1
|
Kunte SC, Unterrainer LM, Kunz WG, Winkelmann M, Lindner S, Jurkschat K, Wängler C, Wängler B, Schirrmacher R, Bartenstein P, Belka C, Schichor C, Albert NL, Unterrainer M. Lenticulostriatal Ischemia Shows Relevant SSTR Expression on PET/CT Imaging Using the Novel SSTR-Targeting Peptide 18 F-SiTATE. Clin Nucl Med 2024; 49:e689-e690. [PMID: 39354685 DOI: 10.1097/rlu.0000000000005482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
ABSTRACT A 64-year-old woman with meningioma presented with left-sided lenticulostriatal ischemia following craniotomy and debulking of a sphenoid wing meningioma. For subsequent radiotherapy planning, an SSTR-targeted PET/CT using the novel ligand 18 F-SiTATE was performed 2.5 months thereafter. The meningioma remnants showed transosseous, intrasellar, and perivascular extension around the internal carotid artery with strong SSTR expression. Moreover, there was focal 18 F-SiTATE uptake in the left caudate and corresponding contrast enhancement due to postischemic blood-brain barrier disruption and reactive SSTR expression. Therefore, increased cortical or subcortical SSTR PET signal may be related to ischemic changes even in the subacute stage after initial stroke.
Collapse
Affiliation(s)
- Sophie C Kunte
- From the Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | | | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Michael Winkelmann
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Simon Lindner
- From the Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Klaus Jurkschat
- Fakultät für Chemie und Chemische Biologie, Technische Universität Dortmund, Dortmund, Germany
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Ralf Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Peter Bartenstein
- From the Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Christian Schichor
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Nathalie L Albert
- From the Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | | |
Collapse
|
2
|
Ruiz-Viroga V, Urbanavicius J, Torterolo P, Lagos P. In vivo uptake of a fluorescent conjugate of melanin-concentrating hormone in the rat brain. J Chem Neuroanat 2021; 114:101959. [PMID: 33848617 DOI: 10.1016/j.jchemneu.2021.101959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 04/03/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022]
Abstract
Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide synthesized by posterior hypothalamic and incerto-hypothalamic neurons that project throughout the central nervous system. The MCHergic system modulates several important functions such as feeding behavior, mood and sleep. MCH exerts its biological functions through interaction with the MCHR-1 receptor, the only functional receptor present in rodents. The internalization process of MCHR-1 triggered by MCH binding was described in vitro in non-neuronal heterologous systems with over-expression of MCHR-1. Reports of in vivo MCHR-1 internalization dynamics are scarce, however, this is an important process to explore based on the critical functions of the MCHergic system. We had previously determined that 60 min after intracerebroventricular (i.c.v.) microinjections of MCH conjugated with fluorophore rhodamine (R-MCH), the dorsal and median raphe nucleus presented R-MCH positive labeled neurons. In the present work, we further studied the in vivo uptake process focusing on the distribution and time-dependent pattern of R-MCH positive cells 10, 20 and 60 min (T10, T20 and T60, respectively) after i.c.v. microinjection of R-MCH. We also explored this uptake process to see whether it was receptor- and clathrin-dependent and examined the phenotype of R-MCH positive cells and their proximity to MCHergic fibers. We found a great number of R-MCH positive cells with high fluorescence intensity in the lateral septum, nucleus accumbens and hippocampus at T20 and T60 (but not at T10), while a lower number with low intensity was observed in the dorsal raphe nucleus. At T20, in rats pre-treated with a MCHR-1 antagonist (ATC-0175) or with phenylarsine oxide (PAO), a clathrin endocytosis inhibitor, a robust decrease (> 50 %) of R-MCH uptake occurred in these structures. The R-MCH positive cells were identified as neurons (NeuN positive, GFAP negative) and some MCHergic fibers run in the vicinities of them. We concluded that neurons localized at structures that were close to the ventricular surfaces could uptake R-MCH in vivo through a receptor-dependent and clathrin-mediated process. Our results support volume transmission of MCH through the cerebrospinal fluid to reach distant targets. Finally, we propose that R-MCH would be an effective tool to study MCH-uptake in vivo.
Collapse
Affiliation(s)
- Vicente Ruiz-Viroga
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Jessika Urbanavicius
- Departamento de Neurofarmacología Experimental, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Pablo Torterolo
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Patricia Lagos
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
3
|
Riedemann T, Sutor B. Long-lasting actions of somatostatin on pyramidal cell excitability in the mouse cingulate cortex. Neurosci Lett 2019; 698:217-223. [PMID: 30668961 DOI: 10.1016/j.neulet.2019.01.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 02/07/2023]
Abstract
Many neurological diseases are related to disturbances of somatostatin- (SOM-) expressing interneurons in the cingulate cortex. Therefore, their role within the circuitry of the cingulate cortex needs to be investigated. We describe here the physiological time course of SOM effects onto pyramidal cell excitability and action potential discharge pattern. Furthermore, we show that the GRK2 inhibitor Gallein had no effect on the reduced SOM-induced response following repetitive SOM applications.
Collapse
Affiliation(s)
- Therese Riedemann
- Biomedical Center, Ludwig-Maximilians-Universität, Physiological Genomics, Großhaderner Str. 9, 82152 Planegg-Martinsried, Germany.
| | - Bernd Sutor
- Biomedical Center, Ludwig-Maximilians-Universität, Physiological Genomics, Großhaderner Str. 9, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
4
|
Adjuvant celecoxib and lanreotide following transarterial chemoembolisation for unresectable hepatocellular carcinoma: a randomized pilot study. Oncotarget 2018; 8:48303-48312. [PMID: 28430638 PMCID: PMC5564648 DOI: 10.18632/oncotarget.15684] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/15/2017] [Indexed: 02/05/2023] Open
Abstract
Recurrence of hepatocellular carcinoma (HCC) after transarterial chemoembolisation (TACE) is common due to neoangiogenesis. Cyclooxygenase-2 inhibitors and somatostatin analogues were reported to inhibit tumour angiogenesis. The pilot randomized controlled trial was aimed to prospectively evaluate the protocol of TACE combined with celecoxib and lanreotide (TACE+C+L) in patients with unresectable and advanced HCC. A total of 71 patients with HCC were enrolled and randomly assigned to either TACE (n=35) or TACE+C+L (n=36) group. Overall survival, disease control rate (DCR), and adverse events were assessed during a 3-year follow-up period. The median overall survival of the TACE+C+L group (15.0 months) was doubled compared to that of TACE group (7.5 months), p = 0.012. DCR of the TACE+C+L group was significantly higher than that of the TACE group either at 6 months (72.2% vs 42.9%, p = 0.012) or at 12 months (61.1% vs 28.6%, p = 0.006). The median overall survivals (13 months vs 4.5 months, p = 0.013) and DCR at 12 months (50% vs 13.6%, p = 0.008) of patients with advanced HCC in TACE+C+L groups were significantly higher than those in TACE group. No significant difference of adverse events was observed between the two groups. The occurrence of post-embolisation syndrome in TACE+C+L group was significantly lower than that in TACE group (16.7% vs 60.0%, p = 0.001). In conclusion, the regimen of TACE+C+L prolonged overall survival, enhanced tumour response, reduced post-embolisation syndrome and was well-tolerable in the patients with unresectable HCC. It may be more beneficial for advanced HCC.
Collapse
|
5
|
|
6
|
Kozhemyakin M, Rajasekaran K, Todorovic MS, Kowalski SL, Balint C, Kapur J. Somatostatin type-2 receptor activation inhibits glutamate release and prevents status epilepticus. Neurobiol Dis 2013; 54:94-104. [PMID: 23473742 DOI: 10.1016/j.nbd.2013.02.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 02/06/2013] [Accepted: 02/25/2013] [Indexed: 02/07/2023] Open
Abstract
Newer therapies are needed for the treatment of status epilepticus (SE) refractory to benzodiazepines. Enhanced glutamatergic neurotransmission leads to SE, and AMPA receptors are modified during SE. Reducing glutamate release during SE is a potential approach to terminate SE. The neuropeptide somatostatin (SST) is proposed to diminish presynaptic glutamate release by activating SST type-2 receptors (SST2R). SST exerts an anticonvulsant action in some experimental models of seizures. Here, we investigated the mechanism of action of SST on excitatory synaptic transmission at the Schaffer collateral-CA1 synapses and the ability of SST to treat SE in rats using patch-clamp electrophysiology and video-EEG monitoring of seizures. SST reduced action potential-dependent EPSCs (sEPSCs) at Schaffer collateral-CA1 synapses at concentrations up to 1μM; higher concentrations had no effect or increased the sEPSC frequency. SST also prevented paired-pulse facilitation of evoked EPSCs and did not alter action-potential-independent miniature EPSCs (mEPSCs). The effect of SST on EPSCs was inhibited by the SST2R antagonist cyanamid-154806 and was mimicked by the SST2R agonists, octreotide and lanreotide. Both SST and octreotide reduced the firing rate of CA1 pyramidal neurons. Intraventricular administration of SST, within a range of doses, either prevented or attenuated pilocarpine-induced SE or delayed the median time to the first grade 5 seizure by 11min. Similarly, octreotide or lanreotide prevented or attenuated SE in more than 65% of animals. Compared to the pilocarpine model, octreotide was highly potent in preventing or attenuating continuous hippocampal stimulation-induced SE in all animals within 60min of SE onset. Our results demonstrate that SST, through the activation of SST2Rs, diminishes presynaptic glutamate release and attenuates SE.
Collapse
Affiliation(s)
- Maxim Kozhemyakin
- Department of Neurology and Neuroscience, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | | | | | | | |
Collapse
|
7
|
González-Hernández A, Hernández-Ortega S, Gómez E, Fernández-G JM. The structure of a triosmium carbonyl cluster-phenylarsine oxide derivative. J Organomet Chem 2011. [DOI: 10.1016/j.jorganchem.2011.06.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
8
|
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | | |
Collapse
|
9
|
Lu R, Li Y, Zhang Y, Chen Y, Shields AD, Winder DG, Angelotti T, Jiao K, Limbird LE, Zhou Y, Wang Q. Epitope-tagged receptor knock-in mice reveal that differential desensitization of alpha2-adrenergic responses is because of ligand-selective internalization. J Biol Chem 2009; 284:13233-43. [PMID: 19276088 DOI: 10.1074/jbc.m807535200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although ligand-selective regulation of G protein-coupled receptor-mediated signaling and trafficking are well documented, little is known about whether ligand-selective effects occur on endogenous receptors or whether such effects modify the signaling response in physiologically relevant cells. Using a gene targeting approach, we generated a knock-in mouse line, in which N-terminal hemagglutinin epitope-tagged alpha(2A)-adrenergic receptor (AR) expression was driven by the endogenous mouse alpha(2A)AR gene locus. Exploiting this mouse line, we evaluated alpha(2A)AR trafficking and alpha(2A)AR-mediated inhibition of Ca(2+) currents in native sympathetic neurons in response to clonidine and guanfacine, two drugs used for treatment of hypertension, attention deficit and hyperactivity disorder, and enhancement of analgesia through actions on the alpha(2A)AR subtype. We discovered a more rapid desensitization of Ca(2+) current suppression by clonidine than guanfacine, which paralleled a more marked receptor phosphorylation and endocytosis of alpha(2A)AR evoked by clonidine than by guanfacine. Clonidine-induced alpha(2A)AR desensitization, but not receptor phosphorylation, was attenuated by blockade of endocytosis with concanavalin A, indicating a critical role for internalization of alpha(2A)AR in desensitization to this ligand. Our data on endogenous receptor-mediated signaling and trafficking in native cells reveal not only differential regulation of G protein-coupled receptor endocytosis by different ligands, but also a differential contribution of receptor endocytosis to signaling desensitization. Taken together, our data suggest that these HA-alpha(2A)AR knock-in mice will serve as an important model in developing ligands to favor endocytosis or nonendocytosis of receptors, depending on the target cell and pathophysiology being addressed.
Collapse
Affiliation(s)
- Roujian Lu
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Rubio A, Pérez M, de Lecea L, Avila J. Effect of cortistatin on tau phosphorylation at Ser262 site. J Neurosci Res 2008; 86:2462-75. [PMID: 18438934 DOI: 10.1002/jnr.21689] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The development of intraneuronal lesions as a result of the progressive deposition of hyperphosphorylated tau at specific brain regions (such as hippocampus and cortex) plays a key role in the pathological process of Alzheimer's disease. However, the mechanisms by which tau phosphorylation is regulated, mainly in the pathology found in the cortex, are still poorly understood. Here, we analyzed the effect of cortistatin, a cortical neuropeptide related to somatostatin, on tau phosphorylation at Ser262 in cultures of murine cortical neurons. Both somatostatin and cortistatin induce tau phosphorylation at Ser262, a site modified in Alzheimer's disease, although with different kinetics in cortex. The effect of cortistatin likely is mediated by heterodimeric receptors composed of somatostatin receptor subtypes 2 and 4 and also by protein kinase C signaling. Cortistatin-deficient mice show decreased tau phosphorylation at Ser262 in the cortex but not in other brain regions tested. Our results suggest an important role for cortistatin in the regulation of tau phosphorylation that may be associated with the pathophysiology of Alzheimer's disease in regions such as the cerebral cortex.
Collapse
Affiliation(s)
- Alicia Rubio
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | |
Collapse
|
11
|
Jacobs S, Schulz S. Intracellular trafficking of somatostatin receptors. Mol Cell Endocrinol 2008; 286:58-62. [PMID: 18045773 DOI: 10.1016/j.mce.2007.10.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Revised: 09/03/2007] [Accepted: 10/10/2007] [Indexed: 01/28/2023]
Abstract
The somatostatin receptor subtypes 1-5 (sst(1)-sst(5)) exhibit different intracellular trafficking and endosomal sorting after agonist exposure. The internalization of the somatostatin receptor subtypes sst(2), sst(3) and sst(5) occurs to a much higher extent after somatostatin exposure than of sst(1) or sst(4). After endocytosis, sst(2) and sst(5) recycle to the plasma membrane, whereas sst(3) is predominantly down-regulated. This review will focus on the molecular mechanisms of the differential intracellular trafficking of sst(2), sst(3) and sst(5), and discusses our current knowledge on somatostatin receptor interacting proteins.
Collapse
Affiliation(s)
- Stefan Jacobs
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Würzburg, Germany
| | | |
Collapse
|
12
|
Kumar U. Colocalization of somatostatin receptor subtypes (SSTR1-5) with somatostatin, NADPH-diaphorase (NADPH-d), and tyrosine hydroxylase in the rat hypothalamus. J Comp Neurol 2007; 504:185-205. [PMID: 17626271 DOI: 10.1002/cne.21444] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The hypothalamus is a major site of somatostatin (SST) production and action. SST is synthesized in several hypothalamic nuclei and involved in a variety of functions. Using SST receptor (SSTR)-specific antibodies, we localized SSTR subtypes in the rat hypothalamus. In addition, we also demonstrated SSTRs colocalization with SST, NADPH-diaphorase (NADPH-d), and tyrosine hydroxylase (TH). SSTR1 is strongly localized in neurons in all major hypothalamic nuclei as well as in nerve fibers in the zona externa of the median eminence and the ependyma of the third ventricle. SSTR2 is also well expressed in most regions but with a relatively lower abundance in comparison to SSTR1. In contrast, SSTR3 is localized primarily in the paraventricular nucleus, dorsomedial hypothalamic nucleus, arcuate nucleus, and median eminence. SSTR4-like immunoreactivity is mainly confined to the arcuate nucleus, ventromedial hypothalamic nucleus, median eminence, and ependymal cells of third ventricle, with the rare SSTR4-positive neuron in the paraventricular nucleus. SSTR5 is the least expressed subtype occurring only in few cells in the inner layer of the median eminence. Overall, SSTR1 is the predominant subtype, followed by SSTR2, 4, 3, and 5. Combined immunofluorescence, immunocytochemistry, and histochemistry were used to demonstrate SSTRs colocalization with SST, TH, and NADPH-d. SSTRs colocalization with SST, TH, and NADPH-d displays in a region and receptor specificity. Colocalization of SST and NADPH-d with SSTRs in hypothalamic regions was similar, suggesting that SST and NADPH-d producing cells are same. In contrast, TH was selectively coexpressed with SSTRs in the hypothalamus in a receptor-specific manner. Taken together, these data suggest that SSTRs may interact with NADPH-d and TH to exert a physiological role in concert within the hypothalamus.
Collapse
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
13
|
Abstract
The experimental data reviewed in the present paper deal with the molecular events underlying the agonist-dependent regulation of the distinct somatostatin receptor subtypes and may suggest important clues about the clinical use of somatostatin analogs with different pattern of receptor specificity for the in vivo targeting of tumoral somatostatin receptors. Somatostatin receptor subtypes are characterized by differential beta-arrestin trafficking and endosomal sorting upon agonist binding due, at least in part, to the differences in their C-terminal tails. Moreover, the subcellular expression pattern of somatostatin receptor subtypes and their activity in response to agonist treatment are affected by intracellular complements, such as proteins involved in intracellular vesicle trafficking. Different somatostatin analogs may induce distinct conformations of the receptor/ligand complex, preferentially coupled to either receptor signaling or receptor endocytosis.
Collapse
|
14
|
Wang Y, Su B, Xia Z. Brain-derived Neurotrophic Factor Activates ERK5 in Cortical Neurons via a Rap1-MEKK2 Signaling Cascade. J Biol Chem 2006; 281:35965-74. [PMID: 17003042 DOI: 10.1074/jbc.m605503200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The extracellular signal-regulated kinase 5 (ERK5) is activated in neurons of the central nervous system by neurotrophins including brain-derived neurotrophic factor (BDNF). Although MEK5 is known to mediate BDNF stimulation of ERK5 in central nervous system neurons, other upstream signaling components have not been identified. Here, we report that BDNF induces a sustained activation of ERK5 in rat cortical neurons and activates Rap1, a small GTPase, as well as MEKK2, a MEK5 kinase. Our data indicate that activation of Rap1 or MEKK2 is sufficient to stimulate ERK5, whereas inhibition of either Rap1 or MEKK2 attenuates BDNF activation of ERK5. Furthermore, BDNF stimulation of MEKK2 is regulated by Rap1. Our evidence also indicates that Ras and MEKK3, a MEK5 kinase in non-neuronal cells, do not play a significant role in BDNF activation of ERK5. This study identifies Rap1 and MEKK2 as critical upstream signaling molecules mediating BDNF stimulation of ERK5 in central nervous system neurons.
Collapse
Affiliation(s)
- Yupeng Wang
- Toxicology Program, Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195-7234, USA
| | | | | |
Collapse
|
15
|
Garduño-Torres B, Arias-Montaño JA. Homologous down-regulation of histamine H3 receptors in rat striatal slices. Synapse 2006; 60:165-71. [PMID: 16715497 DOI: 10.1002/syn.20288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Preincubation of striatal slices with the selective histamine H3-receptor agonist immepip (100 nM) decreased the specific binding of N-alpha-[methyl-3H]-histamine ([3H]-NMHA) to membranes obtained from the treated slices. The binding decrease was significant after 5 min, remained at similar reduced levels between 5- and 30-min incubations with agonist, and only a partial recovery was observed after 90-min washout (34, 41, and 44% at 90, 120, and 150 min, respectively). Saturation analysis showed a significant decrease in both receptor density (-44% +/- 9%) and affinity (dissociation constant, Kd 1.15 +/- 0.23 nM from 0.59 +/- 0.17 nM). The effect of immepip was mimicked by histamine and the H3 agonists imetit and R-alpha-methylhistamine, and was blocked by the H3 antagonist thioperamide. The reduction in [3H]-NMHA binding was fully and partially prevented by incubation at 4 degrees C and in hypertonic medium, respectively, but not by the endocytosis inhibitor phenylarsine oxide (10 microM). None of the following protein kinase inhibitors, Ro-318220 and Gö-6976 (PKC), H-89 (PKA) and staurosporine (general inhibitor) prevented the effect of immepip. In [3H]-adenine-labeled slices the preincubation with immepip (100 nM, 15 min) prevented the inhibitory effect of H3 receptor activation on forskolin-induced [3H]-cAMP accumulation (99% +/- 9% vs. 76% +/- 4% of control values). Taken together our results indicate that agonist binding promotes the down-regulation of striatal H3 receptors resulting in a significant loss of function.
Collapse
Affiliation(s)
- Belén Garduño-Torres
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Apdo. Postal 14-740, 07000 México, D.F., Mexico
| | | |
Collapse
|
16
|
Stroh T, Sarret P, Tannenbaum GS, Beaudet A. Immunohistochemical Distribution and Subcellular Localization of the Somatostatin Receptor Subtype 1 (sst1) in the Rat Hypothalamus. Neurochem Res 2006; 31:247-57. [PMID: 16518576 DOI: 10.1007/s11064-005-9013-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2005] [Indexed: 10/25/2022]
Abstract
The aim of the present study was to examine the cellular and sub-cellular distribution of the somatostatin (SRIF) receptor subtype sst1 in the rat hypothalamus. Receptors were immunolabeled using an antibody directed against an antigenic sequence in the N-terminus of the receptor. Immunopositive neuronal cell bodies and dendrites were observed throughout the mediobasal hypothalamus, including the medial preoptic area, paraventricular, periventricular, and arcuate nuclei. Immunoreactive axons and axon terminals were also observed in the median eminence, suggesting that sst1 is also located pre-synaptically. Electron microscopic examination of the arcuate nucleus revealed a predominant association of immunoreactive sst1 with perikarya and dendrites. Most immunoreactive receptors were intracellular and localized to tubulovesicular compartments and organelles such as the Golgi apparatus, but 14% were associated with the plasma membrane. Of the latter, 47% were apposed to abbuting afferent axon terminals and 20% localized immediately adjacent to an active synaptic zone. These results demonstrate a widespread distribution of sst1 receptors in rat hypothalamus. They also show that somatodendritic sst1 receptors in the arcuate nucleus are ideally poised to mediate SRIF's modulation of afferent synaptic inputs, including central SRIF effects on growth hormone-releasing hormone neurons documented in this area.
Collapse
Affiliation(s)
- Thomas Stroh
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, 3801, University Street, H3A 2B4 Montréal, Québec, Canada.
| | | | | | | |
Collapse
|
17
|
Masmoudi O, Gandolfo P, Tokay T, Leprince J, Ravni A, Vaudry H, Tonon MC. Somatostatin down-regulates the expression and release of endozepines from cultured rat astrocytes via distinct receptor subtypes. J Neurochem 2005; 94:561-71. [PMID: 16033415 DOI: 10.1111/j.1471-4159.2005.03076.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Endozepines, a family of regulatory peptides related to diazepam-binding inhibitor (DBI), are synthesized and released by astroglial cells. Because rat astrocytes express various subtypes of somatostatin receptors (sst), we have investigated the effect of somatostatin on DBI mRNA level and endozepine secretion in rat astrocytes in secondary culture. Somatostatin reduced in a concentration-dependent manner the level of DBI mRNA in cultured astrocytes. This inhibitory effect was mimicked by the selective sst4 receptor agonist L803-087 but not by the selective sst1, sst2 and sst3 receptor agonists L779-591, L779-976 and L797-778, respectively. Somatostatin was unable to further reduce DBI mRNA level in the presence of the MEK inhibitor U0126. Somatostatin and the sst1, sst2 and sst4 receptor agonists induced a concentration-dependent inhibition of endozepine release. Somatostatin and the sst1, sst2 and sst4 receptor agonists also inhibited cAMP formation dose-dependently. In addition, somatostatin reduced forskolin-induced endozepine release. H89 mimicked the inhibitory effect of somatostatin on endozepine secretion. In contrast the PLC inhibitor U73122, the PKC activator PMA and the PKC inhibitor calphostin C had no effect on somatostatin-induced inhibition of endozepine release. The present data demonstrate that somatostatin reduces DBI mRNA level mainly through activation of sst4 receptors negatively coupled to the MAPK pathway, and inhibits endozepine release through activation of sst1, sst2 and sst4 receptors negatively coupled to the adenylyl cyclase/PKA pathway.
Collapse
Affiliation(s)
- Olfa Masmoudi
- European Institute for Peptide Research (IFRMP 23), Laboratory of Cellular and Molecular Neuroendocrinology, INSERM U413, UA CNRS, University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | |
Collapse
|
18
|
Moody TW, Fuselier J, Coy DH, Mantey S, Pradhan T, Nakagawa T, Jensen RT. Camptothecin-somatostatin conjugates inhibit the growth of small cell lung cancer cells. Peptides 2005; 26:1560-6. [PMID: 16112393 DOI: 10.1016/j.peptides.2005.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Revised: 02/22/2005] [Accepted: 02/23/2005] [Indexed: 11/20/2022]
Abstract
The effects of camptothecin-somatostatin (CPT-SS) conjugates were investigated on small cell lung cancer (SCLC) cells. CPT was coupled to a SS agonist (SSA), c(Cys-Phe-DTrp-Lys-Thr-Cys)Thr-NH2 using the built in nucleophile assisted-releasing group (L1) N-methyl-aminoethyl-Gly-Dser-Nle-Dtyr-Dser or (L2) aminoethyl-Gly-Dser-Nle-Dtyr-Dser. The resulting CPT-L1-SSA and CPT-L2-SSA inhibited the specific binding of [125I-Tyr11]SS to NCI-H69 cell membranes with IC50 values of 0.2 and 2.1 nM, respectively. [125I]CPT-L1-SSA was internalized by SCLC cells at 37 degrees C but not at 4 degrees C. CPT-L1-SSA and CPT-L2-SSA inhibited in a dose-dependent manner the increase in adenylylcyclase activity caused by 25 microM forskolin. In vitro, 0.3 microM CPT-L1-SSA half-maximally inhibited the clonal growth of SCLC cells and 1 microM CPT-L1-SSA strongly inhibited 3H-thymidine incorporation into DNA and trypan-blue exclusion. These results suggest that CPT conjugated peptides such as CPT-L1-SSA may prove useful for exploring the efficacy of receptor-directed cytotoxicity to inhibit the proliferation of SCLC cells.
Collapse
Affiliation(s)
- Terry W Moody
- DHHS, NIH, NCI Office of the Director, CCR, 31 Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Ben-Shlomo A, Wawrowsky KA, Proekt I, Wolkenfeld NM, Ren SG, Taylor J, Culler MD, Melmed S. Somatostatin receptor type 5 modulates somatostatin receptor type 2 regulation of adrenocorticotropin secretion. J Biol Chem 2005; 280:24011-21. [PMID: 15857828 DOI: 10.1074/jbc.m501998200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Somatostatin inhibits adrenocorticotropin (ACTH) secretion from pituitary tumor cells. To assess the contribution of somatostatin receptor subtype 5 (SST5) to somatostatin receptor subtype 2 (SST2) action in these cells, we assessed multipathway responses to novel highly monoreceptor-selective peptide agonists and multireceptor agonists, including octreotide and somatostatin-28. Octreotide and somatostatin-28 cell membrane binding affinities correlated with their respective SST2-selective peptide ligand. Although octreotide had similar inhibiting potency (picomolar) for cAMP accumulation and ACTH secretion as an SST2-selective agonist, somatostatin-28 exhibited a higher potency (femtomolar). Baseline spontaneous calcium oscillations assessed by fluorescent confocal microscopy revealed two distinct effects: SST2 activation reduced oscillations at femtomolar concentrations reflected by high inhibiting potency of averaged normalized oscillation amplitude, whereas SST5 activation induces brief oscillation pauses and increased oscillation amplitude. Octreotide exhibits an integrated effect of both receptors; however, somatostatin-28 exhibited a complex response with two separate inhibitory potencies. SST2 internalization was visualized with SST2-selective agonist at lower concentrations than for octreotide or somatostatin-28, whereas SST5 did not internalize. Using monoreceptor-selective peptide agonists, the results indicate that, in AtT-20 cells, SST5 regulates the dominant SST2 action, attenuating SST2 effects on intracellular calcium oscillation and internalization. This may explain superior somatostatin-28 potency and provides a rationale for somatostatin ligand design to treat ACTH-secreting pituitary tumors.
Collapse
Affiliation(s)
- Anat Ben-Shlomo
- Department of Medicine, Cedars Sinai Research Institute, David Geffen School of Medicine, UCLA, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Pujol F, Kitabgi P, Boudin H. The chemokine SDF-1 differentially regulates axonal elongation and branching in hippocampal neurons. J Cell Sci 2005; 118:1071-80. [PMID: 15731012 DOI: 10.1242/jcs.01694] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent data have shown that the chemokine SDF-1 plays a critical role in several aspects of brain development such as cell migration and axon pathfinding. However, its potential function in the generation of axons and dendrites is poorly characterized. In order to better understand the role of SDF-1 in the development of central neurons, we studied the cellular distribution of the SDF-1 receptor CXCR4 by immunocytochemistry of developing hippocampal neurons and tested the effect of SDF-1 in process patterning at the early stages of neuronal development. We found that CXCR4 immunoreactivity undergoes a striking redistribution during development. At the early stages, from day 2 to day 4 in culture, CXCR4 is particularly concentrated at the leading edge of growing neurites. As the cells mature, staining declines at the tip of the processes and becomes more broadly distributed along axons and, to a lesser extent, dendrites. SDF-1 stimulation of neurons at day 1-2 in culture triggers several effects on neuronal morphogenesis. SDF-1 reduces growth cone number and axonal outgrowth but stimulates axonal branching. These latter two effects are not observed in other neurites. This study unravels a new role for SDF-1/CXCR4 in specifying hippocampal neuron morphology by regulating axonal patterning at an early stage of neuronal development.
Collapse
Affiliation(s)
- Fabien Pujol
- INSERM E0350, Hospital St Antoine, 184 rue du Fg St Antoine, 75571 Paris CEDEX 12, France
| | | | | |
Collapse
|
21
|
Kopecky M, Semecky V, Trejtnar F, Laznicek M, Laznickova A, Nachtigal P, Decristoforo C, Mather SJ, Mäcke HR. Analysis of accumulation of 99mTc-octreotide and 99mTc-EDDA/HYNIC-Tyr3-octreotide in the rat kidneys. Nucl Med Biol 2004; 31:231-9. [PMID: 15013489 DOI: 10.1016/j.nucmedbio.2003.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2002] [Revised: 01/31/2003] [Accepted: 06/26/2003] [Indexed: 11/17/2022]
Abstract
The aim of this study was to compare renal handling and distribution of (99m)Tc-octreotide and (99m)Tc-EDDA/HYNIC-Tyr(3)-octreotide (HYNIC-TOC) in rats. In kidney perfusion experiments, the renal clearance value of (99m)Tc-octreotide was three times lower than that of (99m)Tc-EDDA/HYNIC-TOC. The predominant renal excretion of (99m)Tc-EDDA/HYNIC-TOC was associated with a high and long-term renal accumulation up to 48 hrs. Microautoradiographic results indicated that (99m)Tc-EDDA/HYNIC-TOC was retained mainly in the renal medulla within the cells of the collecting ducts and in the surrounding tissue. Lower positivity was found in the proximal and distal tubular cells. We conclude that the mechanism of renal accumulation of somatostatin analogues renal accumulation is complex and that proximal tubular reabsorption is probably not the main mechanism for uptake of (99m)Tc-EDDA/HYNIC-TOC in the kidneys. The presence of the somatostatin receptors, differences in the tonicity level within kidneys and other possible mechanisms could participate in their renal accumulation.
Collapse
Affiliation(s)
- Martin Kopecky
- Department of Biological and Medical Sciences, Charles University, Faculty of Pharmacy, Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Basheer R, Strecker RE, Thakkar MM, McCarley RW. Adenosine and sleep–wake regulation. Prog Neurobiol 2004; 73:379-96. [PMID: 15313333 DOI: 10.1016/j.pneurobio.2004.06.004] [Citation(s) in RCA: 358] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2003] [Accepted: 06/28/2004] [Indexed: 10/26/2022]
Abstract
This review addresses three principal questions about adenosine and sleep-wake regulation: (1) Is adenosine an endogenous sleep factor? (2) Are there specific brain regions/neuroanatomical targets and receptor subtypes through which adenosine mediates sleepiness? (3) What are the molecular mechanisms by which adenosine may mediate the long-term effects of sleep loss? Data suggest that adenosine is indeed an important endogenous, homeostatic sleep factor, likely mediating the sleepiness that follows prolonged wakefulness. The cholinergic basal forebrain is reviewed in detail as an essential area for mediating the sleep-inducing effects of adenosine by inhibition of wake-promoting neurons via the A1 receptor. The A2A receptor in the subarachnoid space below the rostral forebrain may play a role in the prostaglandin D2-mediated somnogenic effects of adenosine. Recent evidence indicates that a cascade of signal transduction induced by basal forebrain adenosine A1 receptor activation in cholinergic neurons leads to increased transcription of the A1 receptor; this may play a role in mediating the longer-term effects of sleep deprivation, often called sleep debt.
Collapse
Affiliation(s)
- Radhika Basheer
- Neuroscience Laboratory, Department of Psychiatry, Harvard Medical School and Boston VA Healthcare System, Brockton, MA 02301, USA
| | | | | | | |
Collapse
|
23
|
Zhang Q, Pangrsic T, Kreft M, Krzan M, Li N, Sul JY, Halassa M, Van Bockstaele E, Zorec R, Haydon PG. Fusion-related release of glutamate from astrocytes. J Biol Chem 2004; 279:12724-33. [PMID: 14722063 DOI: 10.1074/jbc.m312845200] [Citation(s) in RCA: 192] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although cell culture studies have implicated the presence of vesicle proteins in mediating the release of glutamate from astrocytes, definitive proof requires the identification of the glutamate release mechanism and the localization of this mechanism in astrocytes at synaptic locales. In cultured murine astrocytes we show an array of vesicle proteins, including SNARE proteins, and vesicular glutamate transporters that are required to fill vesicles with glutamate. Using immunocytochemistry and single-cell multiplex reverse transcription-PCR we demonstrate the presence of these proteins and their transcripts within astrocytes freshly isolated from the hippocampus. Moreover, immunoelectron microscopy demonstrates the presence of VGLUT1 in processes of astrocytes of the hippocampus. To determine whether calcium-dependent glutamate release is mediated by exocytosis, we expressed the SNARE motif of synaptobrevin II to prevent the formation of SNARE complexes, which reduces glutamate release from astrocytes. To further determine whether vesicular exocytosis mediates calcium-dependent glutamate release from astrocytes, we performed whole cell capacitance measurements from individual astrocytes and demonstrate an increase in whole cell capacitance, coincident with glutamate release. Together, these data allow us to conclude that astrocytes in situ express vesicle proteins necessary for filling vesicles with the chemical transmitter glutamate and that astrocytes release glutamate through a vesicle- or fusion-related mechanism.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Liu Q, Reubi JC, Wang Y, Knoll BJ, Schonbrunn A. In vivo phosphorylation of the somatostatin 2A receptor in human tumors. J Clin Endocrinol Metab 2003; 88:6073-9. [PMID: 14671213 DOI: 10.1210/jc.2003-030986] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Hormone-stimulated receptor internalization and desensitization occur widely in the G protein-coupled receptor (GPCR) family. A critical first step in both these processes is thought to be receptor phosphorylation, a reaction which has been extensively characterized in cell culture. However, little is known about GPCR phosphorylation in vivo. The somatostatin (SS) receptor subtype (sst)2A is widely distributed in human neuroendocrine tumors, and SS analogs are commonly used to target this receptor for both therapy and diagnosis. In cultured pituitary cells sst2A is rapidly phosphorylated and internalized after hormone binding. The aim of the present study was to go one crucial step further and characterize the phosphorylation state of this receptor in human neuroendocrine tumors using a newly developed gel-shift assay. The receptor from a somatostatinoma was completely phosphorylated. In contrast, only unphosphorylated sst2A was present in human tumors that were not exposed to autocrine stimulation. Both in vivo and in cultured cells, the phosphorylation state of the sst2A receptor was correlated with its subcellular localization: phosphorylated receptor was mostly intracellular, whereas unphosphorylated receptor was localized at the cell surface. These results are the first to demonstrate ligand-stimulated GPCR phosphorylation in human tissue in situ, providing a crucial step toward a better understanding of receptor regulation in vivo. Analysis of sst2A phosphorylation promises to provide a sensitive indicator of the effectiveness of SS analogs in diagnostic and therapeutic situations in tumor patients.
Collapse
Affiliation(s)
- Qisheng Liu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center-Houston, Houston, Texas 77225, USA
| | | | | | | | | |
Collapse
|
25
|
Konishi SI, Kasagi Y, Katsumata H, Minami S, Imaki T. Regulation of corticotropin-releasing factor (CRF) type-1 receptor gene expression by CRF in the hypothalamus. Endocr J 2003; 50:21-36. [PMID: 12733706 DOI: 10.1507/endocrj.50.21] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We reported previously that acute stress and intracerebroventricular (i.c.v.) injection of corticotropin-releasing factor (CRF) increased neuronal activation and CRF type-1 receptor (CRFR-1) mRNA expression in the CRF-producing neurons of the parvocellular paraventricular nucleus (PVN) of the hypothalamus. In this study, to determine whether CRF can act directly on hypothalamic CRF neurons, thereby increasing CRFR-1 expression, microinjection of CRF into PVN neurons in vivo and primary cultures of dispersed rat fetal hypothalami in vitro were performed. Microinjection of 0.1 microg of CRF into the PVN significantly increased c-fos and CRFR-1 mRNA expression in the CRF-producing parvocellular PVN, 30 min or 180 min after injection, respectively. This effect was blocked by a CRF antagonist, alpha-helical CRF. CRF, when injected into the lateral ventricle at the same dose, increased neither CRFR-1 nor c-fos mRNA levels in the PVN. Primary culture of hypothalamic neurons revealed that CRFR-1 like immunoreactivity was located in CRF-containing neurons, and that the CRFR-1 mRNA level was significantly increased 4 h after incubation with 10(-8) M CRF. These results demonstrate that CRF directly affects hypothalamic neurons to increase CRFR-1 mRNA expression, providing evidence of a direct role for CRF in the regulation of CRFR-1 expression of hypothalamic neurons.
Collapse
Affiliation(s)
- Shun-ichiro Konishi
- Department of Bioregulation, Institute of Gerontology, Nippon Medical School, Kawasaki 211-8533, Japan
| | | | | | | | | |
Collapse
|
26
|
Peineau S, Potier B, Petit F, Dournaud P, Epelbaum J, Gardette R. AMPA-sst2 somatostatin receptor interaction in rat hypothalamus requires activation of NMDA and/or metabotropic glutamate receptors and depends on intracellular calcium. J Physiol 2003; 546:101-17. [PMID: 12509482 PMCID: PMC2342459 DOI: 10.1113/jphysiol.2002.025890] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Modulation of glutamatergic transmission by neuropeptides is an essential aspect of neuronal network activity. Activation of the hypothalamic somatostatin sst2 receptor subtype by octreotide decreases AMPA glutamate responses, indicating a central link between a neurohormonal and neuromodulatory peptide and the main hypothalamic fast excitatory neurotransmitter. In mediobasal hypothalamic slices, sst2 activation inhibits the AMPA component of glutamatergic synaptic responses but is ineffective when AMPA currents are pharmacologically isolated. In mediobasal hypothalamic cultures, the decrease of AMPA currents induced by octreotide requires a concomitant activation of sst2 receptors with either NMDA and/or metabotropic glutamate receptors. This modulation depends on changes in intracellular calcium concentration induced by calcium flux through NMDA receptors or calcium release from intracellular stores following metabotropic glutamate receptor activation. These results highlight an unusual regulatory mechanism in which the simultaneous activation of at least three different types of receptor is necessary to allow somatostatin-induced modulation of fast synaptic glutamatergic transmission in the hypothalamus.
Collapse
Affiliation(s)
- Stéphane Peineau
- INSERM U549, IFR Broca Sainte Anne, 2ter rue d'Alésia, 75014 Paris, France
| | | | | | | | | | | |
Collapse
|
27
|
Csaba Z, Simon A, Helboe L, Epelbaum J, Dournaud P. Neurochemical characterization of receptor-expressing cell populations by in vivo agonist-induced internalization: insights from the somatostatin sst2A receptor. J Comp Neurol 2002; 454:192-9. [PMID: 12412143 DOI: 10.1002/cne.10430] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Characterization of both neurochemical phenotype of G protein-coupled receptor (GPCR)-expressing cells and receptor compartmentalization is a prerequisite for the elucidation of receptor functions in the central nervous system. However, it is often prevented by the diffuse and homogeneous distribution of receptor immunoreactivity. This is particularly true for the somatostatin (SRIF) sst2A receptor, which is largely distributed in the mammalian brain. By using this receptor as a model, we investigated whether receptor internalization, a biochemical property shared by numerous GPCRs, would reveal sst2A-expressing cell populations in the rat dorsolateral septum (LSD), a region in which SRIF might play an important modulatory role. Thirty minutes to 1 hour after intracerebroventricular injection of the sst2A receptor agonist octreotide, numerous sst2A-immunoreactive neurons and processes became apparent due to intracytoplasmic accumulation of intensely stained granules. Double-immunolabeling experiments with synaptophysin and MAP2 provided evidence that internalized sst2A receptors are predominantly localized in the somatodendritic compartment. Revealing sst2A receptor-expressing cell bodies permitted to analyze their neurotransmitter content. Quantitative analysis demonstrated an extensive overlap (approximately 85%) between SRIF- and sst2A-expressing neuronal populations. Additionally, numerous SRIF-immunoreactive axon-like terminals were found in close apposition with sst2A-positive cell bodies and dendrites. Taken together, these data suggest that the sst2A receptor is predominantly expressed in LSD neurons as a postsynaptic autoreceptor, thus providing novel neuroanatomic clues to elucidate SRIF neurotransmission in this region. More generally, in vivo agonist-induced internalization appears as a rapid and powerful tool for the neurochemical characterization of GPCR-expressing cell populations in the mammalian brain.
Collapse
Affiliation(s)
- Zsolt Csaba
- INSERM U549, IFR Broca-Sainte Anne, Centre Paul Broca, 75014 Paris, France
| | | | | | | | | |
Collapse
|
28
|
Nagele RG, D'Andrea MR, Anderson WJ, Wang HY. Intracellular accumulation of beta-amyloid(1-42) in neurons is facilitated by the alpha 7 nicotinic acetylcholine receptor in Alzheimer's disease. Neuroscience 2002; 110:199-211. [PMID: 11958863 DOI: 10.1016/s0306-4522(01)00460-2] [Citation(s) in RCA: 319] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Amyloid beta(1-42), a major component of amyloid plaques, binds with exceptionally high affinity to the alpha 7 nicotinic acetylcholine receptor and accumulates intracellularly in neurons of Alzheimer's disease brains. In this study, we investigated the possibility that this binding plays a key role in facilitating intraneuronal accumulation of amyloid beta(1-42). Consecutive section immunohistochemistry and digital imaging were used to reveal the spatial relationship between amyloid beta(1-42) and the alpha 7 receptor in affected neurons of Alzheimer's disease brains. Results showed that neurons containing substantial intracellular accumulations of amyloid beta(1-42) invariably express relatively high levels of the alpha 7 receptor. Furthermore, this receptor is highly co-localized with amyloid beta(1-42) within neurons of Alzheimer's disease brains. To experimentally test the possibility that the binding interaction between exogenous amyloid beta(1-42) and the alpha 7 receptor facilitates internalization and intracellular accumulation of amyloid beta(1-42) in Alzheimer's disease brains, we studied the fate of exogenous amyloid beta(1-42) and its interaction with the alpha 7 receptor in vitro using cultured, transfected neuroblastoma cells that express elevated levels of this receptor. Transfected cells exhibited rapid binding, internalization and accumulation of exogenous amyloid beta(1-42), but not amyloid beta(1-40). Furthermore, the rate and extent of amyloid beta(1-42) internalization was related directly to the alpha 7 receptor protein level, since (1) the rate of amyloid beta(1-42) accumulation was much lower in untransfected cells that express much lower levels of this receptor and (2) internalization was effectively blocked by alpha-bungarotoxin, an alpha 7 receptor antagonist. As in neurons of Alzheimer's disease brains, the alpha 7 receptor in transfected cells was precisely co-localized with amyloid beta(1-42) in prominent intracellular aggregates. Internalization of amyloid beta(1-42) in transfected cells was blocked by phenylarsine oxide, an inhibitor of endocytosis. We suggest that the intraneuronal accumulation of amyloid beta(1-42) in Alzheimer's disease brains occurs predominantly in neurons that express the alpha 7 receptor. In addition, internalization of amyloid beta(1-42) may be facilitated by the high-affinity binding of amyloid beta(1-42) to the alpha 7 receptor on neuronal cell surfaces, followed by endocytosis of the resulting complex. This provides a plausible explanation for the selective vulnerability of neurons expressing the alpha 7 receptor in Alzheimer's disease brains and for the fact that amyloid beta(1-42) is the dominant amyloid beta peptide species in intracellular accumulations and amyloid plaques.
Collapse
Affiliation(s)
- R G Nagele
- Department of Molecular Biology, University of Medicine and Dentistry of New Jersey, School of Osteopathic Medicine, Stratford, 08084, USA.
| | | | | | | |
Collapse
|
29
|
Saha S, Henderson Z, Batten TFC. Somatostatin immunoreactivity in axon terminals in rat nucleus tractus solitarii arising from central nucleus of amygdala: coexistence with GABA and postsynaptic expression of sst2A receptor. J Chem Neuroanat 2002; 24:1-13. [PMID: 12084407 DOI: 10.1016/s0891-0618(02)00013-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Axon terminals synapsing on neurones in the nucleus tractus solitarii (NTS) that originate from the central nucleus of the amygdala (CeA) have been shown to contain gamma-aminobutyric acid (GABA) immunoreactivity. Here we investigated whether such terminals also contain somatostatin (SOM), a neuropeptide found in axons distributed throughout the NTS and in somata in the CeA, and known to modulate cardiovascular reflexes when microinjected into the NTS. With fluorescence microscopy, SOM immunoreactivity was seen in the varicosities of some axons throughout the NTS that were anterogradely labelled with biotin dextran amine injected into the CeA. Such varicosities were frequently observed in close proximity to dendrites of NTS neurones that were immunoreactive for the SOM receptor sst(2A) subtype, and in many cases also for catecholamine synthesising enzymes. In the caudal, cardioregulatory zone of NTS, SOM immunoreactivity was localised by electron microscopic pre-embedding gold labelling to boutons containing dense-cored and clear pleomorphic vesicles and forming symmetrical synapses, mostly onto dendrites. Additional post-embedding gold labelling for GABA suggested that a subpopulation (29%) of GABAergic terminals sampled in this area of NTS contained SOM. Almost all boutons anterogradely labelled from the amygdala were GABA-immunoreactive (-IR) and 21% of these were SOM-IR. A similar proportion of these boutons (22%) formed synapses onto dendrites containing immunoreactivity for the SOM receptor sst(2A) subtype. These observations provide evidence that some of the GABAergic projection neurones in the CeA that inhibit baroreceptor reflex responses in the NTS in response to fear or emotional stimuli could release SOM, which might modulate the activity of NTS neurones via an action on sst(2A) receptors.
Collapse
Affiliation(s)
- S Saha
- Institute for Cardiovascular Research, School of Medicine, Worsley Building, University of Leeds, Leeds LS2 9JT, UK
| | | | | |
Collapse
|
30
|
Lee MC, Cahill CM, Vincent JP, Beaudet A. Internalization and trafficking of opioid receptor ligands in rat cortical neurons. Synapse 2002; 43:102-11. [PMID: 11754488 DOI: 10.1002/syn.10014] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The binding, internalization, and trafficking of the fluorescently labeled opioid peptides Fluo-dermorphin and Fluo-deltorphin were quantitatively studied by confocal microscopy in primary cortical neurons in culture. Specific binding of these selective ligands to neurons naturally expressing mu (mu) and delta (delta) opioid receptors (OR), respectively, resulted in their internalization into neuronal somas and processes, as indicated by the persistence of fluorescent labeling following removal of cell surface binding by hypertonic acid wash. This internalization was receptor-specific, as the fluorescent signal was completely abolished when the cells were concomitantly incubated with the opioid receptor antagonist naloxone. It also was clathrin-dependent, as it was totally prevented by the endocytosis inhibitor phenylarsine oxide. Accordingly, internalized ligands were detected inside small, endosome-like vesicles. These labeled vesicles accumulated within nerve cell bodies between 5-30 min of incubation with the fluorescent ligands. This accumulation was abolished after treatment with the antitubular agent nocodazole, suggesting that it was due to a microtubule-dependent, retrograde transport of the internalized ligands from processes to the soma. By contrast, there was no change in the compartmentalization of internalized (mu)OR or deltaOR, as assessed by immunocytochemistry, suggesting that the latter were recycled locally. The present results provide the first demonstration of receptor-mediated internalization of opioid peptides in cultured neurons. It is proposed that their retrograde transport into target cells might be involved in mediating some of the long-term, transcriptional effects of opioids.
Collapse
Affiliation(s)
- Mao-Cheng Lee
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
31
|
Riad M, Watkins KC, Doucet E, Hamon M, Descarries L. Agonist-induced internalization of serotonin-1a receptors in the dorsal raphe nucleus (autoreceptors) but not hippocampus (heteroreceptors). J Neurosci 2001; 21:8378-86. [PMID: 11606626 PMCID: PMC6762788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2001] [Revised: 08/16/2001] [Accepted: 08/23/2001] [Indexed: 02/21/2023] Open
Abstract
Serotonin-1A (5-HT(1A)) receptors in the CNS are a major target for psychotropic drugs. In nucleus raphe dorsalis (NRD) and hippocampus (CA3), the selective 5-HT(1A) agonist (+)-8-hydroxy-2-(di-N-propylamino) tetralin (8-OH-DPAT) reduces the firing activity of serotoninergic (5-HT) and pyramidal neurons, respectively. When located on 5-HT (autoreceptors), but not on non-5-HT (heteroreceptors) neurons, 5-HT(1A) receptors are known to be subject to desensitization. Using quantitative electron microscopy after pre-embedding immunogold labeling with specific antibodies, we examined the subcellular distribution of these receptors after acute administration of 8-OH-DPAT (0.5 mg/kg, i.v.). Silver-intensified immunogold particles associated with the plasma membrane or the cytoplasm were counted in somata and dendrites within the NRD, 15 min, 1 hr and 24 hr after 8-OH-DPAT injection, and in hippocampal dendrites 1 hr after the same treatment. Significant decrease in the density of membrane labeling and concomitant increase of cytoplasmic labeling were demonstrated in the NRD, 15 min and 1 hr after 8-OH-DPAT administration, with a return to baseline level at 24 hr. Internalization was blocked by previous administration of the 5-HT(1A) antagonist N-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-N-(2-pyridinyl) cyclohexane-carboxamide (WAY 100635), which, by itself, was without apparent effect. In hippocampus (CA3), there were no apparent changes in the distribution of the receptor after 8-OH-DPAT administration. These findings are in line with earlier results showing a desensitization of 5-HT(1A) autoreceptors but not heteroreceptors after treatment with 5-HT(1A) receptor agonist. They suggest that this desensitization is the result of autoreceptor internalization.
Collapse
Affiliation(s)
- M Riad
- Départements de Pathologie et Biologie Cellulaire et de Physiologie, and Centre de Recherche en Sciences Neurologiques, Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada H3C 3J7.
| | | | | | | | | |
Collapse
|
32
|
Fontenele-Neto JD, Massarelli EE, Gurgel Garrido PA, Beaudet A, Ferro ES. Comparative fine structural distribution of endopeptidase 24.15 (EC3.4.24.15) and 24.16 (EC3.4.24.16) in rat brain. J Comp Neurol 2001; 438:399-410. [PMID: 11559896 DOI: 10.1002/cne.1323] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Endopeptidase 24.15 (EP24.15) and 24.16 (EP24.16) are closely related metalloendopeptidases implicated in the metabolism of several neuropeptides and widely expressed in mammalian brain. To gain insight into the functional role of these two enzymes in the central nervous system, we examined their cellular and subcellular distribution in rat brain by using electron microscopic immunogold labeling. In all areas examined, EP24.15 and EP24.16 immunoreactivity were observed in selective subpopulations of neuronal and glial cells. Subcellular localization of EP24.15 in neurons revealed that this enzyme was predominantly concentrated in the nucleus, whereas EP24.16 was almost exclusively cytoplasmic. The amount of EP24.15 found in the nucleus was inversely correlated with that found in the cytoplasm, suggesting that the enzyme could be mobilized from one compartment to the other. Within the cytoplasm, EP24.15 and EP24.16 immunoreactivity showed comparable distributional patterns. Both enzymes were detected throughout perikarya and dendrites, as well as within axons and axon terminals. In all neuronal compartments, EP24.15 and EP24.16 showed a major association with membranes of neurosecretory elements, including Golgi cisternae, tubulovesicular organelles, synaptic vesicles, and endosomes. However, whereas EP24.15 always faced the cytoplasmic face of the membranes, EP24.16 was observed on both cytoplasmic and luminal sides, suggesting that the latter was more likely to contribute to the processing of peptides or to the degradation of internalized ligands. Taken together, the present results suggest that EP24.15 could play a major role in the hydrolysis of intranuclear substrates, whereas EP24.16 would be predominantly involved in the processing and inactivation of signaling peptides.
Collapse
Affiliation(s)
- J D Fontenele-Neto
- Department of Histology and Embryology, Cell Biology Program, Biomedical Sciences Institute, USP, São Paulo 05508-900, SP, Brazil
| | | | | | | | | |
Collapse
|
33
|
Souazé F. Maintaining cell sensitivity to G-protein coupled receptor agonists: neurotensin and the role of receptor gene activation. J Neuroendocrinol 2001; 13:473-9. [PMID: 11328459 DOI: 10.1046/j.1365-2826.2001.00658.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the last few years, a number of studies have brought new insights into the fundamental mechanisms of cell desensitization and internalization of G-protein coupled receptors. Such studies have demonstrated that cells remain desensitized from a few minutes to several hours, after exposure to high concentrations of agonist. However, in vivo, agonists such as hormones are always present, even in small amounts, and such long desensitization is not conceivable, since constant stimulation of cells is required for physiological responses. Under such circumstances, cells would require a means to permanently maintain sensitivity to various internal or external stimuli. In the present review, we have taken as an example the expression of the high affinity neurotensin receptor, a seven transmembrane G-protein coupled receptor, upon prolonged exposure to its agonist, and observed that cells remained sensitive only if the receptor gene was activated by the agonist. Consequently, new receptors were synthesized, and either delivered to the cell surface or accumulated in submembrane pools. This regulation takes place only after prolonged and intense agonist stimulation. Under these conditions, it is proposed that receptor turnover is accelerated in proportion to the agonist concentration in order to allow the cells to produce an adapted cellular response to external stimuli. Such mechanisms thus play a key role in cell sensitivity to hormones.
Collapse
Affiliation(s)
- F Souazé
- INSERM U339, Hôpital St Antoine, Paris, France.
| |
Collapse
|
34
|
Csaba Z, Bernard V, Helboe L, Bluet-Pajot MT, Bloch B, Epelbaum J, Dournaud P. In vivo internalization of the somatostatin sst2A receptor in rat brain: evidence for translocation of cell-surface receptors into the endosomal recycling pathway. Mol Cell Neurosci 2001; 17:646-61. [PMID: 11312601 DOI: 10.1006/mcne.2000.0958] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To determine whether cellular compartmentalization of somatostatin receptors can be regulated in vivo, we examined the immunocytochemical distribution of the sst2A receptor (sst2AR) after stereotaxical injections of somatostatin analogs into the rat parietal cortex. Whereas CH-275, a sst1R agonist, failed to induce changes in the diffuse sst2AR immunostaining pattern characteristic of control animals, somatodendritic profiles displaying intracytoplasmic immunoreactive granules became apparent short-term after injection of either somatostatin or the sst2R agonist octreotide. Confocal microscopy revealed that 90% of sst2AR-immunoreactive endosome-like organelles displayed transferrin receptor immunoreactivity. At the electron microscopic level, the percentage of sst2AR immunoparticles dramatically decreased at the plasmalemma of perikarya and dendrites after octreotide injection. Conversely, it significantly increased in endosomes-like organelles. These results demonstrate that sst2ARs undergo, in vivo, rapid and massive internalization into the endocytic recycling compartment in response to acute agonist stimulation and provide important clues toward elucidating somatostatin receptor signaling in the mammalian brain.
Collapse
Affiliation(s)
- Z Csaba
- INSERM U549, IFR Broca-Sainte Anne, Centre Paul Broca, Paris, 75014, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Somatostatin, and the recently discovered neuropeptide cortistatin, exert their physiological actions via a family of six G protein-coupled receptors (sst1, sst2A, sst2B, sst3, sst4, sst5). Following the cloning of somatostatin receptors significant advances have been made in our understanding of their molecular, pharmacological and signaling properties although much progress remains to be done to define their physiological role in vivo. In this review, the present knowledge regarding neuroanatomical localization, signal transduction pathways, desensitization and internalization properties of somatostatin receptors is summarized. Evidence that somatostatin receptors can form homo- and heterodimers and can physically interact with members of the SSTRIP/Shank/ProSAP1/CortBP1 family is also discussed.
Collapse
Affiliation(s)
- Z Csaba
- Inserm U549, IFR Broca-Sainte Anne, Centre Paul Broca, Paris, France
| | | |
Collapse
|