoradiography was negative, and Western blots showed no band at the usual 70-90 kDa location for sst2A. Instead, a band was observed at 205 kDa. This band comigrated with the rabbit myosin standard, which was also stained with R2-88, although antibody sensitivity for myosin was less than 0.002% of that for the sst2A receptor. Rigorous computer-based sequence analysis demonstrated the peptide sequence chosen for antibody production was unique. Moreover, standard sequence alignment protocols were unable to identify the sequences in myosin responsible for the observed reactivity with the R2-88 antiserum. The observed cross-reactivity emphasizes the need for extensive controls to prove the specificity of immunostaining for such low abundance proteins as receptors even when the peptide sequence chosen for antibody production is unique. This study demonstrates for the first time the presence of specific sst2A receptor protein by immunohistochemistry in the human gastrointestinal lymphatic and nervous components, but not in gastrointestinal circular and longitudinal smooth muscle.


Subject(s)
Digestive System/chemistry , Lymphatic System/chemistry , Muscle, Smooth/chemistry , Peripheral Nervous System/chemistry , Receptors, Somatostatin/analysis , Animals , Artifacts , Cross Reactions , Humans , Immunohistochemistry , Myosins/chemistry , Myosins/immunology , Rabbits , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/immunology , Sequence Alignment
13.
Endocrinology ; 140(8): 3790-6, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433240

ABSTRACT

Somatostatin, also known as somatotropin release-inhibiting factor (SRIF), is secreted by pancreatic delta-cells and inhibits the secretion of both insulin and glucagon. SRIF initiates its actions by binding to a family of six G protein-coupled receptors (sst1, -2A, -2B, -3, -4, and -5) encoded by five genes. Messenger RNA for both sst2 and sst5 have been reported in the rat pancreas, and the sst2A receptor protein has been localized to rat pancreatic alpha and pancreatic polypeptide-secreting cells in the islets as well as to pancreatic acinar cells. In this study we have used double immunostaining to show that the sst5 protein is expressed exclusively in the beta-cells of rat pancreatic islets and localizes with insulin-secreting alpha-cells. The sst5 receptor is not colocalized with sst2A. Thus, in the rat SRIF inhibits pancreatic insulin and glucagon secretion via different sst receptor subtypes.


Subject(s)
Insulin/analysis , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Receptors, Somatostatin/analysis , Receptors, Somatostatin/genetics , Alternative Splicing , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Humans , Immunohistochemistry , Insulin/metabolism , Insulin Secretion , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Rats , Receptors, Somatostatin/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic , Transfection
14.
Ann Oncol ; 10 Suppl 2: S17-21, 1999.
Article in English | MEDLINE | ID: mdl-10399028

ABSTRACT

Genes for five somatostatin receptor subtypes, designated sst1-5, have been cloned and shown to belong to the seven transmembrane domain receptor family. The sst2 mRNA transcript is alternatively spliced to generate two related receptor products (sst2A and sst2B) which differ in their carboxylterminal sequence whereas each of the other genes is transcribed to give a single unique receptor protein. The six sst receptor subtypes all bind SRIF14, SRIF28 and the cortistatins with high affinity but vary in their affinity for analogs, such as octreotide. Although the tissue distribution of sst mRNAs has been extensively examined, much less is known about the cellular distribution of the individual receptor proteins. Recent studies with sst subtype specific antibodies have localized individual sst receptors to specific cell types within the rat gastrointestinal tract, pancreas, pituitary and brain. Furthermore, sst receptors have recently been identified in human tumors by immunocytochemistry, providing a significantly improved method for sst receptor detection. All six sst receptor subtypes are linked to guanine nucleotide binding proteins (G proteins) and lead to inhibition of adenylyl cyclase following hormone binding. The sst receptors also regulate a variety of different effectors via G proteins, including calcium and potassium channels and serine and tyrosine phosphatases. In addition to signalling, two other processes are activated by hormone binding: receptor desensitization and receptor internalization. The extent to which these occur seems to vary for the different receptor subtypes. Recent studies have shown that the sst2A receptor is rapidly phosphorylated upon hormone binding, suggesting that this phosphorylation may be responsible for the desensitization and/or internalization of this receptor. The importance of receptor regulation in cellular responsiveness to somatostatin and for receptor detection as well as the molecular mechanisms by which these processes occur provide important areas for future investigations.


Subject(s)
Receptors, Somatostatin/physiology , Animals , Forecasting , Humans , Receptors, Somatostatin/classification , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism
15.
Eur J Clin Invest ; 29(7): 630-6, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10411670

ABSTRACT

BACKGROUND: In a previous study, we demonstrated the presence of receptors for somatostatin, a neuropeptide with immunoregulatory properties, in the inflammatory lesions of patients suffering from sarcoidosis and other granulomatous diseases by in vivo somatostatin receptor scintigraphy and in vitro autoradiography. However, it was not possible to identify exactly which cell types expressed the somatostatin receptors and which subtype was expressed. In this study we used a polyclonal antiserum directed against the sst2A receptor to identify more accurately the sst2A-expressing cells in sarcoidosis and other granulomatous diseases. DESIGN: Tissue biopsies from 12 patients with sarcoidosis, one patient with giant cell arteritis and one patient with Wegener's granulomatosis were studied by immunohistochemistry with the sst2A-specific antiserum. Two of the sarcoidosis patients were treated with the somatostatin analogue octreotide (100 microg t.i.d.). RESULTS: Epithelioid cells, multinucleated giant cells and a subset of CD68+ macrophages stained positive for sst2A in 9 out of 12 of the sarcoid biopsies and in both non-sarcoid granuloma biopsies. Treatment with octreotide resulted in clinical improvement in one out of two treated patients. CONCLUSION: The identification of somatostatin receptors on granuloma macrophages, epithelioid cells and giant cells, and the successful treatment of one patient with sarcoidosis with a somatostatin analogue, may offer new possibilities for treatment of granulomatous diseases.


Subject(s)
Giant Cell Arteritis/pathology , Granulomatosis with Polyangiitis/pathology , Receptors, Somatostatin/analysis , Sarcoidosis/pathology , Adult , Aged , Autoradiography , Biopsy , Female , Giant Cell Arteritis/drug therapy , Giant Cell Arteritis/immunology , Granulomatosis with Polyangiitis/drug therapy , Granulomatosis with Polyangiitis/immunology , Humans , Immunohistochemistry , Iodine Radioisotopes , Lymph Nodes/pathology , Macrophages/immunology , Macrophages/pathology , Male , Middle Aged , Octreotide/therapeutic use , Sarcoidosis/drug therapy , Sarcoidosis/immunology , Skin/pathology , Tritium
16.
J Rheumatol ; 26(3): 532-5, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10090157

ABSTRACT

OBJECTIVE: To identify the somatostatin receptor-expressing cells in rheumatoid synovium using a recently developed antiserum directed against the somatostatin receptor subtype 2A (sst2A). METHODS: We carried out immunohistochemical studies of synovial biopsies from 7 patients with rheumatoid arthritis (RA) and one non-RA patient, using a rabbit polyclonal antiserum directed against sst2A and monoclonal antibodies directed against phenotypic markers. RESULTS: SSt2A was expressed by the endothelial cells of the synovial venules but also by a subset of synovial macrophages. CONCLUSION: The identification of somatostatin receptors on macrophages, which are thought to be important effector cells in RA, may offer mechanistic insights into the potential therapeutic effect of somatostatin (analogs) in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Receptors, Somatostatin/metabolism , Synovial Membrane/metabolism , Animals , Antibodies, Monoclonal/analysis , Arthritis, Rheumatoid/pathology , Autoradiography , Biomarkers/analysis , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Immunoenzyme Techniques , Microcirculation , Rabbits , Receptors, Somatostatin/immunology , Synovial Membrane/blood supply , Synovial Membrane/pathology
17.
J Clin Endocrinol Metab ; 84(2): 775-80, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10022452

ABSTRACT

Although in situ hybridization has been used to examine the distribution of messenger RNA for somatostatin receptor subtypes (sst) in human tumors, the cellular localization of sst1 and sst2A receptors has not been reported. In this study, we describe the cellular localization of human sst1 and sst2A receptor proteins in both cryostat- and paraffin-embedded sections of 25 human tumor tissues using two recently developed polyclonal antibodies. Six somatostatin (SS) receptor (SSR) positive tumors (two gastrinomas, three carcinoids, one pheochromocytoma) and one SSR negative tumor (renal cell carcinoma), selected by positive and negative SSR autoradiography, respectively, were studied by both immunohistochemistry and Western blot analysis. The six SSR positive tumors expressed sst2A, while 4 of 5 expressed sst1 as well. The SSR negative tumor did not express either sst1 or sst2A. Western blot analysis of wheat germ agglutinin purified membrane proteins confirmed the presence of the sst1 and sst2A glycosylated receptors. The paraffin-embedded sections gave best information with respect to the subcellular localization. Sst1 immunoreactivity was observed both on the membrane and in the cytoplasm, while sst2A showed predominantly membrane-associated immunoreactivity. This subcellular distribution of sst1 or sst2A receptors was confirmed in paraffin-embedded sections of 8 additional intestinal carcinoids, 5 gastrinomas and 5 pheochromocytomas. Sst1 receptors were detected in 7 out of 8 carcinoids, in all gastrinomas, and in 4 out of 5 pheochromocytomas, while 6 out of 8 carcinoids, all gastrinomas, and 3 out of 5 pheochromocytomas expressed sst2A receptors. In conclusion, sst1 and sst2A receptors show a differential subcellular localization in human SSR positive tumors. The use of SSR subtype selective antibodies to detect the subcellular distribution of SSR subtypes in individual tumor cells is an important step forward to understand more about the pathophysiological role of the different SSR subtypes in human tumors.


Subject(s)
Immunohistochemistry , Neoplasms/chemistry , Receptors, Somatostatin/analysis , Adrenal Gland Neoplasms/chemistry , Carcinoid Tumor/chemistry , Carcinoma, Renal Cell/chemistry , Gastrinoma/chemistry , Glycosylation , Humans , Immunoblotting , Intestinal Neoplasms/chemistry , Pancreatic Neoplasms/chemistry , Pheochromocytoma/chemistry , RNA, Messenger/analysis , Receptors, Somatostatin/genetics
18.
Mol Pharmacol ; 54(5): 889-98, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9804624

ABSTRACT

The mechanisms regulating receptor internalization are not well understood and vary among different G protein-coupled receptors. The bombesin (Bn)/gastrin-releasing peptide receptor GRP-R, which is coupled to phospholipase C via the Gq family of transducing proteins, is internalized rapidly after Bn binding. Agonist stimulation leads to rapid receptor phosphorylation, as does activation of protein kinase C (PKC) by phorbol-12-myristate-13-acetate (PMA). However, agonist- and PMA-induced phosphorylation occur at different receptor sites. Here, we examined the role of PKC in GRP-R internalization after agonist and antagonist binding. We synthesized [D-Tyr6]Bn(6-13)propylamide ([D-Tyr6]Bn(6-13)PA) and found that it potently inhibited Bn-stimulated insulin release and [125I-Tyr4]Bn binding (Ki = 4.72 nM) in the HIT-T15 pancreatic cell line. The radiolabeled antagonist peptide, [125I-D-Tyr6]Bn(6-13)PA, bound with high affinity (KD = 0.29 nM at 4 degrees) to a single class of receptor sites, and competition binding studies exhibited the analog specificity expected for the GRP-R subtype. Although the agonist [125I-Tyr4]Bn was internalized rapidly at 37 degrees and subsequently degraded, [125I-D-Tyr6]Bn(6-13)PA was not internalized and was released into the medium mainly as intact peptide. The lysosomal inhibitor chloroquine (200 microM) increased the intracellular accumulation of [125I-Tyr4]Bn but had no effect on the subcellular distribution of [125I-D-Tyr6]Bn(6-13)PA. Consistent with these observations, the treatment of cells with 100 nM Bn at 37 degrees reduced cell surface receptors within minutes, whereas [D-Tyr6]Bn(6-13)PA had no effect. The addition of PMA did not induce the internalization of antagonist-occupied receptors, but pharmacological inhibition of PKC decreased the rate of agonist-induced receptor internalization. These results therefore demonstrate that although PKC contributes to agonist-induced internalization of the GRP-R, it does not elicit receptor internalization of the antagonist-occupied receptor.


Subject(s)
Protein Kinase C/physiology , Receptors, Bombesin/metabolism , Receptors, Cell Surface/physiology , Animals , Bombesin/analogs & derivatives , Bombesin/metabolism , Bombesin/pharmacology , Carcinogens/pharmacology , Cell Line , Enzyme Activation , Insulin/metabolism , Insulin Secretion , Kinetics , Pancreas/drug effects , Pancreas/metabolism , Receptors for Activated C Kinase , Receptors, Bombesin/agonists , Receptors, Bombesin/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
19.
J Clin Endocrinol Metab ; 83(10): 3746-9, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9768695

ABSTRACT

Somatostatin and octreotide inhibit endocrine pancreatic functions in man, via specific somatostatin receptors. However, the cellular distribution of the different somatostatin receptor subtype proteins has not been determined in the human pancreas. Here, the immunohistochemical distribution of the sst2A receptor was investigated using the sst2A receptor specific anti-peptide antibody R2-88 in cryostat as well as in formalin-fixed paraffin-embedded sections of human pancreatic tissue, and compared with insulin, glucagon and somatostatin immunostaining of adjacent sections. All pancreatic islets were immunostained with R2-88. Most islet cells were labeled: the sst2A receptors were present in insulin as well as glucagon producing cells, but were not detected in intra-islet vessels nor in adjacent acinar tissue. Absorption of the sst2A antibody with 100 nM of the antigen peptide abolished specific staining in tissue sections. Immunohistochemical staining with R2-88 correlated with the labeling observed after receptor autoradiography using the sst2-preferring radioligand, 125I-Tyr3-octreotide. Therefore, the clinical efficacy of octreotide on glucagon and insulin release can be explained by the presence of sst2A receptors in human A and B pancreatic islet cells. Moreover, absence of sst2A receptors in human acinar tissue suggests that the action of somatostatin on pancreatic exocrine secretion is mediated either indirectly or through a different somatostatin receptor subtype on acinar cells.


Subject(s)
Islets of Langerhans/metabolism , Receptors, Somatostatin/metabolism , Glucagon/metabolism , Humans , Immunohistochemistry , Infant, Newborn , Insulin/metabolism , Pancreatic Neoplasms/metabolism , Reference Values , Tissue Distribution
20.
Am J Pathol ; 153(1): 233-45, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9665484

ABSTRACT

Human tumors frequently express somatostatin receptors. However, none of the receptor subtype proteins have been individually visualized in normal or neoplastic human tissues. Here, the distribution of the sst2A receptor was investigated using immunohistochemistry with the specific anti-peptide antibody R2-88 in 47 human tumors. All tumors selected for their abundance of sst2 mRNA and/or strong binding of the sst2-preferring ligand 125I-labeled Tyr3-octreotide were specifically immunostained with R2-88. Conversely, all tumors without somatostatin binding or expressing predominantly other somatostatin receptor subtype mRNAs (sst1 or sst3) were not specifically immunostained by R2-88. Specificity was shown in immunoblots, demonstrating receptor migration as a 70-kd broad band. In immunohistochemical and immunoblotting experiments, the abolition of staining after antibody blockade with antigen peptide was demonstrated. Immunostaining was identified in cryostat and in formalin-fixed, paraffin-embedded sections. Heat-induced epitope retrieval was necessary to visualize sst2A receptors in formalin-fixed sections. Moreover, because of occasional high nonspecific staining, the demonstration of complete abolition of immunostaining by treatment with antigen peptide was a prerequisite for the correct identification of sst2A-positive tumors. The sst2A receptors were clearly located at the membrane of the tumor cells. These results provide the first localization of a somatostatin receptor subtype in human tissues at the cellular level. The sst2A receptor identification and visualization in tumors with simple immunohistochemical methods in formalin-fixed, paraffin-embedded material will open new diagnostic opportunities for pathologists.


Subject(s)
Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Animals , Blotting, Western , CHO Cells , Cricetinae , Humans , Immunohistochemistry/methods , In Situ Hybridization , Paraffin Embedding , Rats , Tissue Fixation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...