Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 434
Filtrar
1.
Elife ; 132024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39392867

RESUMEN

More than 20 recurrent missense gain-of-function (GOF) mutations have been identified in the sodium-activated potassium (KNa) channel gene KCNT1 in patients with severe developmental and epileptic encephalopathies (DEEs), most of which are resistant to current therapies. Defining the neuron types most vulnerable to KCNT1 GOF will advance our understanding of disease mechanisms and provide refined targets for precision therapy efforts. Here, we assessed the effects of heterozygous expression of a Kcnt1 GOF variant (Kcnt1Y777H) on KNa currents and neuronal physiology among cortical glutamatergic and GABAergic neurons in mice, including those expressing vasoactive intestinal polypeptide (VIP), somatostatin (SST), and parvalbumin (PV), to identify and model the pathogenic mechanisms of autosomal dominant KCNT1 GOF variants in DEEs. Although the Kcnt1Y777H variant had no effects on glutamatergic or VIP neuron function, it increased subthreshold KNa currents in both SST and PV neurons but with opposite effects on neuronal output; SST neurons became hypoexcitable with a higher rheobase current and lower action potential (AP) firing frequency, whereas PV neurons became hyperexcitable with a lower rheobase current and higher AP firing frequency. Further neurophysiological and computational modeling experiments showed that the differential effects of the Kcnt1Y777H variant on SST and PV neurons are not likely due to inherent differences in these neuron types, but to an increased persistent sodium current in PV, but not SST, neurons. The Kcnt1Y777H variant also increased excitatory input onto, and chemical and electrical synaptic connectivity between, SST neurons. Together, these data suggest differential pathogenic mechanisms, both direct and compensatory, contribute to disease phenotypes, and provide a salient example of how a pathogenic ion channel variant can cause opposite functional effects in closely related neuron subtypes due to interactions with other ionic conductances.


Asunto(s)
Neuronas GABAérgicas , Mutación con Ganancia de Función , Parvalbúminas , Somatostatina , Animales , Somatostatina/metabolismo , Somatostatina/genética , Ratones , Neuronas GABAérgicas/metabolismo , Parvalbúminas/metabolismo , Parvalbúminas/genética , Heterocigoto , Corteza Cerebral/metabolismo , Masculino , Potenciales de Acción , Femenino , Mutación Missense , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo
2.
Sci Rep ; 14(1): 13525, 2024 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866945

RESUMEN

The traditional nomenclature of enteroendocrine cells (EECs), established in 1977, applied the "one cell - one hormone" dogma, which distinguishes subpopulations based on the secretion of a specific hormone. These hormone-specific subpopulations included S cells for secretin (SCT), K cells for glucose-dependent insulinotropic polypeptide (GIP), N cells producing neurotensin (NTS), I cells producing cholecystokinin (CCK), D cells producing somatostatin (SST), and others. In the past 15 years, reinvestigations into murine and human organoid-derived EECs, however, strongly questioned this dogma and established that certain EECs coexpress multiple hormones. Using the Gut Cell Atlas, the largest available single-cell transcriptome dataset of human intestinal cells, this study consolidates that the original dogma is outdated not only for murine and human organoid-derived EECs, but also for primary human EECs, showing that the expression of certain hormones is not restricted to their designated cell type. Moreover, specific analyses into SCT-expressing cells reject the presence of any cell population that exhibits significantly elevated secretin expression compared to other cell populations, previously referred to as S cells. Instead, this investigation indicates that secretin production is realized jointly by other enteroendocrine subpopulations, validating corresponding observations in murine EECs also for human EECs. Furthermore, our findings corroborate that SCT expression peaks in mature EECs, in contrast, progenitor EECs exhibit markedly lower expression levels, supporting the hypothesis that SCT expression is a hallmark of EEC maturation.


Asunto(s)
Células Enteroendocrinas , Perfilación de la Expresión Génica , Secretina , Análisis de la Célula Individual , Humanos , Células Enteroendocrinas/metabolismo , Secretina/metabolismo , Secretina/genética , Análisis de la Célula Individual/métodos , Ratones , Animales , Transcriptoma , Diferenciación Celular , Organoides/metabolismo , Organoides/citología , Colecistoquinina/metabolismo , Colecistoquinina/genética , Somatostatina/metabolismo , Somatostatina/genética , Análisis de Expresión Génica de una Sola Célula
3.
FEBS Lett ; 598(16): 1996-2010, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38794908

RESUMEN

Neuronostatin suppresses the differentiation of white preadipocytes. However, the role of neuronostatin in brown adipose tissue remains elusive. Therefore, we investigated the impact of neuronostatin on the proliferation and differentiation of isolated rat brown preadipocytes. We report that neuronostatin and its receptor (GPR107) are synthesized in brown preadipocytes and brown adipose tissue. Furthermore, neuronostatin promotes the replication of brown preadipocytes via the AKT pathway. Notably, neuronostatin suppresses the expression of markers associated with brown adipogenesis (PGC-1α, PPARγ, PRDM16, and UCP1) and reduces cellular mitochondria content. Moreover, neuronostatin impedes the differentiation of preadipocytes by activating the JNK signaling pathway. These effects were not mimicked by somatostatin. Our results suggest that neuronostatin is involved in regulating brown adipogenesis.


Asunto(s)
Adipocitos Marrones , Diferenciación Celular , Proliferación Celular , Animales , Ratas , Adipocitos Marrones/metabolismo , Adipocitos Marrones/citología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/citología , Adipogénesis , Células Cultivadas , Masculino , Ratas Sprague-Dawley , Somatostatina/metabolismo , Somatostatina/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mitocondrias/metabolismo , Fragmentos de Péptidos
4.
Signal Transduct Target Ther ; 8(1): 186, 2023 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-37193687

RESUMEN

Seizures due to cortical dysplasia are notorious for their poor prognosis even with medications and surgery, likely due to the widespread seizure network. Previous studies have primarily focused on the disruption of dysplastic lesions, rather than remote regions such as the hippocampus. Here, we first quantified the epileptogenicity of the hippocampus in patients with late-stage cortical dysplasia. We further investigated the cellular substrates leading to the epileptic hippocampus, using multiscale tools including calcium imaging, optogenetics, immunohistochemistry and electrophysiology. For the first time, we revealed the role of hippocampal somatostatin-positive interneurons in cortical dysplasia-related seizures. Somatostatin-positive were recruited during cortical dysplasia-related seizures. Interestingly, optogenetic studies suggested that somatostatin-positive interneurons paradoxically facilitated seizure generalization. By contrast, parvalbumin-positive interneurons retained an inhibitory role as in controls. Electrophysiological recordings and immunohistochemical studies revealed glutamate-mediated excitatory transmission from somatostatin-positive interneurons in the dentate gyrus. Taken together, our study reveals a novel role of excitatory somatostatin-positive neurons in the seizure network and brings new insights into the cellular basis of cortical dysplasia.


Asunto(s)
Interneuronas , Convulsiones , Humanos , Interneuronas/metabolismo , Hipocampo , Somatostatina/genética , Somatostatina/metabolismo , Giro Dentado/metabolismo
5.
Development ; 150(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37254876

RESUMEN

RAS/MAPK gene dysfunction underlies various cancers and neurocognitive disorders. Although the roles of RAS/MAPK genes have been well studied in cancer, less is known about their function during neurodevelopment. There are many genes that work in concert to regulate RAS/MAPK signaling, suggesting that if common brain phenotypes could be discovered they could have a broad impact on the many other disorders caused by distinct RAS/MAPK genes. We assessed the cellular and molecular consequences of hyperactivating the RAS/MAPK pathway using two distinct genes in a cell type previously implicated in RAS/MAPK-mediated cognitive changes, cortical GABAergic interneurons. We uncovered some GABAergic core programs that are commonly altered in each of the mutants. Notably, hyperactive RAS/MAPK mutants bias developing cortical interneurons towards those that are somatostatin positive. The increase in somatostatin-positive interneurons could also be prevented by pharmacological inhibition of the core RAS/MAPK signaling pathway. Overall, these findings present new insights into how different RAS/MAPK mutations can converge on GABAergic interneurons, which may be important for other RAS/MAPK genes and related disorders.


Asunto(s)
Transducción de Señal , Somatostatina , Alelos , Somatostatina/genética , Somatostatina/metabolismo , Transducción de Señal/genética , Sistema de Señalización de MAP Quinasas/genética , Interneuronas/metabolismo , Neuronas GABAérgicas/metabolismo
6.
Neurotox Res ; 40(6): 1824-1837, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36378411

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by loss of neurons and synapses. The aim of this study was to investigate the effect of somatostatin analogue Vapreotide in an in vitro Alzheimer's model and its effects based on the relationship between somatostatinergic transmission and neurodegenerative functions. In this study, tau transfection was performed using the MAPT gene cloned into the pcDNA3.1 vector and transfection reagent into the SH-SY5Y cell line. In viability experiments using 10 µM Memantine as a positive control, it was observed that Vapreotide at 50 µM (p < 0.0001) and 100 µM (p < 0.05) had a protective effect on cell viability, 100 µM CYN154806 was found to decrease (p < 0.05) cell viability. It was determined that Vapreotide, decreased the expression levels (50 µM-p < 0.001; 100 µM-p < 0.001; 200 µM-p < 0.0001) and phosphorylation of Tau and p-Tau proteins by western blots. With the qRT-PCR method, it was found that Vapreotide, decreased the BAX/BCL2 (50 µM-p < 0.001; 100 µM-p < 0.01; 200 µM-p < 0.001) expression level and decreased the expression level (50 µM-p < 0.01; 100 µM-p < 0.01; 200 µM-p < 0.001) of the APOE4 gene, which constitutes a genetic risk for AD. This study demonstrates a potential therapeutic role for a somatostatin analogue Vapreotide in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Enfermedades Neurodegenerativas , Fármacos Neuroprotectores , Humanos , Enfermedad de Alzheimer/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Péptidos beta-Amiloides/metabolismo , Línea Celular Tumoral , Neuroblastoma/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Somatostatina/uso terapéutico , Transfección , Fosforilación
7.
Sci Rep ; 12(1): 16885, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207478

RESUMEN

Somatostatin, a growth hormone-release inhibiting peptide, exerts antiproliferative and antiangiogenic effects on tumor cells. However, the short half-life of somatostatin limits its application in human therapy, and long-acting somatostatin fusion protein is also limited by its severe terminal degradation. Therefore, oncolytic virus delivery system was introduced to express somatostatin fusion protein and the anti-tumor effects of both somatostatin and oncolytic virus were combined to destroy tumor tissues. Here, a vaccinia VG9/(SST-14)2-HSA recombinant was constructed by replacing somatostatin fusion gene into TK locus of attenuated VG9 strain via homologous recombination. Results showed that vaccinia VG9/(SST-14)2-HSA possessed a combined anti-tumor effect on sstr-positive tumor cells in vitro. In the tumor burden models, BALB/c mice with complete immunity are most suitable for evaluating tumor regression and immune activation. Complete tumor regression was observed in 3 out of 10 mice treated with vaccinia VG9/TK- or VG9/(SST-14)2-HSA, and the survival of all mice in both groups was significantly prolonged. Besides, vaccinia VG9/(SST-14)2-HSA is more effective in prolonging survival than VG9/TK-. Vaccinia VG9/(SST-14)2-HSA exerts a combined anti-tumor efficacy including the oncolytic ability provided by the virus and the anti-tumor effect contributed by (SST-14)2-HSA, which is expected to become a potent therapeutic agent for cancer treatment.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Vaccinia , Animales , Hormona del Crecimiento/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Viroterapia Oncolítica/métodos , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Virus Vaccinia
8.
Int J Mol Sci ; 23(15)2022 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-35955787

RESUMEN

Acromegaly is a chronic and systemic disease due to excessive growth hormone and insulin-like growth factor type I caused, in the vast majority of cases, by a GH-secreting pituitary adenoma. About 40% of these tumors have somatic mutations in the stimulatory G protein alpha-subunit 1 gene. The pathogenesis of the remaining tumors, however, is still not fully comprehended. Surgery is the first-line therapy for these tumors, and first-generation somatostatin receptor ligands (fg-SRL) are the most prescribed medications in patients who are not cured by surgery. MicroRNAs are small, non-coding RNAs that control the translation of many mRNAs, and are involved in the post-transcriptional regulation of gene expression. Differentially expressed miRNAs can explain differences in the pathogenesis of acromegaly and tumor resistance. In this review, we focus on the most validated miRNAs, which are mainly involved in acromegaly's tumorigenesis and fg-SRL resistance, as well as in circulating miRNAs in acromegaly.


Asunto(s)
Acromegalia , Adenoma , Hormona de Crecimiento Humana , MicroARNs , Acromegalia/genética , Adenoma/metabolismo , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , MicroARNs/genética , MicroARNs/uso terapéutico , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Somatostatina/genética , Somatostatina/uso terapéutico
9.
J Comp Neurol ; 530(17): 2977-2993, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35844047

RESUMEN

The olfactory placode (OP) of vertebrates generates several classes of migrating cells, including hypothalamic gonadotropin-releasing hormone (GnRH)-producing neurons, which play essential roles in the reproduction system. Previous studies using OP cell labeling have demonstrated that OP-derived non-GnRH cells enter the developing forebrain; however, their final fates and phenotypes are less well understood. In chick embryos, a subpopulation of migratory cells from the OP that is distinct from GnRH neurons transiently expresses somatostatin (SS). We postulated that these cells are destined to develop into brain neurons. In this study, we examined the expression pattern of SS mRNA in the olfactory-forebrain region during development, as well as the destination of OP-derived migratory cells, including SS mRNA-expressing cells. Utilizing the Tol2 genomic integration system to induce long-term fluorescent protein expression in OP cells, we found that OP-derived migratory cells labeled at embryonic day (E) 3 resided in the olfactory nerve and medial forebrain at E17-19. A subpopulation of green fluorescent protein (GFP)-labeled GnRH neurons that remained in the olfactory nerve was considered to comprise terminal nerve neurons. In the forebrain, GFP-labeled cells showed a distribution pattern similar to that of GnRH neurons. A large proportion of GFP-labeled cells expressed the mature neuronal marker NeuN. Among the GFP-labeled cells, the percentage of GnRH neurons was low, while the remaining GnRH-negative neurons either expressed SS mRNA, neuropeptide Y, or calbindin D-28k or did not express any of them. These results indicate that a diverse population of OP-derived neuronal cells, other than GnRH neurons, integrates into the chick medial forebrain.


Asunto(s)
Hormona Liberadora de Gonadotropina , Neuropéptido Y , Animales , Calbindinas/metabolismo , Movimiento Celular/fisiología , Embrión de Pollo , Pollos/metabolismo , Hormona Liberadora de Gonadotropina/genética , Proteínas Fluorescentes Verdes/metabolismo , Neuronas/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Prosencéfalo/metabolismo , ARN Mensajero/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
10.
Mol Biol Evol ; 39(4)2022 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-35383850

RESUMEN

Somatostatin and its related peptides (SSRPs) form an important family of hormones with diverse physiological roles. The ubiquitous presence of SSRPs in vertebrates and several invertebrate deuterostomes suggests an ancient origin of the SSRP signaling system. However, the existence of SSRP genes outside of deuterostomes has not been established, and the evolutionary history of this signaling system remains poorly understood. Our recent discovery of SSRP-like toxins (consomatins) in venomous marine cone snails (Conus) suggested the presence of a related signaling system in mollusks and potentially other protostomes. Here, we identify the molluscan SSRP-like signaling gene that gave rise to the consomatin family. Following recruitment into venom, consomatin genes experienced strong positive selection and repeated gene duplications resulting in the formation of a hyperdiverse family of venom peptides. Intriguingly, the largest number of consomatins was found in worm-hunting species (>400 sequences), indicating a homologous system in annelids, another large protostome phylum. Consistent with this, comprehensive sequence mining enabled the identification of SSRP-like sequences (and their corresponding orphan receptor) in annelids and several other protostome phyla. These results established the existence of SSRP-like peptides in many major branches of bilaterians and challenge the prevailing hypothesis that deuterostome SSRPs and protostome allatostatin-C are orthologous peptide families. Finally, having a large set of predator-prey SSRP sequences available, we show that although the cone snail's signaling SSRP-like genes are under purifying selection, the venom consomatin genes experience rapid directional selection to target receptors in a changing mix of prey.


Asunto(s)
Conotoxinas , Caracol Conus , Animales , Conotoxinas/genética , Caracol Conus/genética , Neuropéptidos , Péptidos/genética , Somatostatina/genética , Ponzoñas
11.
Dis Markers ; 2022: 4570290, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35242243

RESUMEN

BACKGROUND: DNA methylation is an important epigenetic modification in tumorigenesis, and similar epigenetic regulation mechanisms have been found in the gastrointestinal tract (GIT) cancers. Somatostatin (SST) has been confirmed to be expressed throughout the GIT. This study aimed to simultaneously explore the relationships between the SST methylation and the risks of three GIT cancers (esophageal cancer (EC), gastric cancer (GC), and colorectal cancer (CRC)) and to evaluate its diagnostic value. METHODS: Differentially methylated regions (DMRs) of the SST gene, including TSS200, 1stExon, and the gene body, were identified in GIT cancers by The Cancer Genome Atlas (TCGA) database analysis. Further analyses were conducted in tissue samples of EC (n = 50), GC (n = 99), and CRC (n = 80). The SST methylation was detected by bisulfite-sequencing PCR (BSP), and the SST expression was detected by quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR). RESULTS: In GIT cancers, DMR-related CpG islands were mainly located in the 1stExon. The methylation status of the SST 1stExon in the tumor tissues was significantly higher than that in the adjacent noncancerous tissues, and the methylation rates of the specific CpG sites were correlated with clinical phenotypes. The average methylation rate (AMR) of the SST 1stExon was negatively correlated with the SST gene expression in GC and CRC (both P < 0.001). For the diagnosis of GIT cancers, the combined detection of methylation at CpG sites +18 and +129 showed the highest area under the curve (AUC 0.698), with a sensitivity of 59.3% and a specificity of 72.8%. CONCLUSIONS: The site-specific hypermethylation of the SST 1stExon increases the risk of GIT cancers and might be a potential predictive marker for pan-GIT cancers.


Asunto(s)
Biomarcadores de Tumor/genética , Metilación de ADN , Epigénesis Genética , Neoplasias Gastrointestinales/genética , Medición de Riesgo , Somatostatina/genética , Islas de CpG/genética , Neoplasias Esofágicas/genética , Femenino , Humanos , Masculino , Análisis de Secuencia de ADN , Neoplasias Gástricas/genética , Transcriptoma
12.
J Reprod Dev ; 68(3): 190-197, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35249910

RESUMEN

Reproductive function is suppressed during lactation owing to the suckling-induced suppression of the kisspeptin gene (Kiss1) expression in the arcuate nucleus (ARC) and subsequent suppression of luteinizing hormone (LH) release. Our previous study revealed that somatostatin (SST) neurons mediate suckling-induced suppression of LH release via SST receptor 2 (SSTR2) in ovariectomized lactating rats during early lactation. This study examined whether central SST-SSTR2 signaling mediates the inhibition of ARC Kiss1 expression and LH release in lactating rats during late lactation and whether the inhibition of glutamatergic neurons, stimulators of LH release, is involved in the suppression of LH release mediated by central SST-SSTR2 signaling in lactating rats. A central injection of the SSTR2 antagonist CYN154806 (CYN) significantly increased ARC Kiss1 expression in lactating rats on day 16 of lactation. Dual in situ hybridization revealed that few ARC Kiss1-positive cells co-expressed Sstr2, and some of the ARC Slc17a6 (a glutamatergic neuronal marker)-positive cells co-expressed Sstr2. Furthermore, almost all ARC Kiss1-positive cells co-expressed Grin1, a subunit of N-methyl-D-aspartate (NMDA) receptors. The numbers of Slc17a6/Sstr2 double-labeled and Slc17a6 single-labeled cells were significantly lower in lactating dams than in non-lactating rats whose pups had been removed after parturition. A central injection of an NMDA antagonist reversed the CYN-induced increase in LH release in lactating rats. Overall, these results suggest that central SST-SSTR2 signaling, at least partly, mediates the suppression of ARC Kiss1 expression and LH release by inhibiting ARC glutamatergic interneurons in lactating rats.


Asunto(s)
Interneuronas , Kisspeptinas , Lactancia , Hormona Luteinizante , Receptores de Somatostatina , Somatostatina , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Interneuronas/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Lactancia/metabolismo , Hormona Luteinizante/metabolismo , N-Metilaspartato/metabolismo , Oligopéptidos/farmacología , Ratas , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
13.
Biochim Biophys Acta Mol Cell Res ; 1869(1): 119146, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34599984

RESUMEN

Gaba-ergic neurons are a diverse cell class with extensive influence over cortical processing, but their role in experience-dependent plasticity is not completely understood. Here we addressed the role of cortical somatostatin- (SOM-INs) and vasoactive intestinal polypeptide- (VIP-INs) containing interneurons in a Pavlovian conditioning where stimulation of the vibrissae is used as a conditioned stimulus and tail shock as unconditioned one. This procedure induces a plastic change observed as an enlargement of the cortical functional representation of vibrissae activated during conditioning. Using layer-targeted, cell-selective DREADD transductions, we examined the involvement of SOM-INs and VIP-INs activity in learning-related plastic changes. Under optical recordings, we injected DREADD-expressing vectors into layer IV (L4) barrels or layer II/III (L2/3) areas corresponding to the activated vibrissae. The activity of the interneurons was modulated during all conditioning sessions, and functional 2-deoxyglucose (2DG) maps were obtained 24 h after the last session. In mice with L4 but not L2/3 SOM-INs suppressed during conditioning, the plastic change of whisker representation was absent. The behavioral effect of conditioning was disturbed. Both L4 SOM-INs excitation and L2/3 VIP-INs inhibition during conditioning did not affect the plasticity or the conditioned response. We found the activity of L4 SOM-INs is indispensable in the formation of learning-induced plastic change. We propose that L4 SOM-INs may provide disinhibition by blocking L4 parvalbumin interneurons, allowing a flow of information into upper cortical layers during learning.


Asunto(s)
Interneuronas/fisiología , Aprendizaje , Inhibición Neural , Plasticidad Neuronal , Corteza Somatosensorial/fisiología , Animales , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Moduladores del Transporte de Membrana/farmacología , Ratones , Corteza Somatosensorial/citología , Somatostatina/genética , Somatostatina/metabolismo , Vibrisas/inervación , Vibrisas/fisiología
14.
Int J Cancer ; 150(3): 440-449, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34558659

RESUMEN

Methylation of host-cell deoxyribonucleic acid (DNA) has been proposed as a promising biomarker for triage of high-risk (hr) human papillomavirus (HPV) positive women at screening. Our study aims to validate recently identified host-cell DNA methylation markers for triage in an hrHPV-positive cohort derived from primary HPV-based cervical screening in The Netherlands. Methylation markers ASCL1, LHX8, ST6GALNAC5, GHSR, ZIC1 and SST were evaluated relative to the ACTB reference gene by multiplex quantitative methylation-specific PCR (qMSP) in clinician-collected cervical samples (n = 715) from hrHPV-positive women (age 29-60 years), who were enrolled in the Dutch IMPROVE screening trial (NTR5078). Primary clinical end-point was cervical intraepithelial neoplasia grade 3 (CIN3) or cancer (CIN3+). The single-marker and bi-marker methylation classifiers developed for CIN3 detection in a previous series of hrHPV-positive clinician-collected cervical samples were applied. The diagnostic accuracy was visualised using receiver operating characteristic (ROC) curves and assessed through area under the ROC curve (AUC). The performance of the methylation markers to detect CIN3+ was determined using the predefined threshold calibrated at 70% clinical specificity. Individual methylation makers showed good performance for CIN3+ detection, with highest AUC for ASCL1 (0.844) and LHX8 (0.830). Combined as a bi-marker panel with predefined threshold, ASCL1/LHX8 yielded a CIN3+ sensitivity of 76.9% (70/91; 95% CI 68.3-85.6%) at a specificity of 74.5% (465/624; 95% CI 71.1-77.9%). In conclusion, our study shows that the individual host-cell DNA methylation classifiers and the bi-marker panel ASCL1/LHX8 have clinical utility for the detection of CIN3+ in hrHPV-positive women invited for routine screening.


Asunto(s)
Metilación de ADN , Papillomaviridae/aislamiento & purificación , Triaje , Displasia del Cuello del Útero/genética , Neoplasias del Cuello Uterino/genética , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Estudios de Cohortes , Femenino , Humanos , Proteínas con Homeodominio LIM/genética , Persona de Mediana Edad , Receptores de Ghrelina/genética , Sialiltransferasas/genética , Somatostatina/genética , Factores de Transcripción/genética , Neoplasias del Cuello Uterino/virología , Displasia del Cuello del Útero/virología
15.
Cell Rep ; 37(3): 109837, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686328

RESUMEN

The selection of goal-directed behaviors is supported by neural circuits located within the frontal cortex. Frontal cortical afferents arise from multiple brain areas, yet the cell-type-specific targeting of these inputs is unclear. Here, we use monosynaptic retrograde rabies mapping to examine the distribution of afferent neurons targeting distinct classes of local inhibitory interneurons and excitatory projection neurons in mouse infralimbic frontal cortex. Interneurons expressing parvalbumin, somatostatin, or vasoactive intestinal peptide receive a large proportion of inputs from the hippocampus, while interneurons expressing neuron-derived neurotrophic factor receive a large proportion of inputs from thalamic regions. A similar dichotomy is present among the four different excitatory projection neurons. These results show a prominent bias among long-range hippocampal and thalamic afferent systems in their targeting to specific sets of frontal cortical neurons. Moreover, they suggest the presence of two distinct local microcircuits that control how different inputs govern frontal cortical information processing.


Asunto(s)
Lóbulo Frontal/fisiología , Hipocampo/fisiología , Interneuronas/fisiología , Sinapsis/fisiología , Tálamo/fisiología , Animales , Conducta Animal , Lóbulo Frontal/citología , Lóbulo Frontal/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Interneuronas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Inhibición Neural , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiología , Técnicas de Trazados de Vías Neuroanatómicas , Parvalbúminas/genética , Parvalbúminas/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Sinapsis/metabolismo , Tálamo/citología , Tálamo/metabolismo , Péptido Intestinal Vasoactivo/genética , Péptido Intestinal Vasoactivo/metabolismo
16.
Epigenomics ; 13(15): 1205-1219, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34318683

RESUMEN

Aim: Our study aimed at investigating how LINC01133 functions in gastric cancer (GC) progression. Materials & methods: Gain-of-function and loss-of-function approaches were applied to analyze the effects of LINC01133, microRNA-576-5p (miR-576-5p) and somatostatin (SST) on the biological behaviors of GC cells and in tumor-bearing nude mice. Results: GC tissues and cells showed low expression of LINC01133, and LINC01133 overexpression decreased malignant phenotypes of GC cells. Moreover, LINC01133 upregulated SST through binding to miR-576-5p. Overexpressing miR-576-5p or suppressing SST reversed the functions of LINC01133 in biological potentials of GC cells and tumor growth. Conclusion: LINC01133 overexpression may inhibit GC development by downregulation of miR-576-5p and upregulation of SST, which suggests new therapeutic targets for GC.


Asunto(s)
MicroARNs/genética , Interferencia de ARN , ARN Largo no Codificante/genética , Somatostatina/biosíntesis , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Animales , Línea Celular Tumoral , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Modelos Biológicos , Somatostatina/genética , Neoplasias Gástricas/patología , Transcriptoma
17.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070785

RESUMEN

Somatostatin is widely diffused in the central nervous system, where it participates to control the efficiency of synaptic transmission. This peptide mainly colocalizes with GABA, in inhibitory, GABA-containing interneurons from which it is actively released in a Ca2+ dependent manner upon application of depolarizing stimuli. Once released in the synaptic cleft, somatostatin acts locally, or it diffuses in the extracellular space through "volume diffusion", a mechanism(s) of distribution which mainly operates in the cerebrospinal fluid and that assures the progression of neuronal signalling from signal-secreting sender structures towards receptor-expressing targeted neurons located extrasynaptically, in a non-synaptic, inter-neuronal form of communication. Somatostatin controls the efficiency of central glutamate transmission by either modulating presynaptically the glutamate exocytosis or by metamodulating the activity of glutamate receptors colocalized and functionally coupled with somatostatin receptors in selected subpopulations of nerve terminals. Deciphering the role of somatostatin in the mechanisms of "volume diffusion" and in the "receptor-receptor interaction" unveils new perspectives in the central role of this fine tuner of synaptic strength, paving the road to new therapeutic approaches for the cure of central disorders.


Asunto(s)
Sistema Nervioso Central/metabolismo , Ácido Glutámico/metabolismo , Interneuronas/metabolismo , Neuronas/metabolismo , Receptores de Somatostatina/genética , Somatostatina/genética , Sinapsis/metabolismo , Animales , Calcio/metabolismo , Sistema Nervioso Central/citología , AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Humanos , Interneuronas/citología , Neuronas/citología , Potasio/metabolismo , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Transmisión Sináptica
18.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799501

RESUMEN

The growth hormone (GH)/insulin-like growth factor I (IGF-I) axis is involved in metabolic control. Malnutrition reduces IGF-I and modifies the thermogenic capacity of brown adipose tissue (BAT). Leptin has effects on the GH/IGF-I axis and the function of BAT, but its interaction with IGF-I and the mechanisms involved in the regulation of thermogenesis remains unknown. We studied the GH/IGF-I axis and activation of IGF-I-related signaling and metabolism related to BAT thermogenesis in chronic central leptin infused (L), pair-fed (PF), and control rats. Hypothalamic somatostatin mRNA levels were increased in PF and decreased in L, while pituitary GH mRNA was reduced in PF. Serum GH and IGF-I concentrations were decreased only in PF. In BAT, the association between suppressor of cytokine signaling 3 and the IGF-I receptor was reduced, and phosphorylation of the IGF-I receptor increased in the L group. Phosphorylation of Akt and cyclic AMP response element binding protein and glucose transporter 4 mRNA levels were increased in L and mRNA levels of uncoupling protein-1 (UCP-1) and enzymes involved in lipid anabolism reduced in PF. These results suggest that modifications in UCP-1 in BAT and changes in the GH/IGF-I axis induced by negative energy balance are dependent upon leptin levels.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Hormona del Crecimiento/genética , Factor I del Crecimiento Similar a la Insulina/genética , Leptina/farmacología , Termogénesis/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Metabolismo Energético/genética , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Hormona del Crecimiento/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Fosforilación/efectos de los fármacos , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Termogénesis/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
19.
Medicine (Baltimore) ; 100(4): e24435, 2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33530245

RESUMEN

ABSTRACT: Obstructive sleep apnea (OSA) is a common chronic disease and increases the risk of cardiovascular disease, metabolic and neuropsychiatric disorders, resulting in a considerable socioeconomic burden. This study aimed to identify potential key genes influence the mechanisms and consequences of OSA.Gene expression profiles related to OSA were obtained from Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) in subcutaneous adipose tissues from OSA compared with normal tissues were screened using R software, followed by gene ontology (GO) and pathway enrichment analyses. Subsequently, a protein-protein interaction (PPI) network for these DEGs was constructed by STRING, and key hub genes were extracted from the network with plugins in Cytoscape. The hub genes were further validated in another GEO dataset and assessed by receiver operating characteristic (ROC) analysis and Pearson correlation analysis.There were 373 DEGs in OSA samples in relative to normal controls, which were mainly associated with olfactory receptor activity and olfactory transduction. Upon analyses of the PPI network, GDNF, SLC2A2, PRL, and SST were identified as key hub genes. Decreased expression of the hub genes was association with OSA occurrence, and exhibited good performance in distinguishing OSA from normal samples based on ROC analysis. Besides, the Pearson method revealed a strong correlation between hub genes, which indicates that they may act in synergy, contributing to OSA and related disorders.This bioinformatics research identified 4 hub genes, including GDNF, SLC2A2, PRL, and SST which may be new potential biomarkers for OSA and related disorders.


Asunto(s)
Tamizaje Masivo/métodos , Análisis por Micromatrices/métodos , Apnea Obstructiva del Sueño/genética , Biomarcadores/análisis , Biología Computacional , Bases de Datos Genéticas , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Transportador de Glucosa de Tipo 2/genética , Humanos , Prolactina/genética , Mapas de Interacción de Proteínas , Curva ROC , Somatostatina/genética , Transcriptoma
20.
Surgery ; 169(1): 155-161, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32611516

RESUMEN

BACKGROUND: Patients with pancreatic neuroendocrine tumors often present with metastases, which reduce survival. Molecular features associated with pancreatic neuroendocrine tumor tumorigenesis have been reported, but mechanisms of metastasis remain incompletely understood. METHODS: RNA sequencing was performed on primary and metastatic pancreatic neuroendocrine tumors from 43 patients. Differentially expressed genes were identified, and quantitative polymerase chain reaction used to confirm expression differences. BON cells were transfected with short interfering RNAs and short hairpin RNAs to create knockdowns. Expression changes were confirmed by quantitative polymerase chain reaction, cell viability assessed, and protein levels evaluated by Western blot and immunofluorescence. RESULTS: Nodal and hepatic metastases had decreased expression of somatostatin compared with primary tumors (P = .003). Quantitative polymerase chain reaction in a validation cohort confirmed 5.3-fold lower somatostatin expression in hepatic metastases (P = .043) with no difference in somatostatin receptor, synaptophysin, or chromogranin A expression. Somatostatin knockdown in BON cells increased cell metabolic activity, viability, and growth. Somatostatin-knockdown cells had significantly higher levels of phosphorylated Akt protein and higher mTOR compared with controls. CONCLUSION: Pancreatic neuroendocrine tumor metastases have lower expression of somatostatin than primary tumors, and somatostatin knockdown increased growth in pancreatic neuroendocrine tumor cell lines. This was associated with increased activation of Akt, identifying this pathway as a potential mechanism by which loss of somatostatin expression promotes the metastatic phenotype.


Asunto(s)
Neoplasias Hepáticas/genética , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Somatostatina/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Hígado/patología , Hígado/cirugía , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/cirugía , Metástasis Linfática/patología , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/secundario , Tumores Neuroendocrinos/cirugía , Páncreas/patología , Páncreas/cirugía , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , RNA-Seq , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA