Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 407
Filtrar
1.
Mol Brain ; 14(1): 130, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34429141

RESUMO

Somatostatin-expressing interneurons (SOM-INs) are a major subpopulation of GABAergic cells in CA1 hippocampus that receive excitation from pyramidal cells (PCs), and, in turn, provide feedback inhibition onto PC dendrites. Excitatory synapses onto SOM-INs show a Hebbian long-term potentiation (LTP) mediated by type 1a metabotropic glutamate receptors (mGluR1a) that is implicated in hippocampus-dependent learning. The neuropeptide somatostatin (SST) is also critical for hippocampal long-term synaptic plasticity, as well as learning and memory. SST effects on hippocampal PCs are well documented, but its actions on inhibitory interneurons remain largely undetermined. In the present work, we investigate the involvement of SST in long-term potentiation of CA1 SOM-IN excitatory synapses using pharmacological approaches targeting the somatostatinergic system and whole cell recordings in slices from transgenic mice expressing eYFP in SOM-INs. We report that application of exogenous SST14 induces long-term potentiation of excitatory postsynaptic potentials in SOM-INs via somatostatin type 1-5 receptors (SST1-5Rs) but does not affect synapses of PC or parvalbumin-expressing interneurons. Hebbian LTP in SOM-INs was prevented by inhibition of SSTRs and by depletion of SST by cysteamine treatment, suggesting a critical role of endogenous SST in LTP. LTP of SOM-IN excitatory synapses induced by SST14 was independent of NMDAR and mGluR1a, activity-dependent, and prevented by blocking GABAA receptor function. Our results indicate that endogenous SST may contribute to Hebbian LTP at excitatory synapses of SOM-INs by controlling GABAA inhibition, uncovering a novel role for SST in regulating long-term synaptic plasticity in somatostatinergic cells that may be important for hippocampus-dependent memory processes.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Neurônios GABAérgicos/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Somatostatina/fisiologia , Sinapses/efeitos dos fármacos , Animais , Proteínas de Bactérias , Cisteamina/farmacologia , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/metabolismo , Técnicas de Introdução de Genes , Genes Reporter , Humanos , Interneurônios/metabolismo , Proteínas Luminescentes , Masculino , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Peptídeos Cíclicos/farmacologia , Receptores de Glutamato Metabotrópico/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/fisiologia , Somatostatina/farmacologia , Sinapses/fisiologia
2.
Curr Oncol Rep ; 23(7): 80, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33937962

RESUMO

PURPOSE OF REVIEW: Pancreatic neuroendocrine neoplasms (panNENs) often present as advanced disease and there is little data to guide treatment sequencing in the advance disease setting. Therefore, we aim to provide a comprehensive summary of the current evidence supporting the use of systemic treatment for patients with diagnosis of advanced and metastatic panNENs, as well as to provide strategies for treatment selection and address challenges for treatment selection and sequencing of therapy. RECENT FINDINGS: Substantial advances have been made and many clinical trials have been performed over the past two decades expanding therapeutic options available for patients with advanced panNETs. Available systemic treatments for patients with well-differentiated pancreatic neuroendocrine tumors include somatostatin receptors ligands (SRLs), traditional cytotoxic chemotherapy regimens, peptide receptor radiotherapy (PRRT), and biologically targeted therapies, whereas patients with poorly differentiated neurodocrine carcinomas have more limited treatment options. Despite these advances, no clear guidelines exist to support the best sequence of treatments, not only the first-line, but also subsequent lines of therapy in patients with panNENs. Advances in molecular research and discovery of biomarkers for response allowing a more personalized approach to the multimodality therapy of panNENs are still limited. Understanding the impact of previous therapies on subsequent treatment efficacy and toxicity is also an ongoing research question. In the absence of definite predictive markers and paucity of comparative randomized trials, along with the heterogeneity of this patient population, systemic therapy selection in advanced non-resectable disease should be patient centered and often require evaluation within a multidisciplinary setting. The specific clinical context of the patient, with assessment of individual patient clinical and pathological features, somatostatin receptors imaging, and goals of treatment must all be considered when deciding on systemic therapy in the patient. Additional research is needed to address the gap in knowledge regarding optimal sequencing and timing of therapies and to provide individual care.


Assuntos
Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Humanos , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/uso terapêutico , Receptores de Somatostatina/fisiologia
3.
Sci Rep ; 11(1): 10962, 2021 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-34040115

RESUMO

Neuropeptides have been reported to regulate progenitor proliferation and neurogenesis in the central nervous system. However, these studies have typically been conducted using pharmacological agents in ex vivo preparations, and in vivo evidence for their developmental function is generally lacking. Recent scRNA-Seq studies have identified multiple neuropeptides and their receptors as being selectively expressed in neurogenic progenitors of the embryonic mouse and human retina. This includes Sstr2, whose ligand somatostatin is transiently expressed by immature retinal ganglion cells. By analyzing retinal explants treated with selective ligands that target these receptors, we found that Sstr2-dependent somatostatin signaling induces a modest, dose-dependent inhibition of photoreceptor generation, while correspondingly increasing the relative fraction of primary progenitor cells. These effects were confirmed by scRNA-Seq analysis of retinal explants but abolished in Sstr2-deficient retinas. Although no changes in the relative fraction of primary progenitors or photoreceptor precursors were observed in Sstr2-deficient retinas in vivo, scRNA-Seq analysis demonstrated accelerated differentiation of neurogenic progenitors. We conclude that, while Sstr2 signaling may act to negatively regulate retinal neurogenesis in combination with other retinal ganglion cell-derived secreted factors such as Shh, it is dispensable for normal retinal development.


Assuntos
Proteínas do Olho/fisiologia , Neurogênese/fisiologia , Neuropeptídeos/fisiologia , Receptores de Somatostatina/fisiologia , Retina/citologia , Animais , Relação Dose-Resposta a Droga , Proteínas do Olho/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Idade Gestacional , Humanos , Ligantes , Camundongos , Camundongos Knockout , Neuropeptídeos/agonistas , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/farmacologia , Fenótipo , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Receptores de Somatostatina/deficiência , Receptores de Somatostatina/efeitos dos fármacos , Retina/embriologia , Transdução de Sinais/fisiologia , Análise de Célula Única
4.
Front Endocrinol (Lausanne) ; 12: 652363, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33796080

RESUMO

Somatostatin (SST) and somatostatin receptors (SSTRs) play an important role in the brain and gastrointestinal (GI) system. SST is produced in various organs and cells, and the inhibitory function of somatostatin-containing cells is involved in a range of physiological functions and pathological modifications. The GI system is the largest endocrine organ for digestion and absorption, SST-endocrine cells and neurons in the GI system are a critical effecter to maintain homeostasis via SSTRs 1-5 and co-receptors, while SST-SSTRs are involved in chemo-sensory, mucus, and hormone secretion, motility, inflammation response, itch, and pain via the autocrine, paracrine, endocrine, and exoendocrine pathways. It is also a power inhibitor for tumor cell proliferation, severe inflammation, and post-operation complications, and is a first-line anti-cancer drug in clinical practice. This mini review focuses on the current function of producing SST endocrine cells and local neurons SST-SSTRs in the GI system, discusses new development prognostic markers, phosphate-specific antibodies, and molecular imaging emerging in diagnostics and therapy, and summarizes the mechanism of the SST family in basic research and clinical practice. Understanding of endocrines and neuroendocrines in SST-SSTRs in GI will provide an insight into advanced medicine in basic and clinical research.


Assuntos
Trato Gastrointestinal/fisiologia , Receptores de Somatostatina/fisiologia , Somatostatina/fisiologia , Animais , Antineoplásicos/farmacologia , Comunicação Celular/efeitos dos fármacos , Proliferação de Células , Modelos Animais de Doenças , Sistema Nervoso Entérico/fisiologia , Homeostase , Humanos , Inflamação , Ligantes , Neurônios/metabolismo , Sistema Nervoso Parassimpático/fisiologia , Prognóstico , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Células Secretoras de Somatostatina/metabolismo , Sistema Nervoso Simpático/fisiologia
5.
J Neurosci ; 41(17): 3932-3943, 2021 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-33741721

RESUMO

The Abelson-helper integration site 1 (AHI1) gene encodes for a ciliary transition zone localizing protein that when mutated causes the human ciliopathy, Joubert syndrome. We prepared and examined neuronal cultures derived from male and female embryonic Ahi1+/+ and Ahi1-/- mice (littermates) and found that the distribution of ciliary melanin-concentrating hormone receptor-1 (MchR1) was significantly reduced in Ahi1-/- neurons; however, the total and surface expression of MchR1 on Ahi1-/- neurons was similar to controls (Ahi1+/+). This indicates that a pathway for MchR1 trafficking to the surface plasma membrane is intact, but the process of targeting MchR1 into cilia is impaired in Ahi1-deficient mouse neurons, indicating a role for Ahi1 in localizing MchR1 to the cilium. Mouse Ahi1-/- neurons that fail to accumulate MchR1 in the ciliary membrane have significant decreases in two downstream MchR1 signaling pathways [cAMP and extracellular signal-regulated kinase (Erk)] on MCH stimulation. These results suggest that the ciliary localization of MchR1 is necessary and critical for MchR1 signaling, with Ahi1 participating in regulating MchR1 localization to cilia, and further supporting cilia as critical signaling centers in neurons.SIGNIFICANCE STATEMENT Our work here demonstrates that neuronal primary cilia are powerful and focused signaling centers for the G-protein-coupled receptor (GPCR), melanin-concentrating hormone receptor-1 (MCHR1), with a role for the ciliary transition zone protein, Abelson-helper integration site 1 (AHI1), in mediating ciliary trafficking of MCHR1. Moreover, our manuscript further expands the repertoire of cilia functions on neurons, a cell type that has not received significant attention in the cilia field. Lastly, our work demonstrates the significant influence of ciliary GPCR signaling in the overall signaling of neurons.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Cílios/fisiologia , Neurônios/fisiologia , Receptores de Somatostatina/fisiologia , Transdução de Sinais/fisiologia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/fisiopatologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Membrana Celular/fisiologia , Cerebelo/anormalidades , Cerebelo/fisiopatologia , AMP Cíclico/metabolismo , Anormalidades do Olho/genética , Anormalidades do Olho/fisiopatologia , Feminino , Doenças Renais Císticas/genética , Doenças Renais Císticas/fisiopatologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Knockout , Gravidez , Receptores de Somatostatina/genética , Retina/anormalidades , Retina/fisiopatologia , Transdução de Sinais/genética
6.
Sci Rep ; 11(1): 4880, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33649390

RESUMO

An understanding of adipocyte responsiveness to G-protein-coupled receptor-(GPCR) derived signals must take into consideration the role of membrane microenvironments; that individual sub-populations of proteins may vary significantly across different regions of the cell, and that cell differentiation alters those microenvironments. 3T3-L1 pre-adipocytes undergo a dramatic phenotypic transformation during differentiation into adipocytes, requiring the development of a transient primary cilium. We demonstrate that melanin-concentrating hormone (MCH) receptor 1, a GPCR that stimulates appetite, translocates to the transient primary cilium during early 3T3-L1 cell adipogenesis. Furthermore, we used RNA-Seq to investigate whether MCH signaling is influenced by its receptor localization and whether MCH can influence the transcriptome of early adipocyte development. We found that MCH signaling is sensitive to receptor localization to cilia, and this alters the adipogenic transcriptional program. Also, novel MCH signaling pathways in 3T3-L1 cells are identified, including those for circadian rhythm, the inflammatory response, and ciliary biogenesis. The presence of active MCH-signaling pathways in pre-adipocytes and the discovery that these pathways intersect with the early adipogenic program, among other newly-identified signaling pathways, suggests that the use of MCH receptor 1 antagonists for clinical interventions may have unintended consequences on adipose tissue development.


Assuntos
Adipócitos , Adipogenia , Cílios/metabolismo , Receptores de Somatostatina/fisiologia , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Diferenciação Celular , Microambiente Celular , Camundongos , Transcriptoma
7.
Cancer ; 127(3): 345-353, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33270905

RESUMO

Pancreatic neuroendocrine neoplasms (panNENs) are the second most common epithelial tumors of the pancreas. Despite improvements in prognostic grading and staging systems, it remains a challenge to predict the clinical behavior of panNENs and the response to specific therapies given the high degree of heterogeneity of these tumors. Most panNENs are nonfunctional and present as advanced disease. However, systemic therapies provide modest benefits. Therefore, there is a need for predictive biomarkers to develop personalized treatment and to advance new drug development. The somatostatin receptors remain the only clinically established prognostic and predictive biomarkers in panNENs. Oncogenic drivers are at a very low frequency. Commonly mutated genes in panNENs include MEN1, chromatin remodeling genes (DAXX and ATRX), and mammalian target of rapamycin pathway genes. In contrast, poorly differentiated neuroendocrine carcinomas (panNECs), which carry a very poor prognosis, have distinctive mutations in certain genes (eg, RB1 and p53). Ongoing research to integrate epigenomics will provide tremendous opportunities to improve current understanding of the clinical heterogeneity of pancreatic neuroendocrine tumors and provide invaluable insight into the biology of these tumors, new drug development, and establishing personalized therapies.


Assuntos
Tumores Neuroendócrinos/genética , Neoplasias Pancreáticas/genética , Instabilidade Cromossômica , Reparo do DNA , Epigênese Genética , Humanos , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/imunologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/imunologia , Receptores de Somatostatina/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia
8.
Microscopy (Oxf) ; 69(5): 277-285, 2020 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-32627821

RESUMO

G-protein-coupled receptors (GPCRs) comprise the largest and most diverse cell surface receptor family, with more than 800 known GPCRs identified in the human genome. Binding of an extracellular cue to a GPCR results in intracellular G protein activation, after which a sequence of events, can be amplified and optimized by selective binding partners and downstream effectors in spatially discrete cellular environments. Because GPCRs are widely expressed in the body, they help to regulate an incredible range of physiological processes from sensation to growth to hormone responses. Indeed, it is estimated that ∼ 30% of all clinically approved drugs act by binding to GPCRs. The primary cilium is a sensory organelle composed of a microtubule axoneme that extends from the basal body. The ciliary membrane is highly enriched in specific signaling components, allowing the primary cilium to efficiently convey signaling cascades in a highly ordered microenvironment. Recent data demonstrated that a limited number of non-olfactory GPCRs, including somatostatin receptor 3 and melanin-concentrating hormone receptor 1 (MCHR1), are selectively localized to cilia on several mammalian cell types including neuronal cells. Utilizing cilia-specific cell biological and molecular biological approaches, evidence has accumulated to support the biological importance of ciliary GPCR signaling followed by cilia structural changes. Thus, cilia are now considered a unique sensory platform for integration of GPCR signaling toward juxtaposed cytoplasmic structures. Herein, we review ciliary GPCRs and focus on a novel role of MCHR1 in ciliary length control that will impact ciliary signaling capacity and neuronal function.


Assuntos
Ciclo Celular/fisiologia , Cílios/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Somatostatina/fisiologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Humanos , Proteínas de Membrana/fisiologia , Modelos Biológicos , Neurônios
9.
CNS Neurol Disord Drug Targets ; 18(10): 791-797, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31686636

RESUMO

BACKGROUND: Our previous study has indicated that somatostatin potently inhibits neuropathic pain through the activation of its type 2 receptor (SSTR2) in mouse dorsal root ganglion and spinal cord. However, the underlying mechanism of this activation has not been elucidated clearly. OBJECTIVE: The aim of this study is to perform the pharmacological studies on the basis of sciatic nerve-pinch mice model and explore the underlying mechanism involving SSTR2. METHODS: On the basis of a sciatic nerve-pinch injury model, we aimed at comparing the painful behavior and dorsal root ganglion neurons neurochemical changes after the SSTR2 antibody (anti- SSTR2;5µl,1µg/ml) administration in the mouse. RESULTS: After pinch nerve injury, we found that the mechanical hyperalgesia and severely painful behavior (autotomy) were detected after the application of SSTR2 antibody (anti-SSTR2; 5µl, 1µg/ml) on the pinch-injured nerve. The up-regulated phosphorylated ERK (p-ERK) expression and the apoptotic marker (i.e., Bax) were significantly decreased in DRGs after anti-SSTR2 treatment. CONCLUSION: The current data suggested that inhibitory changes in proteins from the apoptotic pathway in anti-SSTR2-treated groups might be taking place to overcome the protein deficits caused by SSTR2 antibody and supported the new therapeutic intervention with SSTR2 antagonist for neuronal degeneration following nerve injury.


Assuntos
Anticorpos/imunologia , Gânglios Espinais/fisiopatologia , Hiperalgesia/prevenção & controle , Hiperalgesia/fisiopatologia , Receptores de Somatostatina/imunologia , Receptores de Somatostatina/fisiologia , Animais , MAP Quinases Reguladas por Sinal Extracelular/biossíntese , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Hiperalgesia/psicologia , Masculino , Camundongos , Nervo Isquiático/lesões , Proteína X Associada a bcl-2/biossíntese
10.
Neurochem Int ; 129: 104518, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31381945

RESUMO

In the present study, we describe the status of somatostatin receptor 2 and 5 (SSTR2 and SSTR5) as well as cannabinoid type 1 receptor (CB1R) in Huntingtin (Htt) knock-in striatal neuronal cells. In mutant Htt (mHtt) knock-in (STHdhQ111/111) and wild type (STHdhQ7/7) striatal neuronal cells, SSTRs and CB1R exhibit prominent cytoplasmic expression and respond to agonist in a receptor specific manner. In response to quinolinic acid (QUIN)-induced toxicity, STHdhQ111/111 cells are more vulnerable and display suppressed cell survival signaling pathways. Receptor-specific agonists protect cells from QUIN-induced toxicity and activate ERK1/2 in both STHdh cells. Co-activation of SSTRs and CB1R resulted in loss of protective effects, delayed ERK1/2 phosphorylation and altered receptor complex composition. These results provide firsthand evidence in support of the protective role of SSTRs in STHdh cells and the possible crosstalk between SSTRs and CB1R in the modulation of excitotoxicity in Huntington's disease.


Assuntos
Corpo Estriado/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurotoxinas/toxicidade , Ácido Quinolínico/toxicidade , Receptor Cross-Talk , Receptor CB1 de Canabinoide/fisiologia , Receptores de Somatostatina/fisiologia , Animais , Linhagem Celular Transformada , Corpo Estriado/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Introdução de Genes , Genes Reporter , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Neurônios/metabolismo , Neurotoxinas/antagonistas & inibidores , Fosforilação , Processamento de Proteína Pós-Traducional , Ácido Quinolínico/antagonistas & inibidores , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/biossíntese , Receptor CB1 de Canabinoide/genética , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/biossíntese , Receptores de Somatostatina/genética
11.
Geroscience ; 41(5): 631-641, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30903571

RESUMO

The inhibitory neuropeptide somatostatin regulates several functions in the nervous system including memory. Its concentrations decrease by age leading to functional alterations, but there are little known about the receptorial mechanism. We discovered that somatostatin receptor 4 (sst4) mediates analgesic, anti-depressant, and anti-inflammatory effects without endocrine actions, and it is a unique target for drug development. We investigated the exploratory and locomotor activities and learning and memory functions of male and female sst4gene-deficient mice compared with their wild-types (WT) at ages of 3, 12, 17 months in the Y-maze test, open field test (OFT), radial-arm maze (RAM) test and novel object recognition (NOR) test. Young sst4 gene-deficient females visited, repeated, and missed significantly less arms than the WTs in the RAM; males showed decreased exploration in the NOR. Young mice moved significantly more, spend longer time in OFT center, and visited more arms in the Y-maze than older ones. Young WT females spend significantly longer time in the OFT center, visited, missed and repeated more arms of the RAM than males. Old males found more rewards than females. Young males explored longer the novel object than young females and older males in the NOR; the recognition index was smaller in females. We conclude that aging and sex are important factors of behavioral parameters that should be focused on in such studies. Sst4 is likely to influence locomotion and exploratory behavior only in young mice, but not during normal aging, which is a beneficial feature of a good drug target focusing on the elderly.


Assuntos
Envelhecimento/fisiologia , Comportamento Exploratório/fisiologia , Locomoção/fisiologia , Receptores de Somatostatina/fisiologia , Envelhecimento/genética , Animais , Feminino , Aprendizagem/fisiologia , Masculino , Aprendizagem em Labirinto , Memória de Curto Prazo/fisiologia , Camundongos Knockout , Receptores de Somatostatina/genética , Fatores Sexuais , Memória Espacial/fisiologia
12.
Curr Radiopharm ; 12(2): 98-106, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30727940

RESUMO

BACKGROUND: Pancreatic Neuroendocrine Tumors (P-NETs) are a challenge in terms of both diagnosis and therapy; morphological studies need to be frequently implemented with nonstandard techniques such as Endoscopic Ultrasounds, Dynamic CT, and functional Magnetic Resonance. DISCUSSION: The role of nuclear medicine, being scarcely sensitive F-18 Fluorodeoxyglucose, is mainly based on the over-expression of Somatostatin Receptors (SSTR) on neuroendocrine tumor cells surface. Therefore, SSTR can be used as a target for both diagnosis, using radiotracers labeled with gamma or positron emitters, and therapy. SSTRs subtypes are capable of homo and heterodimerization in specific combinations that alter both the response to ligand activation and receptor internalization. CONCLUSION: Although agonists usually provide efficient internalization, also somatostatin antagonists (SS-ANTs) could be used for imaging and therapy. Peptide Receptor Radionuclide Therapy (PRRT) represents the most successful option for targeted therapy. The theranostic model based on SSTR does not work in insulinoma, in which different radiotracers such as F-18 FluoroDOPA or tracers for the glucagon-like peptide-1 receptor have to be preferred.


Assuntos
Tumores Neuroendócrinos/diagnóstico por imagem , Medicina Nuclear/métodos , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Animais , Di-Hidroxifenilalanina/análogos & derivados , Di-Hidroxifenilalanina/farmacologia , Dimerização , Ácido Edético/análogos & derivados , Ácido Edético/farmacologia , Fluordesoxiglucose F18/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/fisiologia , Humanos , Radioisótopos de Índio , Insulinoma/diagnóstico por imagem , Ligantes , Camundongos , Tumores Neuroendócrinos/fisiopatologia , Octreotida/uso terapêutico , Neoplasias Pancreáticas/fisiopatologia , Ácido Pentético/farmacologia , Cintilografia , Compostos Radiofarmacêuticos , Receptores de Somatostatina/fisiologia
13.
Neurosci Res ; 149: 38-49, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30685491

RESUMO

Somatostatin reduces neuronal excitability via somatostatin receptors (Sst1-Sst5) and inhibits seizure activity. However, the expression status of the Sst subtypes in epileptic mice and their role in the antiepileptic effects of somatostatin remain unclear. Here, we show that the Sst subtypes are regulated differently by epileptic neuronal activity in mice. Systemic kainate injection rapidly and transiently elevated the Sst2 and Sst3 mRNA and reduced Sst1 and Sst4 mRNA in the hippocampus; however, among all the subtypes, only Sst2 mRNA was increased in the excitatory neurons of the basolateral amygdala, accompanied by a decrease in the level of Sst2 protein. Following kainate administration, recovery from seizure was delayed by reduced expression of Sst2 in the basolateral amygdala, but not in the dentate gyrus of the hippocampus; higher expression levels of Bdnf, a neuronal activity marker, were observed in both conditions. These results suggest that Sst2 contributes to seizure termination by feedback inhibition in the amygdala. This could be a potential therapeutic target for acute seizures.


Assuntos
Hipocampo/metabolismo , Receptores de Somatostatina/metabolismo , Receptores de Somatostatina/fisiologia , Convulsões/metabolismo , Animais , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ácido Caínico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/fisiologia , Convulsões/induzido quimicamente
14.
Pharmacol Rev ; 70(4): 763-835, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30232095

RESUMO

Somatostatin, also known as somatotropin-release inhibitory factor, is a cyclopeptide that exerts potent inhibitory actions on hormone secretion and neuronal excitability. Its physiologic functions are mediated by five G protein-coupled receptors (GPCRs) called somatostatin receptor (SST)1-5. These five receptors share common structural features and signaling mechanisms but differ in their cellular and subcellular localization and mode of regulation. SST2 and SST5 receptors have evolved as primary targets for pharmacological treatment of pituitary adenomas and neuroendocrine tumors. In addition, SST2 is a prototypical GPCR for the development of peptide-based radiopharmaceuticals for diagnostic and therapeutic interventions. This review article summarizes findings published in the last 25 years on the physiology, pharmacology, and clinical applications related to SSTs. We also discuss potential future developments and propose a new nomenclature.


Assuntos
Receptores de Somatostatina/metabolismo , Animais , Regulação da Expressão Gênica , Humanos , Ligantes , Conformação Proteica , Transporte Proteico , Receptores de Somatostatina/química , Receptores de Somatostatina/genética , Receptores de Somatostatina/fisiologia , Transdução de Sinais , Terminologia como Assunto
15.
Genesis ; 56(8): e23217, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29806135

RESUMO

The neuropeptide, melanin concentrating hormone (MCH), and its G protein-coupled receptor, melanin concentrating hormone receptor 1 (Mchr1), are expressed centrally in adult rodents. MCH signaling has been implicated in diverse behaviors such as feeding, sleep, anxiety, as well as addiction and reward. While a model utilizing the Mchr1 promoter to drive constitutive expression of Cre recombinase (Mchr1-Cre) exists, there is a need for an inducible Mchr1-Cre to determine the roles for this signaling pathway in neural development and adult neuronal function. Here, we generated a BAC transgenic mouse where the Mchr1 promotor drives expression of tamoxifen inducible CreER recombinase. Many aspects of the Mchr1-Cre expression pattern are recapitulated by the Mchr1-CreER model, though there are also notable differences. Most strikingly, compared to the constitutive model, the new Mchr1-CreER model shows strong expression in adult animals in hypothalamic brain regions involved in feeding behavior but diminished expression in regions involved in reward, such as the nucleus accumbens. The inducible Mchr1-CreER allele will help reveal the potential for Mchr1 signaling to impact neural development and subsequent behavioral phenotypes, as well as contribute to the understanding of the MCH signaling pathway in terminally differentiated adult neurons and the diverse behaviors that it influences.


Assuntos
Hormônios Hipotalâmicos/fisiologia , Melaninas/fisiologia , Hormônios Hipofisários/fisiologia , Receptores de Somatostatina/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Integrases , Melaninas/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Animais , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Hormônios Hipofisários/metabolismo , Receptores de Somatostatina/metabolismo , Transdução de Sinais , Tamoxifeno
16.
Med Sci Monit ; 23: 1947-1955, 2017 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-28434012

RESUMO

BACKGROUND Thyrotropin-secreting pituitary adenomas (TSHomas) are a rare cause of hyperthyroidism. Somatostatin analogs have proved to be effective for inhibiting pituitary hormones secretion, working via interactions with somatostatin receptors (SSTRs). Moreover, antiproliferative activity of somatostatin analog is now demonstrated in several studies. In the present study, we determined the relative predominance of SSTR2 and SSTR5 subtypes among the different types of adenomas, especially TSHoma, and investigated the relationship between efficacy of short-term octreotide (OCT) treatment and SSTR expression. MATERIAL AND METHODS Serum hormone determinations and histological findings in resected tissue resulted in 5 diagnoses: 16 TSHomas, 8 acromegaly, 3 prolactinomas, 3 corticotropinomas, 4 clinically nonfunctioning adenomas (NFPAs), and 4 normal pituitary specimens. IHC was performed on formalin-fixed and paraffin-embedded tissue in tissue microarrays. RESULTS IHC of SSTR subtypes in the different cohorts showed SSTR2 staining intensity scores higher than SSTR5 in TSHoma, acromegaly and prolactinoma, whereas the expression of SSTR5 was stronger than SSTR2 in corticotropinoma and NFPA. SSTR2 and SSTR5 expressions were significantly higher in TSHoma than in other pituitary adenomas. OCT treatment for a median of 8.4 days (range: 3-18 days) and with a total median dose of 1.9 mg (range: 0.9-4.2 mg) showed a significant decrease of thyroid hormone levels (TSH [µIU/ml] in all patients. Patients with low SSTR5 expression presented a significantly higher TSH suppression rate (P values <0.05). CONCLUSIONS The present data confirm that somatostatin analogs should be considered as a medical alternative to surgical treatment, especially in patients with TSHoma, and short-term response to OCT therapy may be related to the expression of SSTR5.


Assuntos
Receptores de Somatostatina/genética , Adolescente , Adulto , Idoso , Feminino , Humanos , Hipertireoidismo/genética , Hipertireoidismo/metabolismo , Hipertireoidismo/patologia , Masculino , Pessoa de Meia-Idade , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , RNA Mensageiro/metabolismo , Receptores de Somatostatina/metabolismo , Receptores de Somatostatina/fisiologia , Somatostatina/genética , Somatostatina/metabolismo , Tireotropina/genética , Tireotropina/metabolismo
17.
Neuroscience ; 346: 320-336, 2017 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-28161436

RESUMO

The somatostatin 4 receptor (sst4) is widely expressed in stress-related brain areas (e.g. hippocampus, amygdala) and regulates the emotional behavior in acute situations. Since its importance in chronic stress-induced complex pathophysiological alterations is unknown, we investigated the involvement of sst4 in the responsiveness to chronic variable stress (CVS). Sstr4 gene-deficient (Sstr4-/-) mice and their wildtype counterparts (Sstr4+/+) were used to examine the behavioral and neuroendocrine alterations as well as chronic neuronal activity (FosB expression) changes in response to CVS. In Sstr4+/+ mice, there was no behavioral response to the applied CVS paradigm. In contrast, immobility time in the tail suspension test increased after the CVS in the knockouts. In the forced swim test, Sstr4-/- animals showed increased baseline immobility and then it decreased after the CVS. Light-dark box and open field test behaviors and sucrose preference did not respond to the stress in the knockouts. Adrenal weights increased and thymus weights decreased in both Sstr4+/+ and Sstr4-/- mice demonstrating the effect of chronic stress. The relative adrenal weight of stressed knockouts increased to a greater extent, while relative thymus and body weights decreased only in the Sstr4-/- mice. Basal plasma corticosterone concentrations did not change after the CVS in either genotype. FosB immunopositivity in the central and basolateral amygdaloid nuclei was enhanced in stressed knockouts, but not in wild types. This is the first evidence that sst4 activation is involved in the behavioral and neuroendocrine alterations induced by chronic stress with a crucial role of plastic changes in the amygdala.


Assuntos
Encéfalo/fisiopatologia , Sistemas Neurossecretores/fisiopatologia , Receptores de Somatostatina/fisiologia , Estresse Psicológico/fisiopatologia , Glândulas Suprarrenais/patologia , Tonsila do Cerebelo/fisiopatologia , Animais , Ansiedade/fisiopatologia , Comportamento Animal , Corticosterona/sangue , Hipocampo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Neurônios/metabolismo , Tamanho do Órgão , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Somatostatina/genética , Timo/patologia
18.
Neuropsychopharmacology ; 42(8): 1647-1656, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27986975

RESUMO

Altered brain somatostatin functions recently appeared as key elements for the pathogenesis of stress-related neuropsychiatric disorders. The hippocampus exerts an inhibitory feedback on stress but the mechanisms involved remain unclear. We investigated herein the role of hippocampal somatostatin receptor subtypes in both stress response and behavioral emotionality using C57BL/6, wild type and sst2 or sst4 knockout mice. Inhibitory effects of hippocampal infusions of somatostatin agonists on stress-induced hypothalamo-pituitary-adrenal axis (HPA) activity were tested by monitoring peripheral blood and local hippocampus corticosterone levels, the latter by using microdialysis. Anxiolytic and antidepressant-like effects were determined in the elevated-plus maze, open field, forced swimming, and stress-sensitive beam walking tests. Hippocampal injections of somatostatin analogs and sst2 or sst4, but not sst1 or sst3 receptor agonists produced rapid and sustained inhibition of HPA axis. sst2 agonists selectively produced anxiolytic-like behaviors whereas both sst2 and sst4 agonists had antidepressant-like effects. Consistent with these findings, high corticosterone levels and anxiety were found in sst2KO mice and depressive-like behaviors observed in both sst2KO and sst4KO strains. Both hippocampal sst2 and sst4 receptors selectively inhibit stress-induced HPA axis activation but mediate anxiolytic and antidepressive effects through distinct mechanisms. Such results are to be accounted for in development of pathway-specific somatostatin receptor agents in the treatment of hypercortisolism (Cushing's disease) and stress-related neuropsychiatric disorders.


Assuntos
Emoções/fisiologia , Hipocampo/metabolismo , Receptores de Somatostatina/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Ansiolíticos/farmacologia , Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Corticosterona/sangue , Corticosterona/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Octreotida/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Somatostatina/análogos & derivados , Somatostatina/farmacologia
19.
Eur Neuropsychopharmacol ; 26(11): 1826-1835, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27617778

RESUMO

In order to prepare the mother for the demands of pregnancy and lactation, the maternal brain is subjected to a number of adaptations. Maternal behaviors are regulated by complex neuronal interactions. Here, we show that the melanin concentrating hormone (MCH) system is an important regulator of maternal behaviors. First, we report that melanin concentrating hormone receptor 1 knockout (MCHR1 KO) mice display a disruption of maternal behavior. Early postpartum MCHR1 KO females exhibit poor nesting, deficits in pup retrieval and maternal aggression. In addition, ablation of MCH receptors results in decreased milk production and prolactin mRNA levels. Then we show that these results are in line with those obtained in wild type mice (WT) treated with the specific MCHR1 antagonist GW803430. Furthermore, following pups retrieval, MCHR1 KO mice display a lower level of Fos expression than WT mice in the ventral tegmental area, and nucleus accumbens. With the progression of the lactation period, however, the MCHR1 KO mice improve maternal care towards their pups. This is manifested by an increase in the pups׳ survival rate and the decrease in pups׳ retrieval time beyond the second day after parturition. In conclusion, we show that the MCH system plays a significant role in the initiation of maternal behavior. In this context, MCH may play a role in integrating information from multiple sources, and connecting brain reward, homeostatic and regulatory systems.


Assuntos
Comportamento Materno/efeitos dos fármacos , Receptores de Somatostatina/genética , Receptores de Somatostatina/fisiologia , Agressão , Animais , Animais Recém-Nascidos , Canibalismo/psicologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genes fos/efeitos dos fármacos , Lactação/genética , Camundongos , Camundongos Knockout , Comportamento de Nidação/efeitos dos fármacos , Prolactina/biossíntese , Pirimidinonas/farmacologia , Receptores de Somatostatina/antagonistas & inibidores , Tiofenos/farmacologia
20.
Alcohol Clin Exp Res ; 40(10): 2199-2207, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27579857

RESUMO

BACKGROUND: Reward and energy homeostasis are both regulated by a network of hypothalamic neuropeptide systems. The melanin-concentrating hormone (MCH) and its MCH-1 receptor (MCH1-R) modulate alcohol intake, but it remains unknown to what extent this reflects actions on energy balance or reward. Here, we evaluated the MCH1-R in regulation of caloric intake and motivation to consume alcohol in states of escalated consumption. METHODS: Rats had intermittent access (IA) to alcohol and were divided into high- and low-drinking groups. Food and alcohol consumption was assessed after administration of an MCH1-R antagonist, GW803430. Next, GW803430 was evaluated on alcohol self-administration in protracted abstinence induced by IA in high-drinking rats. Finally, the effect of GW803430 was assessed on alcohol self-administration in acute withdrawal in rats exposed to alcohol vapor. Gene expression of MCH and MCH1-R was measured in the hypothalamus and nucleus accumbens (NAc) in both acute and protracted abstinence. RESULTS: High-drinking IA rats consumed more calories from alcohol than chow and GW803430 decreased both chow and alcohol intake. In low-drinking rats, only food intake was affected. In protracted abstinence from IA, alcohol self-administration was significantly reduced by pretreatment with GW803430 and gene expression of both MCH and the MCH1-R were dysregulated in hypothalamus and NAc. In contrast, during acute withdrawal from vapor exposure, treatment with GW803430 did not affect alcohol self-administration, and no changes in MCH or MCH1-R gene expression were observed. CONCLUSIONS: Our data suggest a dual role of MCH and the MCH1-R in regulation of alcohol intake, possibly through mechanisms involving caloric intake and reward motivation. A selective suppression of alcohol self-administration during protracted abstinence by GW803430 was observed and accompanied by adaptations in gene expression of MCH and MCH1-R. Selective suppression of escalated consumption renders the MCH1-R an attractive target for treatment of alcohol use disorders.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Ingestão de Energia/fisiologia , Hormônios Hipotalâmicos/fisiologia , Melaninas/fisiologia , Motivação/fisiologia , Hormônios Hipofisários/fisiologia , Receptores de Somatostatina/fisiologia , Animais , Ingestão de Alimentos/fisiologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Hormônios Hipotalâmicos/biossíntese , Hipotálamo/metabolismo , Masculino , Melaninas/biossíntese , Núcleo Accumbens/metabolismo , Hormônios Hipofisários/biossíntese , Pirimidinonas/farmacologia , Ratos , Receptores de Somatostatina/antagonistas & inibidores , Autoadministração , Tiofenos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...