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










Publication year range
1.
Eur J Pharmacol ; 973: 176587, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38642667

ABSTRACT

Agonist-induced phosphorylation is a crucial step in the activation/deactivation cycle of G protein-coupled receptors (GPCRs), but direct determination of individual phosphorylation events has remained a major challenge. We have recently developed a bead-based immunoassay for the quantitative assessment of agonist-induced GPCR phosphorylation that can be performed entirely in 96-well plates, thus eliminating the need for western blot analysis. In the present study, we adapted this assay to three novel phosphosite-specific antibodies directed against the neurokinin 1 (NK1) receptor, namely pS338/pT339-NK1, pT344/pS347-NK1, and pT356/pT357-NK1. We found that substance P (SP) stimulated concentration-dependent phosphorylation of all three sites, which could be completely blocked in the presence of the NK1 receptor antagonist aprepitant. The other two endogenous ligands of the tachykinin family, neurokinin A (NKA) and neurokinin B (NKB), were also able to induce NK1 receptor phosphorylation, but to a much lesser extent than substance P. Interestingly, substance P promoted phosphorylation of the two distal sites more efficiently than that of the proximal site. The proximal site was identified as a substrate for phosphorylation by protein kinase C. Analysis of GPCR kinase (GRK)-knockout cells revealed that phosphorylation was mediated by all four GRK isoforms to similar extents at the T344/S347 and the T356/T357 cluster. Knockout of all GRKs resulted in abolition of all phosphorylation signals highlighting the importance of these kinases in agonist-mediated receptor phosphorylation. Thus, the 7TM phosphorylation assay technology allows for rapid and detailed analyses of GPCR phosphorylation.


Subject(s)
Receptors, Neurokinin-1 , Substance P , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-1/agonists , Phosphorylation/drug effects , Humans , Substance P/pharmacology , Animals , Immunoassay/methods , Cricetulus , CHO Cells , Mice , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin A/pharmacology , Neurokinin A/metabolism
2.
Elife ; 122023 Dec 12.
Article in English | MEDLINE | ID: mdl-38085667

ABSTRACT

Free fatty acid receptor 2 (FFAR2) is activated by short-chain fatty acids and expressed widely, including in white adipocytes and various immune and enteroendocrine cells. Using both wild-type human FFAR2 and a designer receptor exclusively activated by designer drug (DREADD) variant we explored the activation and phosphorylation profile of the receptor, both in heterologous cell lines and in tissues from transgenic knock-in mouse lines expressing either human FFAR2 or the FFAR2-DREADD. FFAR2 phospho-site-specific antisera targeting either pSer296/pSer297 or pThr306/pThr310 provided sensitive biomarkers of both constitutive and agonist-mediated phosphorylation as well as an effective means to visualise agonist-activated receptors in situ. In white adipose tissue, phosphorylation of residues Ser296/Ser297 was enhanced upon agonist activation whilst Thr306/Thr310 did not become phosphorylated. By contrast, in immune cells from Peyer's patches Thr306/Thr310 become phosphorylated in a strictly agonist-dependent fashion whilst in enteroendocrine cells of the colon both Ser296/Ser297 and Thr306/Thr310 were poorly phosphorylated. The concept of phosphorylation bar-coding has centred to date on the potential for different agonists to promote distinct receptor phosphorylation patterns. Here, we demonstrate that this occurs for the same agonist-receptor pairing in different patho-physiologically relevant target tissues. This may underpin why a single G protein-coupled receptor can generate different functional outcomes in a tissue-specific manner.


Subject(s)
Fatty Acids, Nonesterified , Receptors, G-Protein-Coupled , Animals , Humans , Mice , Cell Line , Fatty Acids, Volatile/metabolism , Mice, Transgenic , Phosphorylation , Receptors, G-Protein-Coupled/metabolism
3.
bioRxiv ; 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37214942

ABSTRACT

During Hedgehog (Hh) signal transduction in development and disease, the atypical G protein-coupled receptor (GPCR) SMOOTHENED (SMO) communicates with GLI transcription factors by binding the protein kinase A catalytic subunit (PKA-C) and physically blocking its enzymatic activity. Here we show that GPCR kinase 2 (GRK2) orchestrates this process during endogenous Hh pathway activation in the primary cilium. Upon SMO activation, GRK2 rapidly relocalizes from the ciliary base to the shaft, triggering SMO phosphorylation and PKA-C interaction. Reconstitution studies reveal that GRK2 phosphorylation enables active SMO to bind PKA-C directly. Lastly, the SMO-GRK2-PKA pathway underlies Hh signal transduction in a range of cellular and in vivo models. Thus, GRK2 phosphorylation of ciliary SMO, and the ensuing PKA-C binding and inactivation, are critical initiating events for the intracellular steps in Hh signaling. More broadly, our study suggests an expanded role for GRKs in enabling direct GPCR interactions with diverse intracellular effectors.

4.
Commun Biol ; 6(1): 419, 2023 04 15.
Article in English | MEDLINE | ID: mdl-37061609

ABSTRACT

G protein-coupled receptors (GPCRs) are important signal transducers that are phosphorylated upon activation at intracellular serine and threonine residues. Although antibodies that specifically recognize the phosphorylation state of GPCRs have been available for many years, efficient immunolocalization of phosphorylated receptors in their tissues of origin has not been possible. Here, we show that phosphorylation of receptors is highly unstable during routine immunohistochemical procedures, requiring the use of appropriate phosphatase inhibitors particular during tissue perfusion, post-fixation, and cryoprotection but not during immunostaining of tissue sections. We provide proof of concept using phosphorylation state-specific µ-opioid receptor (MOP) and cannabinoid receptor 1 (CB1) antibodies. Indeed, three of four well-characterized phosphosite-specific MOP antibodies, including pS375-MOP, pT376-MOP, and pT379-MOP, showed robust neuronal immunostaining in brain and spinal cord sections of opioid-treated mice only after inclusion of phosphatase inhibitors. We then extended this approach to the CB1 receptor and demonstrated that one of three newly-generated phosphosite-specific CB1 antibodies, namely pS425-CB1, showed striking staining of fibers and varicosities in brain slices from cannabinoid-treated mice. Although subsequent experiments showed that phospho-CB1 immunostaining was less sensitive to phosphatases, we conclude that the use of phosphatase inhibitors should always be considered in the development of immunohistochemical procedures for new phosphosite-specific GPCR antibodies. In summary, we anticipate that this improved protocol will facilitate the widespread use of phosphorylation state-specific antibodies to monitor the activation of endogenous GPCRs under physiological and pharmacological conditions. Our approach may also prove useful to confirm target engagement of GPCR drug candidates in native tissues.


Subject(s)
Analgesics, Opioid , Cannabinoids , Animals , Mice , Analgesics, Opioid/pharmacology , Phosphorylation , Receptors, G-Protein-Coupled , Neurons , Antibodies/pharmacology
5.
Commun Biol ; 5(1): 1206, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36352263

ABSTRACT

Analysis of agonist-driven phosphorylation of G protein-coupled receptors (GPCRs) can provide valuable insights into the receptor activation state and ligand pharmacology. However, to date, assessment of GPCR phosphorylation using high-throughput applications has been challenging. We have developed and validated a bead-based immunoassay for the quantitative assessment of agonist-induced GPCR phosphorylation that can be performed entirely in multiwell cell culture plates. The assay involves immunoprecipitation of affinity-tagged receptors using magnetic beads followed by protein detection using phosphorylation state-specific and phosphorylation state-independent anti-GPCR antibodies. As proof of concept, five prototypical GPCRs (MOP, C5a1, D1, SST2, CB2) were treated with different agonizts and antagonists, and concentration-response curves were generated. We then extended our approach to establish selective cellular GPCR kinase (GRK) inhibitor assays, which led to the rapid identification of a selective GRK5/6 inhibitor (LDC8988) and a highly potent pan-GRK inhibitor (LDC9728). In conclusion, this versatile GPCR phosphorylation assay can be used extensively for ligand profiling and inhibitor screening.


Subject(s)
Receptors, G-Protein-Coupled , Phosphorylation , Ligands , Receptors, G-Protein-Coupled/metabolism , Immunoassay
6.
J Biol Chem ; 298(5): 101932, 2022 05.
Article in English | MEDLINE | ID: mdl-35427647

ABSTRACT

GPR84 is an immune cell-expressed, proinflammatory receptor currently being assessed as a therapeutic target in conditions including fibrosis and inflammatory bowel disease. Although it was previously shown that the orthosteric GPR84 activators 2-HTP and 6-OAU promoted its interactions with arrestin-3, a G protein-biased agonist DL-175 did not. Here, we show that replacement of all 21 serine and threonine residues within i-loop 3 of GPR84, but not the two serines in the C-terminal tail, eliminated the incorporation of [32P] and greatly reduced receptor-arrestin-3 interactions promoted by 2-HTP. GPR84 was phosphorylated constitutively on residues Ser221 and Ser224, while various other amino acids are phosphorylated in response to 2-HTP. Consistent with this, an antiserum able to identify pSer221/pSer224 recognized GPR84 from cells treated with and without activators, whereas an antiserum able to identify pThr263/pThr264 only recognized GPR84 after exposure to 2-HTP and not DL-175. Two distinct GPR84 antagonists as well as inhibition of G protein-coupled receptor kinase 2/3 prevented phosphorylation of pThr263/pThr264, but neither strategy affected constitutive phosphorylation of Ser221/Ser224. Furthermore, mutation of residues Thr263 and Thr264 to alanine generated a variant of GPR84 also limited in 2-HTP-induced interactions with arrestin-2 and -3. By contrast, this mutant was unaffected in its capacity to reduce cAMP levels. Taken together, these results define a key pair of threonine residues, regulated only by subsets of GPR84 small molecule activators and by GRK2/3 that define effective interactions with arrestins and provide novel tools to monitor the phosphorylation and functional status of GPR84.


Subject(s)
Arrestins , Threonine , Arrestins/metabolism , Humans , Ligands , Mutation , Phosphorylation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Serine/metabolism , Threonine/metabolism , beta-Arrestin 2/metabolism
7.
Mol Endocrinol ; 30(6): 645-59, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27101376

ABSTRACT

The human somatostatin receptor 3 (sst3) is expressed in about 50% of all neuroendocrine tumors and hence a promising target for multireceptor somatostatin analogs. The sst3 receptor is unique among ssts in that it exhibits a very long intracellular C-terminal tail containing a huge number of potential phosphate acceptor sites. Consequently, our knowledge about the functional role of the C-terminal tail in sst3 receptor regulation is very limited. Here, we have generated a series of phosphorylation-deficient mutants that enabled us to determine crucial sites for its agonist-induced ß-arrestin mobilization, internalization, and down-regulation. Based on this information, we generated phosphosite-specific antibodies for C-terminal Ser(337)/Thr(341), Thr(348), and Ser(361) that enabled us to investigate the temporal patterns of sst3 phosphorylation and dephosphorylation. We found that the endogenous ligand somatostatin induced a rapid and robust phosphorylation that was completely blocked by the sst3 antagonist NVP-ACQ090. The stable somatostatin analogs pasireotide and octreotide promoted clearly less phosphorylation compared with somatostatin. We also show that sst3 phosphorylation occurred within seconds to minutes, whereas dephosphorylation of the sst3 receptor occurred at a considerable slower rate. In addition, we also identified G protein-coupled receptor kinases 2 and 3 and protein phosphatase 1α and 1ß as key regulators of sst3 phosphorylation and dephosphorylation, respectively. Thus, we here define the C-terminal phosphorylation motif of the human sst3 receptor that regulates its agonist-promoted phosphorylation, ß-arrestin recruitment, and internalization of this clinically relevant receptor.


Subject(s)
Receptors, Somatostatin/metabolism , Amino Acid Sequence , Biocatalysis/drug effects , Down-Regulation/drug effects , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , HEK293 Cells , Humans , Marine Toxins , Membrane Potentials/drug effects , Mutant Proteins/metabolism , Oxazoles/pharmacology , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Transport/drug effects , Receptors, Somatostatin/agonists , Receptors, Somatostatin/chemistry
8.
PLoS One ; 9(3): e91526, 2014.
Article in English | MEDLINE | ID: mdl-24637622

ABSTRACT

We have recently identified protein phosphatase 1ß (PP1ß) as G protein-coupled receptor (GPCR) phosphatase for the sst2 somatostatin receptor using siRNA knockdown screening. By contrast, for the sst5 somatostatin receptor we identified protein phosphatase 1γ (PP1γ) as GPCR phosphatase using the same approach. We have also shown that sst2 and sst5 receptors differ substantially in the temporal dynamics of their dephosphorylation and trafficking patterns. Whereas dephosphorylation and recycling of the sst2 receptor requires extended time periods of ∼30 min, dephosphorylation and recycling of the sst5 receptor is completed in less than 10 min. Here, we examined which receptor domains determine the selection of phosphatases for receptor dephosphorylation. We found that generation of tail-swap mutants between sst2 and sst5 was required and sufficient to reverse the patterns of dephosphorylation and trafficking of these two receptors. In fact, siRNA knockdown confirmed that the sst5 receptor carrying the sst2 tail is predominantly dephosphorylated by PP1ß, whereas the sst2 receptor carrying the sst5 tail is predominantly dephosphorylated by PP1γ. Thus, the GPCR phosphatase responsible for dephosphorylation of individual somatostatin receptor subtypes is primarily determined by their different carboxyl-terminal receptor domains. This phosphatase specificity has in turn profound consequences for the dephosphorylation dynamics and trafficking patterns of GPCRs.


Subject(s)
Protein Interaction Domains and Motifs/physiology , Protein Phosphatase 1/metabolism , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/metabolism , Arrestins , Cell Line , Humans , Isoenzymes , Phosphorylation , Protein Transport , Substrate Specificity , beta-Arrestins
9.
Mol Cell Endocrinol ; 387(1-2): 44-51, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24565897

ABSTRACT

The somatostatin receptor 2 (sst2) is the pharmacological target of somatostatin analogs that are widely used in the diagnosis and treatment of human neuroendocrine tumors. We have recently shown that the stable somatostatin analogs octreotide and pasireotide (SOM230) stimulate distinct patterns of sst2 receptor phosphorylation and internalization. Like somatostatin, octreotide promotes the phosphorylation of at least six carboxyl-terminal serine and threonine residues namely S341, S343, T353, T354, T356 and T359, which in turn leads to a robust receptor endocytosis. Unlike somatostatin, pasireotide stimulates a selective phosphorylation of S341 and S343 of the human sst2 receptor followed by a partial receptor internalization. Here, we show that exchange of S341 and S343 by alanine is sufficient to block pasireotide-driven internalization, whereas mutation of T353, T354, T356 and T359 to alanine is required to strongly inhibited both octreotide- and somatostatin-induced internalization. Yet, combined mutation of T353, T354, T356 and T359 is not sufficient to prevent somatostatin-driven ß-arrestin mobilization and receptor desensitization. Replacement of all fourteen carboxyl-terminal serine and threonine residues by alanine completely abrogates sst2 receptor internalization and ß-arrestin mobilization in HEK293 cells. Together, our findings demonstrate for the first time that agonist-selective sst2 receptor internalization is regulated by multi-site phosphorylation of its carboxyl-terminal tail.


Subject(s)
Endocytosis/drug effects , Octreotide/pharmacology , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Amino Acid Sequence , Antineoplastic Agents, Hormonal/pharmacology , Arrestins/metabolism , Cell Line , HEK293 Cells , Humans , Molecular Sequence Data , Neuroendocrine Tumors/drug therapy , Phosphorylation , Protein Transport/drug effects , Somatostatin/pharmacology , beta-Arrestins
10.
Br J Pharmacol ; 171(7): 1591-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24328848

ABSTRACT

The biological actions of somatostatin are mediated by a family of five GPCRs, named sst1 to sst5 . Somatostatin receptors exhibit equally high-binding affinities to their natural ligand somatostatin-14 and largely overlapping distributions. The overexpression of somatostatin receptors in human tumours is the molecular basis for diagnostic and therapeutic application of the stable somatostatin analogues octreotide, lanreotide and pasireotide. The efficiency of somatostatin receptor signalling is tightly regulated and ultimately limited by the coordinated phosphorylation and dephosphorylation of intracellular carboxyl-terminal serine and threonine residues. Here, we review and discuss recent progress in the generation and application of phosphosite-specific antibodies for human sst2 and sst5 receptors. These phosphosite-specific antibodies are unique tools to monitor the spatial and temporal dynamics of receptors phosphorylation and dephosphorylation. Using a combined approach of phosphosite-specific antibodies and siRNA knock-down screening, relevant kinases and phosphatases were identified. Emerging evidence suggests distinct mechanisms of agonist-selective fine-tuning for individual somatostatin receptors. The recently uncovered differences in phosphorylation and dephosphorylation of these receptors may hence be of physiological significance in mediating responses to acute, persistent or repeated stimuli in a variety of target tissues.


Subject(s)
Receptors, Somatostatin/agonists , Signal Transduction/drug effects , Somatostatin/pharmacology , Amino Acid Sequence , Animals , Antibodies/pharmacology , Humans , Ligands , Molecular Sequence Data , Phosphorylation , RNA Interference , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/metabolism
11.
PLoS One ; 8(5): e64975, 2013.
Article in English | MEDLINE | ID: mdl-23734232

ABSTRACT

The chemokine receptor CXCR4 regulates cell migration during ontogenesis and disease states including cancer and inflammation. Upon stimulation by the endogenous ligand CXCL12, CXCR4 becomes phosphorylated at multiple sites in its C-terminal domain. Mutations in the CXCR4 gene affecting C-terminal phosphorylation sites are a hallmark of WHIM syndrome, a genetic disorder characterized by a gain-of-CXCR4-function. To better understand how multi-site phosphorylation of CXCR4 is organized and how perturbed phosphorylation might affect CXCR4 function, we developed novel phosphosite-specific CXCR4 antibodies and studied the differential regulation and interaction of three C-terminal phosphorylation sites in human embryonic kidney cells (HEK293). CXCL12 promoted a robust phosphorylation at S346/347 which preceded phosphorylation at S324/325 and S338/339. After CXCL12 washout, the phosphosites S338/339 and S324/325 were rapidly dephosphorylated whereas phosphorylation at S346/347 was long-lasting. CXCL12-induced phosphorylation at S346/347 was staurosporine-insensitive and mediated by GRK2/3. WHIM syndrome-associated CXCR4 truncation mutants lacking the S346/347 phosphosite and the recently identified E343K WHIM mutant displayed strongly impaired phosphorylation at S324/325 and S338/339 as well as reduced CXCL12-induced receptor internalization. Relevance of the S346-S348 site was confirmed by a S346-348A mutant showing strongly impaired CXCL12-promoted phosphorylation at S324/325 and S338/339, defective internalization, gain of calcium mobilization, and reduced desensitization. Thus, the triple serine motif S346-S348 contains a major initial CXCR4 phosphorylation site and is required for efficient subsequent multi-site phosphorylation and receptor regulation. Hierarchical organization of CXCR4 phosphorylation explains why small deletions at the extreme CXCR4 C terminus typically associated with WHIM syndrome severely alter CXCR4 function.


Subject(s)
Chemokine CXCL12/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Receptors, CXCR4/metabolism , Binding Sites/genetics , Calcium/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 3/genetics , HEK293 Cells , Humans , Immunoblotting , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Kinetics , Mutation , Phosphorylation , Primary Immunodeficiency Diseases , RNA Interference , Receptors, CXCR4/genetics , Serine/genetics , Serine/metabolism , Warts/genetics , Warts/metabolism
12.
Regul Pept ; 183: 1-6, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23466804

ABSTRACT

BACKGROUND: The somatostatin receptor 1 (sst1) is widely distributed throughout the body and is also present in neoplastic tissues. However, little is known about its precise tissue distribution, regulation and function, which may in part be due to the lack of specific monoclonal anti-sst1 antibodies. METHODS: We have characterized the novel rabbit monoclonal anti-human sst1 antibody UMB-7 using sst1-expressing cells and human pituitary samples. The antibody was then used for immunohistochemical staining of a large panel of formalin-fixed, paraffin-embedded human tissues. RESULTS: Western blot analyses of BON-1 cells and human pituitary revealed a broad band migrating at a molecular weight of 45,000-60,000. After enzymatic deglycosylation the size of this band decreased to a molecular weight of 45,000. UMB-7 yielded an efficient immunostaining of distinct cell populations in the human tissue samples with a predominance of plasma membrane staining, which was completely abolished by preadsorption of UMB-7 with its immunizing peptide. The sst1 receptor was detected in anterior pituitary, pancreatic islets, distal tubules, enteric ganglion cells and nerve fibers, chief cells of the gastric mucosa, macrophages and mast cells. In addition, sst1 was observed in pituitary adenomas, gastrointestinal neuroendocrine tumors and pheochromocytoma as well as in pancreatic adenocarcinomas, gastric carcinomas, urinary bladder carcinomas and sarcomas. CONCLUSIONS: UMB-7 may prove of great value in the identification of sst1-expressing tumors during routine histopathological examinations. This may open up new routes for diagnostic and therapeutic intervention.


Subject(s)
Antibodies, Monoclonal/immunology , Neoplasms/metabolism , Receptors, Somatostatin/biosynthesis , Animals , Humans , Immunohistochemistry , Neoplasms/immunology , Neoplasms/pathology , Paraffin Embedding , Rabbits , Receptors, Somatostatin/analysis , Receptors, Somatostatin/immunology
13.
Mol Endocrinol ; 27(4): 671-82, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23418396

ABSTRACT

The frequent overexpression of the somatostatin receptors sst2 and sst5 in neuroendocrine tumors provides the molecular basis for therapeutic application of novel multireceptor somatostatin analogs. Although the phosphorylation of the carboxyl-terminal region of the sst2 receptor has been studied in detail, little is known about the agonist-induced regulation of the human sst5 receptor. Here, we have generated phosphosite-specific antibodies for the carboxyl-terminal threonines 333 (T333) and 347 (T347), which enabled us to selectively detect either the T333-phosphorylated or the T347-phosphorylated form of sst5. We show that agonist-mediated phosphorylation occurs at T333, whereas T347 is constitutively phosphorylated in the absence of agonist. We further demonstrate that the multireceptor somatostatin analog pasireotide and the sst5-selective ligand L-817,818 but not octreotide or KE108 were able to promote a detectable T333 phosphorylation. Interestingly, BIM-23268 was the only sst5 agonist that was able to stimulate T333 phosphorylation to the same extent as natural somatostatin. Agonist-induced T333 phosphorylation was dose-dependent and selectively mediated by G protein-coupled receptor kinase 2. Similar to that observed for the sst2 receptor, phosphorylation of sst5 occurred within seconds. However, unlike that seen for the sst2 receptor, dephosphorylation and recycling of sst5 were rapidly completed within minutes. We also identify protein phosphatase 1γ as G protein-coupled receptor phosphatase for the sst5 receptor. Together, we provide direct evidence for agonist-selective phosphorylation of carboxyl-terminal T333. In addition, we identify G protein-coupled receptor kinase 2-mediated phosphorylation and protein phosphatase 1γ-mediated dephosphorylation of T333 as key regulators of rapid internalization and recycling of the human sst5 receptor.


Subject(s)
Phosphothreonine/metabolism , Receptors, Somatostatin/metabolism , Amino Acid Sequence , Antibody Specificity/drug effects , Antibody Specificity/immunology , Biocatalysis/drug effects , Endocytosis/drug effects , G-Protein-Coupled Receptor Kinase 2/metabolism , HEK293 Cells , Humans , Marine Toxins , Molecular Sequence Data , Oxazoles/pharmacology , Phosphorylation/drug effects , Protein Phosphatase 1/metabolism , Protein Transport/drug effects , Receptors, Somatostatin/agonists , Receptors, Somatostatin/chemistry
14.
Neuroendocrinology ; 96(4): 301-10, 2012.
Article in English | MEDLINE | ID: mdl-22414742

ABSTRACT

BACKGROUND: Among the five somatostatin receptors (sst(1)-sst(5)), the sst(3) receptor displays a distinct pharmacological profile. Like sst(2), the sst(3) receptor efficiently internalizes radiolabeled somatostatin analogs. Unlike sst(2), however, internalized sst(3) receptors are rapidly transferred to lysosomes for degradation. Apart from this, very little is known about the clinical relevance of the sst(3) receptor, which may in part be due to the lack of specific monoclonal sst(3) antibodies. METHODS: Here, we have extensively characterized the novel rabbit monoclonal anti-human sst(3) antibody UMB-5 using transfected cells and receptor-expressing tissues. UMB-5 was then subjected to immunohistochemical staining of a series of 190 formalin-fixed, paraffin-embedded normal and neoplastic human tissues. RESULTS: Specificity of UMB-5 was demonstrated by detection of a broad band migrating at a molecular weight of 70,000-85,000 in immunoblots from human pituitary. After enzymatic deglycosylation, the size of this band decreased to a molecular weight of 45,000. Tissue immunostaining was completely abolished by pre-adsorption of UMB-5 with its immunizing peptide. In addition, UMB-5 detected distinct cell populations in human tissues like pancreatic islands, anterior pituitary, adrenal cortex, adrenal medulla, and enteric ganglia, similar to that seen with a rabbit polyclonal antibody generated against a different carboxyl-terminal epitope of the sst(3) receptor. In a comparative immunohistochemical study, UMB-5 yielded predominant plasma membrane staining in the majority of pituitary adenomas, pheochromocytomas, and a subset of neuroendocrine tumors. The sst(3) receptor was also present in many glioblastomas, pancreatic, breast, cervix, and ovarian carcinomas. CONCLUSION: The rabbit monoclonal antibody UMB-5 may prove of great value in the identification of sst(3)-expressing tumors during routine histopathological examinations. Given its unique trafficking properties, these tumors may be potential candidates for sst(3)-directed receptor radiotherapy.


Subject(s)
Antibodies, Monoclonal , Gene Expression Regulation, Neoplastic , Receptors, Somatostatin/biosynthesis , Receptors, Somatostatin/genetics , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Molecular Sequence Data , Pituitary Gland/metabolism , Rabbits
15.
Neuroendocrinology ; 94(3): 255-64, 2011.
Article in English | MEDLINE | ID: mdl-21952553

ABSTRACT

OBJECTIVE: The frequent overexpression of somatostatin receptors (sst) in neuroendocrine tumors provides the molecular basis for the diagnostic and therapeutic application of stable somatostatin analogs. Whereas octreotide acts mainly via the sst(2) receptor, the novel pan-somatostatin analog pasireotide exhibits particular high affinity for the sst(5) receptor. To determine whether a patient is a candidate for octreotide or pasireotide therapy, it is important to evaluate the somatostatin receptor status. However, so far highly specific rabbit monoclonal antibodies have been developed for the sst(2) receptor only (clone UMB-1). METHODS: Here, we have extensively characterized a novel rabbit monoclonal antibody for the human sst(5) receptor (clone UMB-4). In a comparative immunohistochemical study, the expression of sst(5) and sst(2) receptors was assessed using UMB-4 and UMB-1, respectively. RESULTS: Western blot experiments unequivocally demonstrated that UMB-4 selectively detected its cognate sst(5) receptor and did not cross-react with other proteins present in crude tissue homogenates. UMB-4 yielded a highly effective immunostaining of distinct cell populations in formalin-fixed, paraffin-embedded human tissues with a predominance of plasma membrane staining. In the pituitary, sst(5) was present on all growth hormone (GH)- and adrenocorticotropin hormone (ACTH)-producing cells whereas sst(2) was only observed on a subpopulation of GH-positive cells. Consequently, sst(5) was detectable on the majority of GH and ACTH adenomas. In contrast, sst(2) was only seen on GH but not on ACTH adenomas. CONCLUSIONS: The rabbit monoclonal antibodies UMB-4 and UMB-1 will facilitate the assessment of the somatostatin receptor status of human tumors during routine histopathological examinations.


Subject(s)
Antibodies, Monoclonal/metabolism , Neoplasms/metabolism , Receptors, Somatostatin/immunology , Receptors, Somatostatin/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Antibody Specificity , Gene Expression Regulation, Neoplastic , HEK293 Cells , Health , Humans , Immunohistochemistry , Neoplasms/pathology , Rabbits , Receptors, Somatostatin/analysis , Tissue Distribution , Tumor Cells, Cultured
16.
Mol Endocrinol ; 25(5): 859-66, 2011 May.
Article in English | MEDLINE | ID: mdl-21330405

ABSTRACT

The clinically used somatostatin (SS-14) analogs octreotide and pasireotide (SOM230) stimulate distinct species-specific patterns of sst(2A) somatostatin receptor phosphorylation and internalization. Like SS-14, octreotide promotes the phosphorylation of at least six carboxyl-terminal serine and threonine residues, namely S341, S343, T353, T354, T356, and T359, which in turn leads to a robust endocytosis of both rat and human sst(2A) receptors. Unlike SS-14, pasireotide fails to induce any substantial phosphorylation or internalization of the rat sst(2A) receptor. Nevertheless, pasireotide is able to stimulate a selective phosphorylation of S341 and S343 of the human sst(2A) receptor followed by a clearly detectable receptor sequestration. Here, we show that transplantation of amino acids 1-180 of the human sst(2A) receptor to the rat sst(2A) receptor facilitates pasireotide-induced internalization. Conversely, construction of a rat-human sst(2A) chimera conferred resistance to pasireotide-induced internalization. We then created a series of site-directed mutants leading to the identification of amino acids 27, 30, 163, and 164 that when exchanged to their human counterparts facilitated pasireotide-driven S341/S343 phosphorylation and internalization of the rat sst(2A) receptor. Exchange of these amino acids to their rat counterparts completely blocked the pasireotide-mediated internalization of the human sst(2A) receptor. Notably, octreotide and SS-14 stimulated a full phosphorylation and internalization of all mutant sst(2A) receptors tested. Together, these findings suggest that pasireotide activates the sst(2A) receptor via a molecular switch that is structurally and functionally distinct from that turned on during octreotide-driven sst(2A) activation.


Subject(s)
Receptors, Somatostatin/metabolism , Signal Transduction , Amino Acid Sequence , Animals , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , HEK293 Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Octreotide/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Transport , Rats , Receptors, Somatostatin/agonists , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
17.
Regul Pept ; 167(1): 9-13, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-20851148

ABSTRACT

BACKGROUND: The immunohistochemical localization of the µ-opioid receptor (MOR, MOP) has been studied in detail in mouse and rat brain using a variety of polyclonal antibodies. However, biochemical analysis of the MOR signaling complex in vivo has been hampered by the lack of suitable monoclonal antibodies for efficient immunoprecipitation of the receptor protein from native sources. Moreover, previous immunohistochemical investigations were restricted to frozen sections from perfusion-fixed rodent brain, largely due to the limited availability of MOR antibodies that effectively stain paraffin-embedded tissues. METHODS: Here, we extensively characterized the novel rabbit monoclonal anti-MOR antibody UMB-3 using transfected cells and MOR-deficient mice. UMB-3 was also subjected to a comparative immunohistochemical study of formalin-fixed, paraffin-embedded mouse and rat organ samples as well as human normal and neoplastic tissues. RESULTS: Specificity of UMB-3 was demonstrated by detection of a broad band migrating at M(r) 70,000-80,000 in immunoprecipitates from crude brain homogenates of MOR+/+ mice but not of MOR⁻/⁻ mice; cell surface staining of MOR-transfected cells; translocation of MOR receptor immunostaining after agonist exposure; distinct immunostaining of neuronal cell bodies and fibers in MOR-expressing brain regions; absence of staining in MOR-deficient mice; and abolition of tissue immunostaining by preadsorption of UMB-3 with its immunizing peptide. CONCLUSIONS: The rabbit monoclonal antibody UMB-3 is an excellent tool for immunoprecipitation of MOR from native sources as well as for immunohistochemical staining of MOR in paraffin-embedded tissue samples of rodent and human origin.


Subject(s)
Antibodies, Monoclonal/metabolism , Brain/metabolism , Neoplasms/metabolism , Receptors, Opioid, mu/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibody Affinity , Brain/cytology , Brain/immunology , Cells, Cultured , Female , Fixatives/chemistry , Formaldehyde/chemistry , Gene Expression , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Male , Mice , Mice, Transgenic , Neoplasms/immunology , Neoplasms/pathology , Paraffin Embedding , Rabbits , Rats , Rats, Wistar , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/immunology , Tissue Fixation , Transfection
18.
J Clin Endocrinol Metab ; 94(2): 654-61, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19001514

ABSTRACT

OBJECTIVE: The clinically used somatostatin analogs, octreotide and lanreotide, act primarily by binding to somatostatin receptor 2 (sst2). In contrast, the novel multireceptor ligand pasireotide (SOM230) binds with high affinity to somatostatin receptor subtypes sst1, sst2, sst3, and sst5. SOM230 is currently under clinical evaluation for treatment of acromegaly, Cushing's disease, and octreotide-resistant carcinoid tumors. However, the effects of SOM230 on internalization and postendosomal sorting of individual human somatostatin receptor subtypes have not been determined so far. RESULTS: Here we show that SOM230 was less potent than octreotide in inducing internalization and signaling of sst2 receptors expressed in human embryonic kidney cells. In contrast, SOM230 was more potent than octreotide in inducing internalization and signaling of sst3 and sst5 receptors. Both SOM230 and octreotide stimulated a rapid down-regulation of sst3 but not of sst2 or sst5 receptors. SOM230 and octreotide profoundly differed in their patterns of sst2-stimulated beta-arrestin mobilization. Whereas octreotide-mediated receptor activation led to the formation of stable complexes facilitating the internalization of sst2 and beta-arrestin-2 into the same endocytic vesicles, SOM230-mediated receptor activation led to the formation of unstable complexes that dissociated at or near the plasma membrane. Consequently, sst2 receptors recycled rapidly to the plasma membrane after endocytosis in SOM230-treated cells, but not in octreotide-treated cells. CONCLUSION: We show that SOM230 modulates somatostatin receptor trafficking in a manner clearly distinct from octreotide and somatostatin. These findings may provide an explanation for the differential regulation of somatostatin receptor responsiveness during long-term administration of stable somatostatin analogs.


Subject(s)
Octreotide/pharmacology , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Antineoplastic Agents, Hormonal/pharmacology , Cells, Cultured , Endocytosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Somatostatin/pharmacology
19.
PLoS One ; 3(12): e4069, 2008.
Article in English | MEDLINE | ID: mdl-19116653

ABSTRACT

BACKGROUND: The CXCR4 chemokine receptor regulates migration and homing of cancer cells to specific metastatic sites. Determination of the CXCR4 receptor status will provide predictive information for disease prognosis and possible therapeutic intervention. However, previous attempts to localize CXCR4 using poorly characterized mouse monoclonal or rabbit polyclonal antibodies have produced predominant nuclear and occasional cytoplasmic staining but did not result in the identification of bona fide cell surface receptors. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we extensively characterized the novel rabbit monoclonal anti-CXCR4 antibody (clone UMB-2) using transfected cells and tissues from CXCR4-deficient mice. Specificity of UMB-2 was demonstrated by cell surface staining of CXCR4-transfected cells; translocation of CXCR4 immunostaining after agonist exposure; detection of a broad band migrating at M(r) 38,000-43,000 in Western blots of homogenates from CXCR4-expressing cells; selective detection of the receptor in tissues from CXCR4+/+ but not from CXCR4-/- mice; and abolition of tissue immunostaining by preadsorption of UMB-2 with its immunizing peptide. In formalin-fixed, paraffin-embedded human tumor tissues, UMB-2 yielded highly effective plasma membrane staining of a subpopulation of tumor cells, which were often heterogeneously distributed throughout the tumor. A comparative analysis of the mouse monoclonal antibody 12G5 and other frequently used commercially available antibodies revealed that none of these was able to detect CXCR4 under otherwise identical conditions. CONCLUSIONS/SIGNIFICANCE: Thus, the rabbit monoclonal antibody UMB-2 may prove of great value in the assessment of the CXCR4 receptor status in a variety of human tumors during routine histopathological examination.


Subject(s)
Antibodies, Monoclonal/immunology , Neoplasms/diagnosis , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Immunohistochemistry , Mice , Mice, Inbred Strains , Neoplasms/immunology , Neoplasms/pathology , Rabbits , Receptors, CXCR4/analysis , Transfection
20.
Biochim Biophys Acta ; 1763(11): 1229-37, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17055077

ABSTRACT

The calcium sensor protein caldendrin is abundantly expressed in neurons and is thought to play an important role in different aspects of synapto-dendritic Ca2+ signaling. Caldendrin is highly abundant in the postsynaptic density of a subset of excitatory synapses in brain and its distinct localization raises several decisive questions about its function. Previous work suggests that caldendrin is tightly associated with Ca2+ - and Ca2+ release channels and might be involved in different aspects of the organization of the postsynaptic scaffold as well as with synapse-to-nucleus communication. In this report we introduce two new EF-hand calcium sensor proteins termed calneurons that apart from calmodulin represent the closest homologues of caldendrin in brain. Calneurons have a different EF-hand organization than other calcium sensor proteins, are prominently expressed in neurons and will presumably bind Ca2+ with higher affinity than caldendrin. Despite some significant structural differences it is conceivable that they are involved in similar Ca2+ regulated processes like caldendrin and neuronal calcium sensor proteins.


Subject(s)
Calcium Signaling , Calcium-Binding Proteins/metabolism , Neurons/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Molecular Sequence Data , Protein Conformation , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...