Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.417
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(41): e2400298121, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39361640

RESUMEN

Somatostatin receptors (SSTRs) exert critical biological functions such as negatively regulating hormone release and cell proliferation, making them popular targets for developing therapeutics to treat endocrine disorders, especially neuroendocrine tumors. Although several panagonists mimicking the endogenous ligand somatostatin are available, the development of more effective and safer somatostatinergic therapies is limited due to a lack of molecular understanding of the ligand recognition and regulation of divergent SSTR subtypes. Here, we report four cryoelectron microscopy structures of Gi-coupled SSTR1 and SSTR3 activated by distinct agonists, including the FDA-approved panagonist pasireotide as well as their selective small molecule agonists L-797591 and L-796778. Our structures reveal a conserved recognition pattern of pasireotide in SSTRs attributed to the binding with a conserved extended binding pocket, distinct from SST14, octreotide, and lanreotide. Together with mutagenesis analyses, our structures further reveal the dynamic feature of ligand binding pockets in SSTR1 and SSTR3 to accommodate divergent agonists, the key determinants of ligand selectivity lying across the orthosteric pocket of different SSTR subtypes, as well as the molecular mechanism underlying diversity and conservation of receptor activation. Our work provides a framework for rational design of subtype-selective SSTR ligands and may facilitate drug development efforts targeting SSTRs with improved therapeutic efficacy and reduced side effects.


Asunto(s)
Microscopía por Crioelectrón , Receptores de Somatostatina , Somatostatina , Receptores de Somatostatina/metabolismo , Receptores de Somatostatina/química , Receptores de Somatostatina/agonistas , Humanos , Ligandos , Somatostatina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/química , Sitios de Unión , Unión Proteica , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/metabolismo
2.
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
3.
Cereb Cortex ; 34(9)2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39344196

RESUMEN

Three major types of GABAergic interneurons, parvalbumin-, somatostatin-, and vasoactive intestinal peptide-expressing (PV, SOM, VIP) cells, play critical but distinct roles in the cortical microcircuitry. Their specific electrophysiology and connectivity shape their inhibitory functions. To study the network dynamics and signal processing specific to these cell types in the cerebral cortex, we developed a multi-layer model incorporating biologically realistic interneuron parameters from rodent somatosensory cortex. The model is fitted to in vivo data on cell-type-specific population firing rates. With a protocol of cell-type-specific stimulation, network responses when activating different neuron types are examined. The model reproduces the experimentally observed inhibitory effects of PV and SOM cells and disinhibitory effect of VIP cells on excitatory cells. We further create a version of the model incorporating cell-type-specific short-term synaptic plasticity (STP). While the ongoing activity with and without STP is similar, STP modulates the responses of Exc, SOM, and VIP cells to cell-type-specific stimulation, presumably by changing the dominant inhibitory pathways. With slight adjustments, the model also reproduces sensory responses of specific interneuron types recorded in vivo. Our model provides predictions on network dynamics involving cell-type-specific short-term plasticity and can serve to explore the computational roles of inhibitory interneurons in sensory functions.


Asunto(s)
Interneuronas , Modelos Neurológicos , Plasticidad Neuronal , Corteza Somatosensorial , Corteza Somatosensorial/fisiología , Corteza Somatosensorial/citología , Interneuronas/fisiología , Plasticidad Neuronal/fisiología , Animales , Péptido Intestinal Vasoactivo/metabolismo , Potenciales de Acción/fisiología , Parvalbúminas/metabolismo , Red Nerviosa/fisiología , Somatostatina/metabolismo , Ratas
4.
Int J Mol Sci ; 25(18)2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39337311

RESUMEN

Diabetes mellitus affects 537 million adults around the world. Adropin is expressed in different cell types. Our aim was to investigate the cellular localization in the endocrine pancreas and its effect on modulating pancreatic endocrine hormone release in streptozotocin (STZ)-induced diabetic rats. Adropin expression in the pancreas was investigated in normal and diabetic rats using immunohistochemistry and immunoelectron microscopy. Serum levels of insulin, glucagon pancreatic polypeptide (PP), and somatostatin were measured using a Luminex® χMAP (Magpix®) analyzer. Pancreatic endocrine hormone levels in INS-1 832/3 rat insulinoma cells, as well as pancreatic tissue fragments of normal and diabetic rats treated with different concentrations of adropin (10-6, 10-9, and 10-12 M), were measured using ELISA. Adropin was colocalized with cells producing either insulin, glucagon, or PP. Adropin treatment reduced the number of glucagon-secreting alpha cells and suppressed glucagon release from the pancreas. The serum levels of GLP-1 and amylin were significantly increased after treatment with adropin. Our study indicates a potential role of adropin in modulating glucagon secretion in animal models of diabetes mellitus.


Asunto(s)
Diabetes Mellitus Experimental , Glucagón , Insulina , Islotes Pancreáticos , Animales , Glucagón/metabolismo , Glucagón/sangre , Diabetes Mellitus Experimental/metabolismo , Ratas , Masculino , Islotes Pancreáticos/metabolismo , Insulina/metabolismo , Insulina/sangre , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/sangre , Células Secretoras de Glucagón/metabolismo , Somatostatina/metabolismo , Polipéptido Pancreático/metabolismo , Polipéptido Pancreático/sangre , Ratas Sprague-Dawley , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Proteínas Sanguíneas , Péptidos
5.
Theranostics ; 14(14): 5400-5412, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39310095

RESUMEN

Rationale: Positron Emission Tomography (PET) using the somatostatin receptor 2 (SSTR2)-antagonist satoreotide trizoxetan (68Ga-SSO120) is a novel, promising imaging modality for small-cell lung cancer (SCLC), which holds potential for theranostic applications. This study aims to correlate uptake in PET imaging with SSTR2 expression in immunohistochemistry (IHC) and to assess the prognostic value of 68Ga-SSO120 PET at initial staging of patients with SCLC. Methods: We analyzed patients who underwent 68Ga-SSO120 PET/CT during initial diagnostic workup of SCLC as part of institutional standard-of-care. SSTR2 expression in IHC was evaluated on a 4-level scale and correlated with normalized standardized uptake values and tumor-to-liver ratios (SUVmax and TLRpeak) in 68Ga-SSO120 PET on a lesion level. Highest lesion SUVmax/TLRpeak per patient, SSTR2 score in IHC, M status according to TNM classification, and other parameters were analyzed for association with overall survival (OS) and time to treatment failure (TTF) by univariate, multivariate (cut-off values were identified on data for best separation), and stratified Cox regression. Results: We included 54 patients (24 men/30 women, median age 65 years, 21 M0/33 M1 according to TNM classification). In 43 patients with available surplus tumor tissue samples, hottest lesion SUVmax/TLRpeak showed a significant correlation with the level of SSTR2-expression by tumor cells in IHC (Spearman's rho 0.86/0.81, both p < 0.001; ANOVA p < 0.001). High SSTR2 expression in IHC, 68Ga-SSO120 SUVmax and TLRpeak of the hottest lesion per patient, whole-body TLRmean, MTV, TLG, M status, and serum LDH showed a significant association with inferior TTF/OS in univariate analysis. In separate multivariate Cox regression (including sex, age, M stage, and LDH) higher hottest-lesion TLRpeak showed a significant association with shorter OS (HR = 0.26, 95%CI: 0.08-0.84, p = 0.02) and SSTR2 expression in IHC with significantly shorter TTF (HR = 0.24, 95%CI: 0.08-0.71, p = 0.001) and OS (HR = 0.22, 95%CI: 0.06-0.84, p = 0.03). In total, 12 patients (22.2%) showed low (< 1), 21 (38.9%) intermediate (≥ 1 but < 2), 14 (25.9%) high (≥ 2 but < 5), and 7 (13.0%) very high (≥ 5) whole-body mean TLRmean. Conclusion: In patients with SCLC, SSTR2 expression assessed by 68Ga-SSO120 PET and by IHC were closely correlated and associated with shorter survival. More than 75% of patients showed higher whole-body 68Ga-SSO120 tumor uptake than liver uptake and almost 40% high or very high uptake, possibly paving the way towards theranostic applications.


Asunto(s)
Inmunohistoquímica , Neoplasias Pulmonares , Tomografía Computarizada por Tomografía de Emisión de Positrones , Receptores de Somatostatina , Carcinoma Pulmonar de Células Pequeñas , Humanos , Femenino , Masculino , Receptores de Somatostatina/metabolismo , Anciano , Persona de Mediana Edad , Carcinoma Pulmonar de Células Pequeñas/diagnóstico por imagen , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Carcinoma Pulmonar de Células Pequeñas/mortalidad , Inmunohistoquímica/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/mortalidad , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Radioisótopos de Galio , Pronóstico , Anciano de 80 o más Años , Adulto , Estudios Retrospectivos , Análisis de Supervivencia , Radiofármacos , Somatostatina/metabolismo , Nanomedicina Teranóstica/métodos , Tomografía de Emisión de Positrones/métodos
6.
Cell Rep ; 43(9): 114763, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39288028

RESUMEN

Recent studies have found dramatic cell-type-specific responses to stimulus novelty, highlighting the importance of analyzing the cortical circuitry at this granularity to understand brain function. Although initial work characterized activity by cell type, the alterations in cortical circuitry due to interacting novelty effects remain unclear. We investigated circuit mechanisms underlying the observed neural dynamics in response to novel stimuli using a large-scale public dataset of electrophysiological recordings in behaving mice and a population network model. The model was constrained by multi-patch synaptic physiology and electron microscopy data. We found generally weaker connections under novel stimuli, with shifts in the balance between somatostatin (SST) and vasoactive intestinal polypeptide (VIP) populations and increased excitatory influences on parvalbumin (PV) and SST populations. These findings systematically characterize how cortical circuits adapt to stimulus novelty.


Asunto(s)
Somatostatina , Animales , Ratones , Somatostatina/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Red Nerviosa/fisiología , Neuronas/fisiología , Neuronas/metabolismo , Parvalbúminas/metabolismo , Modelos Neurológicos , Corteza Cerebral/fisiología , Corteza Cerebral/metabolismo , Sinapsis/fisiología , Sinapsis/metabolismo
7.
Nature ; 633(8029): 398-406, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39198646

RESUMEN

The brain functions as a prediction machine, utilizing an internal model of the world to anticipate sensations and the outcomes of our actions. Discrepancies between expected and actual events, referred to as prediction errors, are leveraged to update the internal model and guide our attention towards unexpected events1-10. Despite the importance of prediction-error signals for various neural computations across the brain, surprisingly little is known about the neural circuit mechanisms responsible for their implementation. Here we describe a thalamocortical disinhibitory circuit that is required for generating sensory prediction-error signals in mouse primary visual cortex (V1). We show that violating animals' predictions by an unexpected visual stimulus preferentially boosts responses of the layer 2/3 V1 neurons that are most selective for that stimulus. Prediction errors specifically amplify the unexpected visual input, rather than representing non-specific surprise or difference signals about how the visual input deviates from the animal's predictions. This selective amplification is implemented by a cooperative mechanism requiring thalamic input from the pulvinar and cortical vasoactive-intestinal-peptide-expressing (VIP) inhibitory interneurons. In response to prediction errors, VIP neurons inhibit a specific subpopulation of somatostatin-expressing inhibitory interneurons that gate excitatory pulvinar input to V1, resulting in specific pulvinar-driven response amplification of the most stimulus-selective neurons in V1. Therefore, the brain prioritizes unpredicted sensory information by selectively increasing the salience of unpredicted sensory features through the synergistic interaction of thalamic input and neocortical disinhibitory circuits.


Asunto(s)
Corteza Visual Primaria , Tálamo , Vías Visuales , Animales , Femenino , Masculino , Ratones , Interneuronas/fisiología , Ratones Endogámicos C57BL , Modelos Neurológicos , Inhibición Neural/fisiología , Estimulación Luminosa , Corteza Visual Primaria/fisiología , Corteza Visual Primaria/citología , Pulvinar/fisiología , Pulvinar/citología , Somatostatina/metabolismo , Tálamo/fisiología , Tálamo/citología , Péptido Intestinal Vasoactivo/metabolismo , Vías Visuales/citología , Vías Visuales/fisiología , Neuronas/fisiología
8.
Int J Mol Sci ; 25(16)2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39201352

RESUMEN

Resistance to first-generation somatostatin receptor ligand (fgSRL) treatment in acromegaly is common, making the identification of biomarkers that predict fgSRL response a desired goal. We conducted a retrospective analysis on 21 patients with acromegaly who underwent surgery and subsequent pharmacological treatment. Through immunohistochemistry (IHC), we assessed the expression of the somatostatin receptor subtypes SSTR2 and SSTR5, E-Cadherin, and cytokeratin granulation pattern (sparsely or densely). Patients were divided into responders and non-responders based on their biochemical response to fgSRL and/or the newer agent, Pasireotide, or the GH-blocker, Pegvisomant. Patients resistant to fgSRL (n = 12) exhibited lower SSTR2 and E-Cadherin expressions. Sparsely granulated tumors were more frequent in the non-responder group. SSTR2 (p = 0.024, r = 0.49) and E-Cadherin (p = 0.009, r = 0.64) positively correlated with the Insulin-like Growth Factor 1 (IGF-1) decrease after fgSRL, while SSTR5 (p = 0.107, r = -0.37) showed a trend towards negative correlation. SSTR5 positivity seemed to be associated with Pasireotide response, albeit the number of treated patients was too low (n = 4). No IHC markers correlated with Pegvisomant response. Our findings suggest that densely granulated tumors, with positive SSTR2 and E-Cadherin seem to be associated with favorable fgSRL responses. The strongest predictive value of the studied markers was found for E-Cadherin, which seems to surpass even SSTR2.


Asunto(s)
Acromegalia , Cadherinas , Receptores de Somatostatina , Humanos , Receptores de Somatostatina/metabolismo , Acromegalia/tratamiento farmacológico , Acromegalia/metabolismo , Femenino , Masculino , Cadherinas/metabolismo , Persona de Mediana Edad , Adulto , Estudios Retrospectivos , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Somatostatina/metabolismo , Anciano , Factor I del Crecimiento Similar a la Insulina/metabolismo , Resultado del Tratamiento , Hormona de Crecimiento Humana/análogos & derivados
9.
Digestion ; 105(5): 400-410, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39033748

RESUMEN

INTRODUCTION: Inflammation in ulcerative colitis (UC) originates in the colorectal mucosa. Transcriptome sequencing analysis of the colorectal mucosa allows the identification of potential neuropeptides related to local neurotransmission. The intestinal mucus lining the surface of the mucosa may harbor biomarkers of mucosal inflammation; however, this has not been sufficiently investigated, given the difficulty in obtaining human samples. We previously reported the feasibility of obtaining mucin samples for proteomic analysis by brushing during colonoscopy. Herein, we aimed to investigate the composition of the intestinal mucus and detect neuropeptides characteristic of UC. METHODS: Mucus and mucosal samples were collected from patients with UC from the colorectum in areas showing remission or active UC using a brush catheter and biopsy forceps during colonoscopy. RNA sequencing findings of mucus samples of active and remission areas were compared. RNA and protein expression levels of significantly upregulated neuropeptides were analyzed. RESULTS: Of the neuropeptides associated with UC, somatostatin (SST) was significantly elevated in areas of remission, according to RNA sequencing results of mucus and expression levels in mucus RNA and proteins. Conversely, SST expression in the mucosa was increased in the inflamed areas. Flow cytometry revealed that the fluorescence intensity of SST-positive cells in the remission zone was higher in the mucus than in the mucosa. CONCLUSION: SST expression in the mucus is considered to be an important factor associated with UC activity.


Asunto(s)
Colitis Ulcerosa , Mucosa Intestinal , Neuropéptidos , Análisis de Secuencia de ARN , Humanos , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/genética , Colitis Ulcerosa/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Femenino , Persona de Mediana Edad , Neuropéptidos/análisis , Neuropéptidos/metabolismo , Neuropéptidos/genética , Adulto , Somatostatina/análisis , Somatostatina/metabolismo , Colonoscopía , Anciano , Moco/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo
10.
J Nucl Med ; 65(8): 1279-1285, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38991751

RESUMEN

Myocardial somatostatin PET uptake is observed not only in most patients with acute myocarditis (AM) but also in some oncology patients referred for routine somatostatin PET. This raises concerns about the specificity of somatostatin PET for detecting myocarditis. The current study aims to identify factors associated with the detection of myocardial uptake on somatostatin PET scans recorded for oncology indications and differential PET criteria that characterize myocardial uptake in AM patients. Methods: We analyzed factors associated with the detection of myocardial [68Ga]Ga-DOTATOC uptake in 508 [68Ga]Ga-DOTATOC PET scans from 178 patients, performed for confirmed or suspected oncologic disease (Onc-PET) and PET criteria that could differentiate myocardial [68Ga]Ga-DOTATOC uptake in 31 patients with MRI-ascertained AM (AM-PET) from that in the Onc-PET group. Results: Significant myocardial uptake was detected in 137 (26.9%) Onc-PET scans and was independently associated with somatostatin analog treatment (exp(ß), 0.805; 95% CI, 0.728-0.890; P < 0.001) and age (exp(ß), 1.005; 95% CI, 1.001-1.009; P = 0.012). A comparable model was selected for predicting the myocardial-to-blood SUVmax ratio using somatostatin analog treatment (P < 0.001) and history of coronary artery disease (P = 0.022). Myocardial uptake was detected in 12.9% (25/193) of Onc-PET scans from patients treated with somatostatin analogs but in 43.4% (59/136) of untreated patients over the median age of 64 y. Myocardial uptake was apparent in all 31 AM-PET scans, with volume and intensity of uptake dramatically higher than in the 137 Onc-PET scans showing myocardial uptake. A myocardial-to-blood SUVmax ratio threshold of 2.20 provided a sensitivity of 87% (27/31) and a specificity of 88% (44/50) for differentiating myocardial uptake between the AM-PET group and an Onc-PET group restricted to patients with clinical characteristics comparable to those of patients in the AM-PET group (≤64 y of age, no coronary artery disease history, and no somatostatin agonists). A myocardial uptake volume threshold of 18 cm3 provided comparable diagnostic accuracy (sensitivity, 84% [26/31]; specificity, 94% [47/50]). Conclusion: Myocardial uptake was detected in 26.9% of somatostatin PET scans recorded for oncology indications. This rate was decreased by somatostatin analog treatments and increased in older individuals. However, somatostatin PET scans, analyzed with the quantitative criterion of uptake intensity or volume, are able to identify AM and to differentiate it from myocardial uptake of other origins.


Asunto(s)
Miocarditis , Miocardio , Octreótido , Somatostatina , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad Aguda , Transporte Biológico , Diagnóstico Diferencial , Corazón/diagnóstico por imagen , Miocarditis/diagnóstico por imagen , Miocarditis/metabolismo , Miocardio/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Octreótido/análogos & derivados , Octreótido/metabolismo , Octreótido/farmacocinética , Compuestos Organometálicos/farmacocinética , Compuestos Organometálicos/metabolismo , Tomografía de Emisión de Positrones , Radiofármacos/farmacocinética , Estudios Retrospectivos , Somatostatina/análogos & derivados , Somatostatina/metabolismo
11.
Nat Commun ; 15(1): 6344, 2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-39068220

RESUMEN

Dysfunction of pancreatic δ cells contributes to the etiology of diabetes. Despite their important role, human δ cells are scarce, limiting physiological studies and drug discovery targeting δ cells. To date, no directed δ-cell differentiation method has been established. Here, we demonstrate that fibroblast growth factor (FGF) 7 promotes pancreatic endoderm/progenitor differentiation, whereas FGF2 biases cells towards the pancreatic δ-cell lineage via FGF receptor 1. We develop a differentiation method to generate δ cells from human stem cells by combining FGF2 with FGF7, which synergistically directs pancreatic lineage differentiation and modulates the expression of transcription factors and SST activators during endoderm/endocrine precursor induction. These δ cells display mature RNA profiles and fine secretory granules, secrete somatostatin in response to various stimuli, and suppress insulin secretion from in vitro co-cultured ß cells and mouse ß cells upon transplantation. The generation of human pancreatic δ cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation studies in diabetes.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes , Humanos , Animales , Ratones , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Somatostatina/citología , Endodermo/citología , Endodermo/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Páncreas/citología , Páncreas/metabolismo , Somatostatina/metabolismo , Linaje de la Célula , Insulina/metabolismo , Secreción de Insulina
12.
Mucosal Immunol ; 17(5): 858-870, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38866206

RESUMEN

Corneal wound healing in diabetic patients is usually delayed and accompanied by excessive inflammation. However, the underlying cellular and molecular mechanisms remain poorly understood. Here, we found that somatostatin (SST), an immunosuppressive peptide produced by corneal nerve fibers, was significantly reduced in streptozotocin-induced diabetic mice. In addition, we discovered that topical administration of exogenous SST significantly improved re-epithelialization and nerve regeneration following diabetic corneal epithelial abrasion. Further analysis showed that topical SST significantly reduced the expression of injury inflammation-related genes, inhibited neutrophil infiltration, and shifted macrophage polarization from pro-inflammatory M1 to anti-inflammatory M2 in diabetic corneas' healing. Moreover, the application of L-817,818, an agonist of the SST receptor type 5 subtype, significantly reduced the inflammatory response following epithelial injury and markedly improved the process of re-epithelialization and nerve regeneration in mice. Taken together, these data suggest that activation of the SST-SST receptor type 5 pathway significantly ameliorates diabetes-induced abnormalities in corneal wound repair in mice. Targeting this pathway may provide a novel strategy to restore impaired corneal wound closure and nerve regeneration in diabetic patients.


Asunto(s)
Córnea , Lesiones de la Cornea , Diabetes Mellitus Experimental , Receptores de Somatostatina , Transducción de Señal , Somatostatina , Cicatrización de Heridas , Animales , Ratones , Somatostatina/metabolismo , Córnea/patología , Córnea/metabolismo , Lesiones de la Cornea/metabolismo , Receptores de Somatostatina/metabolismo , Masculino , Modelos Animales de Enfermedad , Humanos , Macrófagos/metabolismo , Macrófagos/inmunología , Ratones Endogámicos C57BL , Regeneración Nerviosa
13.
Mol Metab ; 86: 101979, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38945296

RESUMEN

OBJECTIVE: Bariatric surgery is an effective treatment to obesity, leading to weight loss and improvement in glycemia, that is characterized by hypersecretion of gastrointestinal hormones. However, weight regain and relapse of hyperglycemia are not uncommon. We set to identify mechanisms that can enhance gastrointestinal hormonal secretion following surgery to sustain weight loss. METHODS: We investigated the effect of somatostatin (Sst) inhibition on the outcomes of bariatric surgery using a mouse model of sleeve gastrectomy (SG). RESULTS: Sst knockout (sst-ko) mice fed with a calorie-rich diet gained weight normally and had a mild favorable metabolic phenotype compared to heterozygous sibling controls, including elevated plasma levels of GLP-1. Mathematical modeling of the feedback inhibition between Sst and GLP-1 showed that Sst exerts its maximal effect on GLP-1 under conditions of high hormonal stimulation, such as following SG. Obese sst-ko mice that underwent SG had higher levels of GLP-1 compared with heterozygous SG-operated controls. The SG-sst-ko mice regained less weight than controls and maintained lower glycemia months after surgery. Obese wild-type mice that underwent SG and were treated daily with a Sst receptor inhibitor for two months had higher GLP-1 levels, regained less weight, and improved metabolic profile compared to saline-treated SG-operated controls, and compared to inhibitor or saline-treated sham-operated obese mice. CONCLUSIONS: Our results suggest that inhibition of Sst signaling enhances the long-term favorable metabolic outcomes of bariatric surgery.


Asunto(s)
Gastrectomía , Péptido 1 Similar al Glucagón , Ratones Noqueados , Obesidad , Somatostatina , Animales , Somatostatina/metabolismo , Ratones , Gastrectomía/métodos , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/sangre , Obesidad/metabolismo , Obesidad/cirugía , Masculino , Cirugía Bariátrica/métodos , Ratones Endogámicos C57BL , Pérdida de Peso , Glucemia/metabolismo , Ratones Obesos
14.
Proc Natl Acad Sci U S A ; 121(26): e2321710121, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38885377

RESUMEN

Somatostatin receptor 5 (SSTR5) is an important G protein-coupled receptor and drug target for neuroendocrine tumors and pituitary disorders. This study presents two high-resolution cryogenicelectron microscope structures of the SSTR5-Gi complexes bound to the cyclic neuropeptide agonists, cortistatin-17 (CST17) and octreotide, with resolutions of 2.7 Å and 2.9 Å, respectively. The structures reveal that binding of these peptides causes rearrangement of a "hydrophobic lock", consisting of residues from transmembrane helices TM3 and TM6. This rearrangement triggers outward movement of TM6, enabling Gαi protein engagement and receptor activation. In addition to hydrophobic interactions, CST17 forms conserved polar contacts similar to somatostatin-14 binding to SSTR2, while further structural and functional analysis shows that extracellular loops differently recognize CST17 and octreotide. These insights elucidate agonist selectivity and activation mechanisms of SSTR5, providing valuable guidance for structure-based drug development targeting this therapeutically relevant receptor.


Asunto(s)
Octreótido , Receptores de Somatostatina , Receptores de Somatostatina/metabolismo , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/química , Humanos , Octreótido/química , Octreótido/farmacología , Octreótido/metabolismo , Neuropéptidos/metabolismo , Neuropéptidos/química , Microscopía por Crioelectrón , Unión Proteica , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/metabolismo , Somatostatina/metabolismo , Somatostatina/química , Somatostatina/análogos & derivados , Modelos Moleculares , Células HEK293
15.
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
16.
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
17.
Cell Rep ; 43(5): 114197, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38733587

RESUMEN

Interneurons (INs), specifically those in disinhibitory circuits like somatostatin (SST) and vasoactive intestinal peptide (VIP)-INs, are strongly modulated by the behavioral context. Yet, the mechanisms by which these INs are recruited during active states and whether their activity is consistent across sensory cortices remain unclear. We now report that in mice, locomotor activity strongly recruits SST-INs in the primary somatosensory (S1) but not the visual (V1) cortex. This diverse engagement of SST-INs cannot be explained by differences in VIP-IN function but is absent in the presence of visual input, suggesting the involvement of feedforward sensory pathways. Accordingly, inactivating the somatosensory thalamus, but not decreasing VIP-IN activity, significantly reduces the modulation of SST-INs by locomotion. Model simulations suggest that the differences in SST-INs across behavioral states can be explained by varying ratios of VIP- and thalamus-driven activity. By integrating feedforward activity with neuromodulation, SST-INs are anticipated to be crucial for adapting sensory processing to behavioral states.


Asunto(s)
Interneuronas , Somatostatina , Péptido Intestinal Vasoactivo , Animales , Interneuronas/metabolismo , Interneuronas/fisiología , Somatostatina/metabolismo , Ratones , Péptido Intestinal Vasoactivo/metabolismo , Corteza Somatosensorial/fisiología , Corteza Somatosensorial/metabolismo , Masculino , Ratones Endogámicos C57BL , Locomoción/fisiología , Conducta Animal/fisiología , Corteza Visual/fisiología , Corteza Visual/metabolismo , Tálamo/fisiología , Tálamo/metabolismo
18.
Acta Neuropathol ; 147(1): 80, 2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714540

RESUMEN

GABAergic interneurons play a critical role in maintaining neural circuit balance, excitation-inhibition regulation, and cognitive function modulation. In tuberous sclerosis complex (TSC), GABAergic neuron dysfunction contributes to disrupted network activity and associated neurological symptoms, assumingly in a cell type-specific manner. This GABAergic centric study focuses on identifying specific interneuron subpopulations within TSC, emphasizing the unique characteristics of medial ganglionic eminence (MGE)- and caudal ganglionic eminence (CGE)-derived interneurons. Using single-nuclei RNA sequencing in TSC patient material, we identify somatostatin-expressing (SST+) interneurons as a unique and immature subpopulation in TSC. The disrupted maturation of SST+ interneurons may undergo an incomplete switch from excitatory to inhibitory GABAergic signaling during development, resulting in reduced inhibitory properties. Notably, this study reveals markers of immaturity specifically in SST+ interneurons, including an abnormal NKCC1/KCC2 ratio, indicating an imbalance in chloride homeostasis crucial for the postsynaptic consequences of GABAergic signaling as well as the downregulation of GABAA receptor subunits, GABRA1, and upregulation of GABRA2. Further exploration of SST+ interneurons revealed altered localization patterns of SST+ interneurons in TSC brain tissue, concentrated in deeper cortical layers, possibly linked to cortical dyslamination. In the epilepsy context, our research underscores the diverse cell type-specific roles of GABAergic interneurons in shaping seizures, advocating for precise therapeutic considerations. Moreover, this study illuminates the potential contribution of SST+ interneurons to TSC pathophysiology, offering insights for targeted therapeutic interventions.


Asunto(s)
Neuronas GABAérgicas , Interneuronas , Esclerosis Tuberosa , Humanos , Neuronas GABAérgicas/patología , Neuronas GABAérgicas/metabolismo , Eminencia Ganglionar , Interneuronas/patología , Interneuronas/metabolismo , Eminencia Media/patología , Eminencia Media/metabolismo , Receptores de GABA-A/metabolismo , Somatostatina/metabolismo , Esclerosis Tuberosa/patología , Esclerosis Tuberosa/metabolismo , Animales
19.
J Endocrinol ; 261(3)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38593829

RESUMEN

Pancreatic alpha cell activity and glucagon secretion lower as glucose levels increase. While part of the decrease is regulated by glucose itself, paracrine signaling by their neighboring beta and delta cells also plays an important role. Somatostatin from delta cells is an important local inhibitor of alpha cells at high glucose. Additionally, urocortin 3 (UCN3) is a hormone that is co-released from beta cells with insulin and acts locally to potentiate somatostatin secretion from delta cells. UCN3 thus inhibits insulin secretion via a negative feedback loop with delta cells, but its role with respect to alpha cells and glucagon secretion is not understood. We hypothesize that the somatostatin-driven glucagon inhibition at high glucose is regulated in part by UCN3 from beta cells. Here, we use a combination of live functional Ca2+ and cAMP imaging as well as direct glucagon secretion measurement, all from alpha cells in intact mouse islets, to determine the contributions of UCN3 to alpha cell behavior. Exogenous UCN3 treatment decreased alpha cell Ca2+ and cAMP levels and inhibited glucagon release. Blocking endogenous UCN3 signaling increased alpha cell Ca2+ by 26.8 ± 7.6%, but this did not result in increased glucagon release at high glucose. Furthermore, constitutive deletion of Ucn3 did not increase Ca2+ activity or glucagon secretion relative to controls. UCN3 is thus capable of inhibiting mouse alpha cells, but, given the subtle effects of endogenous UCN3 signaling on alpha cells, we propose that UCN3-driven somatostatin may serve to regulate local paracrine glucagon levels in the islet instead of inhibiting gross systemic glucagon release.


Asunto(s)
Células Secretoras de Glucagón , Glucagón , Comunicación Paracrina , Urocortinas , Animales , Urocortinas/metabolismo , Urocortinas/genética , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Ratones , Glucagón/metabolismo , Glucosa/metabolismo , Calcio/metabolismo , Masculino , Ratones Endogámicos C57BL , AMP Cíclico/metabolismo , Somatostatina/farmacología , Somatostatina/metabolismo
20.
Cell Rep ; 43(4): 114115, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38607918

RESUMEN

In the CA1 hippocampus, vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) play a prominent role in disinhibitory circuit motifs. However, the specific behavioral conditions that lead to circuit disinhibition remain uncertain. To investigate the behavioral relevance of VIP-IN activity, we employed wireless technologies allowing us to monitor and manipulate their function in freely behaving mice. Our findings reveal that, during spatial exploration in new environments, VIP-INs in the CA1 hippocampal region become highly active, facilitating the rapid encoding of novel spatial information. Remarkably, both VIP-INs and pyramidal neurons (PNs) exhibit increased activity when encountering novel changes in the environment, including context- and object-related alterations. Concurrently, somatostatin- and parvalbumin-expressing inhibitory populations show an inverse relationship with VIP-IN and PN activity, revealing circuit disinhibition that occurs on a timescale of seconds. Thus, VIP-IN-mediated disinhibition may constitute a crucial element in the rapid encoding of novelty and the acquisition of recognition memory.


Asunto(s)
Región CA1 Hipocampal , Interneuronas , Reconocimiento en Psicología , Péptido Intestinal Vasoactivo , Animales , Interneuronas/metabolismo , Interneuronas/fisiología , Péptido Intestinal Vasoactivo/metabolismo , Región CA1 Hipocampal/fisiología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/citología , Ratones , Masculino , Reconocimiento en Psicología/fisiología , Células Piramidales/metabolismo , Células Piramidales/fisiología , Ratones Endogámicos C57BL , Memoria/fisiología , Parvalbúminas/metabolismo , Conducta Exploratoria/fisiología , Somatostatina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA