Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Neurosci ; 18: 1328815, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38601090

RESUMEN

Introduction: Optical Projection Tomography (OPT) and light sheet fluorescence microscopy (LSFM) are high resolution optical imaging techniques, ideally suited for ex vivo 3D whole mouse brain imaging. Although they exhibit high specificity for their targets, the anatomical detail provided by tissue autofluorescence remains limited. Methods: T1-weighted images were acquired from 19 BABB or DBE cleared brains to create an MR template using serial longitudinal registration. Afterwards, fluorescent OPT and LSFM images were coregistered/normalized to the MR template to create fusion images. Results: Volumetric calculations revealed a significant difference between BABB and DBE cleared brains, leading to develop two optimized templates, with associated tissue priors and brain atlas, for BABB (OCUM) and DBE (iOCUM). By creating fusion images, we identified virus infected brain regions, mapped dopamine transporter and translocator protein expression, and traced innervation from the eye along the optic tract to the thalamus and superior colliculus using cholera toxin B. Fusion images allowed for precise anatomical identification of fluorescent signal in the detailed anatomical context provided by MR. Discussion: The possibility to anatomically map fluorescent signals on magnetic resonance (MR) images, widely used in clinical and preclinical neuroscience, would greatly benefit applications of optical imaging of mouse brain. These specific MR templates for cleared brains enable a broad range of neuroscientific applications integrating 3D optical brain imaging.

2.
Front Bioeng Biotechnol ; 11: 1147244, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37545890

RESUMEN

Genetic modification of pancreatic islet organoids, assembled in vitro prior to transplantation is an emerging alternative to direct in vivo genetic manipulations for a number of clinical and research applications. We have previously shown that dispersion of islet cells followed by re-aggregation into islet organoids, or pseudoislets, allows for efficient transduction with viral vectors, while maintaining physiological functions of native islets. Among viruses currently used for genetic manipulations, adeno-associated viruses (AAVs) have the most attractive safety profile making them suitable for gene therapy applications. Studies reporting on pseudoislet transduction with AAVs are, however, lacking. Here, we have characterized in detail the performance of AAV serotype 8 in transduction of islet cells during pseudoislet formation in comparison with human adenovirus type 5 (AdV5). We have assessed such parameters as transduction efficiency, expression kinetics, and endocrine cell tropism of AAV8 alone or in combination with AdV5. Data provided within our study may serve as a reference point for future functional studies using AAVs for gene transfer to islet cell organoids and will facilitate further development of engineered pseudoislets of superior quality suitable for clinical transplantation.

3.
Adv Healthc Mater ; 11(21): e2200782, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36101484

RESUMEN

Impaired diabetic wound healing is associated with the persistence of chronic inflammation and excessive oxidative stress, which has become one of the most serious clinical challenges. Wound dressings with anti-inflammatory and reactive oxygen species (ROS)-scavenging properties are desirable for diabetic wound treatment. In this study, a shape-adaptable, biodegradable, biocompatible, antioxidant, and immunomodulatory interleukin-33 (IL-33)-cytogel is developed by encapsulating IL-33 into physically cross-linked DNA hydrogels and used as wound dressings to promote diabetic wound healing. The porous microstructures and biodegradable properties of the IL-33-cytogel ensure the local sustained-release of IL-33 in the wound area, where the sustained-release of IL-33 is maintained for at least 7 days. IL-33-cytogel can induce local accumulation of group 2 innate lymphoid cells (ILC2s) and regulatory T cells (Tregs), as well as M1-to-M2 transition at the wound sites. Additionally, the antioxidant and biocompatible characteristics of DNA hydrogels promote the scavenging of intracellular ROS without affecting cell viability. As a result, local inflammation in the diabetic wound area is resolved upon IL-33-cytogel treatment, which is accompanied by improved granulation tissue regeneration and accelerated wound closure. This study demonstrates a promising strategy in tissue engineering and regenerative medicine by incorporating DNA hydrogels and cytokine immunotherapy for promoting diabetic wound healing.


Asunto(s)
Diabetes Mellitus , Hidrogeles , Humanos , Hidrogeles/química , Antioxidantes , Interleucina-33 , Inmunidad Innata , Preparaciones de Acción Retardada , Especies Reactivas de Oxígeno , Citocinas , Linfocitos , Cicatrización de Heridas , Inflamación , ADN
4.
Sci Data ; 9(1): 558, 2022 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-36088402

RESUMEN

Mouse models for streptozotocin (STZ) induced diabetes probably represent the most widely used systems for preclinical diabetes research, owing to the compound's toxic effect on pancreatic ß-cells. However, a comprehensive view of pancreatic ß-cell mass distribution subject to STZ administration is lacking. Previous assessments have largely relied on the extrapolation of stereological sections, which provide limited 3D-spatial and quantitative information. This data descriptor presents multiple ex vivo tomographic optical image datasets of the full ß-cell mass distribution in mice subject to single high and multiple low doses of STZ administration, and in glycaemia recovered mice. The data further include information about structural features, such as individual islet ß-cell volumes, spatial coordinates, and shape as well as signal intensities for both insulin and GLUT2. Together, they provide the most comprehensive anatomical record of the effects of STZ administration on the islet of Langerhans in mice. As such, this data descriptor may serve as reference material to facilitate the planning, use and (re)interpretation of this widely used disease model.


Asunto(s)
Diabetes Mellitus Experimental , Islotes Pancreáticos , Animales , Glucemia/análisis , Insulina/análisis , Ratones , Estreptozocina/análisis
5.
J Cell Mol Med ; 26(18): 4847-4858, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35975481

RESUMEN

Significant pancreatic islet dysfunction and loss shortly after transplantation to the liver limit the widespread implementation of this procedure in the clinic. Nonimmune factors such as reactive oxygen species and inflammation have been considered as the primary driving force for graft failure. The adipokine adiponectin plays potent roles against inflammation and oxidative stress. Previous studies have demonstrated that systemic administration of adiponectin significantly prevented islet loss and enhanced islet function at post-transplantation period. In vitro studies indicate that adiponectin protects islets from hypoxia/reoxygenation injury, oxidative stress as well as TNF-α-induced injury. By applying adenovirus mediated transfection, we now engineered islet cells to express exogenous adiponectin gene prior to islet transplantation. Adenovirus-mediated adiponectin transfer to a syngeneic suboptimal islet graft transplanted under kidney capsule markedly prevented inflammation, preserved islet graft mass and improved islet transplant outcomes. These results suggest that adenovirus-mediated adiponectin gene therapy would be a beneficial clinical engineering approach for islet preservation in islet transplantation.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Adenoviridae/genética , Adiponectina/genética , Terapia Genética , Supervivencia de Injerto , Humanos , Inflamación , Trasplante de Islotes Pancreáticos/métodos
6.
Nucleic Acids Res ; 50(13): 7783-7799, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35801912

RESUMEN

CRISPR-based precise gene-editing requires simultaneous delivery of multiple components into living cells, rapidly exceeding the cargo capacity of traditional viral vector systems. This challenge represents a major roadblock to genome engineering applications. Here we exploit the unmatched heterologous DNA cargo capacity of baculovirus to resolve this bottleneck in human cells. By encoding Cas9, sgRNA and Donor DNAs on a single, rapidly assembled baculoviral vector, we achieve with up to 30% efficacy whole-exon replacement in the intronic ß-actin (ACTB) locus, including site-specific docking of very large DNA payloads. We use our approach to rescue wild-type podocin expression in steroid-resistant nephrotic syndrome (SRNS) patient derived podocytes. We demonstrate single baculovirus vectored delivery of single and multiplexed prime-editing toolkits, achieving up to 100% cleavage-free DNA search-and-replace interventions without detectable indels. Taken together, we provide a versatile delivery platform for single base to multi-gene level genome interventions, addressing the currently unmet need for a powerful delivery system accommodating current and future CRISPR technologies without the burden of limited cargo capacity.


Asunto(s)
Baculoviridae , Sistemas CRISPR-Cas , Baculoviridae/genética , Sistemas CRISPR-Cas/genética , ADN/genética , Edición Génica , Vectores Genéticos , Humanos
7.
Sci Transl Med ; 14(638): eaba9112, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35353540

RESUMEN

During progression of type 2 diabetes, pancreatic ß cells are subjected to sustained metabolic overload. We postulated that this state mediates a hypoxic phenotype driven by hypoxia-inducible factor-1α (HIF-1α) and that treatment with the HIF-1α inhibitor PX-478 would improve ß cell function. Our studies showed that the HIF-1α protein was present in pancreatic ß cells of diabetic mouse models. In mouse islets with high glucose metabolism, the emergence of intracellular Ca2+ oscillations at low glucose concentration and the abnormally high basal release of insulin were suppressed by treatment with the HIF-1α inhibitor PX-478, indicating improvement of ß cell function. Treatment of db/db mice with PX-478 prevented the rise of glycemia and diabetes progression by maintenance of elevated plasma insulin concentration. In streptozotocin-induced diabetic mice, PX-478 improved the recovery of glucose homeostasis. Islets isolated from these mice showed hallmarks of improved ß cell function including elevation of insulin content, increased expression of genes involved in ß cell function and maturity, inhibition of dedifferentiation markers, and formation of mature insulin granules. In response to PX-478 treatment, human islet organoids chronically exposed to high glucose presented improved stimulation index of glucose-induced insulin secretion. These results suggest that the HIF-1α inhibitor PX-478 has the potential to act as an antidiabetic therapeutic agent that preserves ß cell function under metabolic overload.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Ratones , Compuestos de Mostaza/metabolismo , Compuestos de Mostaza/farmacología , Fenilpropionatos
8.
Front Endocrinol (Lausanne) ; 12: 652853, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33967961

RESUMEN

The endocrine cells confined in the islets of Langerhans are responsible for the maintenance of blood glucose homeostasis. In particular, beta cells produce and secrete insulin, an essential hormone regulating glucose uptake and metabolism. An insufficient amount of beta cells or defects in the molecular mechanisms leading to glucose-induced insulin secretion trigger the development of diabetes, a severe disease with epidemic spreading throughout the world. A comprehensive appreciation of the diverse adaptive procedures regulating beta cell mass and function is thus of paramount importance for the understanding of diabetes pathogenesis and for the development of effective therapeutic strategies. While significant findings were obtained by the use of islets isolated from the pancreas, in vitro studies are inherently limited since they lack the many factors influencing pancreatic islet cell function in vivo and do not allow for longitudinal monitoring of islet cell plasticity in the living organism. In this respect a number of imaging methodologies have been developed over the years for the study of islets in situ in the pancreas, a challenging task due to the relatively small size of the islets and their location, scattered throughout the organ. To increase imaging resolution and allow for longitudinal studies in individual islets, another strategy is based on the transplantation of islets into other sites that are more accessible for imaging. In this review we present the anterior chamber of the eye as a transplantation and imaging site for the study of pancreatic islet cell plasticity, and summarize the major research outcomes facilitated by this technological platform.


Asunto(s)
Cámara Anterior/metabolismo , Ojo/anatomía & histología , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Páncreas/metabolismo , Animales , Glucemia/metabolismo , Plasticidad de la Célula , Córnea/inmunología , Córnea/fisiología , Ojo/inmunología , Homeostasis , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Ratones , Microscopía Confocal , Monitoreo Fisiológico , Páncreas/fisiología , Ratas
9.
Commun Biol ; 3(1): 541, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32999405

RESUMEN

Mouse models of Streptozotocin (STZ) induced diabetes represent the most widely used preclinical diabetes research systems. We applied state of the art optical imaging schemes, spanning from single islet resolution to the whole organ, providing a first longitudinal, 3D-spatial and quantitative account of ß-cell mass (BCM) dynamics and islet longevity in STZ-treated mice. We demonstrate that STZ-induced ß-cell destruction predominantly affects large islets in the pancreatic core. Further, we show that hyperglycemic STZ-treated mice still harbor a large pool of remaining ß-cells but display pancreas-wide downregulation of glucose transporter type 2 (GLUT2). Islet gene expression studies confirmed this downregulation and revealed impaired ß-cell maturity. Reversing hyperglycemia by islet transplantation partially restored the expression of markers for islet function, but not BCM. Jointly our results indicate that STZ-induced hyperglycemia results from ß-cell dysfunction rather than ß-cell ablation and that hyperglycemia in itself sustains a negative feedback loop restraining islet function recovery.


Asunto(s)
Diabetes Mellitus Experimental/patología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Animales , Biomarcadores/metabolismo , Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo , Transportador de Glucosa de Tipo 2/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestructura , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Fluorescente
10.
EBioMedicine ; 45: 529-541, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31262716

RESUMEN

BACKGROUND: The functional quality of insulin-secreting islet beta cells is a major factor determining the outcome of clinical transplantations for diabetes. It is therefore of importance to develop methodological strategies aiming at optimizing islet cell function prior to transplantation. In this study we propose a synthetic biology approach to genetically engineer cellular signalling pathways in islet cells. METHODS: We established a novel procedure to modify islet beta cell function by combining adenovirus-mediated transduction with reaggregation of islet cells into pseudoislets. As a proof-of-concept for the genetic engineering of islets prior to transplantation, this methodology was applied to increase the expression of the V1b receptor specifically in insulin-secreting beta cells. The functional outcomes were assessed in vitro and in vivo following transplantation into the anterior chamber of the eye. FINDINGS: Pseudoislets produced from mouse dissociated islet cells displayed basic functions similar to intact native islets in terms of glucose induced intracellular signalling and insulin release, and after transplantation were properly vascularized and contributed to blood glucose homeostasis. The synthetic amplification of the V1b receptor signalling in beta cells successfully modulated pseudoislet function in vitro. Finally, in vivo responses of these pseudoislet grafts to vasopressin allowed evaluation of the potential benefits of this approach in regenerative medicine. INTERPRETATION: These results are promising first steps towards the generation of high-quality islets and suggest synthetic biology as an important tool in future clinical islet transplantations. Moreover, the presented methodology might serve as a useful research strategy to dissect cellular signalling mechanisms of relevance for optimal islet function.


Asunto(s)
Diabetes Mellitus/terapia , Ingeniería Genética , Trasplante de Islotes Pancreáticos/métodos , Biosíntesis de Proteínas , Animales , Glucemia , Diabetes Mellitus/patología , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Islotes Pancreáticos , Ratones
11.
Sci Rep ; 7(1): 12440, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28963457

RESUMEN

Functional beta cell mass is an essential biomarker for the diagnosis and staging of diabetes. It has however proven technically challenging to study this parameter during diabetes progression. Here we have detailed the kinetics of the rapid decline in functional beta cell mass in the RIP-DTR mouse, a model of hyperglycemia resulting from diphtheria toxin induced beta cell ablation. A novel combination of imaging modalities was employed to study the pattern of beta cell destruction. Optical projection tomography of the pancreas and longitudinal in vivo confocal microscopy of islets transplanted into the anterior chamber of the eye allowed to investigate kinetics and tomographic location of beta cell mass decay in individual islets as well as at the entire islet population level. The correlation between beta cell mass and function was determined by complementary in vivo and ex vivo characterizations, demonstrating that beta cell function and glucose tolerance were impaired within the first two days following treatment when more than 50% of beta cell mass was remaining. Our results illustrate the importance of acquiring quantitative functional and morphological parameters to assess the functional status of the endocrine pancreas.


Asunto(s)
Diabetes Mellitus Experimental/patología , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Hiperglucemia/patología , Insulina/deficiencia , Islotes Pancreáticos/ultraestructura , Proteínas Recombinantes de Fusión/genética , Animales , Cámara Anterior/cirugía , Glucemia/metabolismo , Recuento de Células , Muerte Celular , Coristoma , Diabetes Mellitus Experimental/diagnóstico por imagen , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Expresión Génica , Prueba de Tolerancia a la Glucosa , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Hiperglucemia/diagnóstico por imagen , Hiperglucemia/genética , Hiperglucemia/metabolismo , Insulina/genética , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos/métodos , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal , Regiones Promotoras Genéticas , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Tomografía Óptica
12.
Sci Rep ; 7(1): 6646, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28751653

RESUMEN

Despite the dramatic increase in the prevalence of diabetes, techniques for in situ studies of the underlying pancreatic biochemistry are lacking. Such methods would facilitate obtaining mechanistic understanding of diabetes pathophysiology and aid in prognostic and/or diagnostic assessments. In this report we demonstrate how a multivariate imaging approach (orthogonal projections to latent structures - discriminant analysis) can be applied to generate full vibrational microspectroscopic profiles of pancreatic tissues. These profiles enable extraction of known and previously unrecorded biochemical alterations in models of diabetes, and allow for classification of the investigated tissue with regards to tissue type, strain and stage of disease progression. Most significantly, the approach provided evidence for dramatic alterations of the pancreatic biochemistry at the initial onset of immune-infiltration in the Non Obese Diabetic model for type 1 diabetes. Further, it enabled detection of a previously undocumented accumulation of collagen fibrils in the leptin deficient ob/ob mouse islets. By generating high quality spectral profiles through the tissue capsule of hydrated human pancreata and by in vivo Raman imaging of pancreatic islets transplanted to the anterior chamber of the eye, we provide critical feasibility studies for the translation of this technique to diagnostic assessments of pancreatic biochemistry in vivo.


Asunto(s)
Diabetes Mellitus Tipo 1/diagnóstico , Páncreas/metabolismo , Análisis Espectral/métodos , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Leptina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Páncreas/fisiopatología
13.
Proc Natl Acad Sci U S A ; 112(20): E2611-9, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25941406

RESUMEN

Insulin resistance and ß-cell failure are the major defects in type 2 diabetes mellitus. However, the molecular mechanisms linking these two defects remain unknown. Elevated levels of apolipoprotein CIII (apoCIII) are associated not only with insulin resistance but also with cardiovascular disorders and inflammation. We now demonstrate that local apoCIII production is connected to pancreatic islet insulin resistance and ß-cell failure. An increase in islet apoCIII causes promotion of a local inflammatory milieu, increased mitochondrial metabolism, deranged regulation of ß-cell cytoplasmic free Ca(2+) concentration ([Ca(2+)]i) and apoptosis. Decreasing apoCIII in vivo results in improved glucose tolerance, and pancreatic apoCIII knockout islets transplanted into diabetic mice, with high systemic levels of the apolipoprotein, demonstrate a normal [Ca(2+)]i response pattern and no hallmarks of inflammation. Hence, under conditions of islet insulin resistance, locally produced apoCIII is an important diabetogenic factor involved in impairment of ß-cell function and may thus constitute a novel target for the treatment of type 2 diabetes mellitus.


Asunto(s)
Apolipoproteína C-III/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/patología , Análisis de Varianza , Animales , Apolipoproteína C-III/genética , Western Blotting , Calcio/metabolismo , Línea Celular Tumoral , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Confocal , Mitocondrias/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Cell Tissue Res ; 358(2): 331-42, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25038744

RESUMEN

The protein nucleobindin 1 (NUCB1; also known as CALNUC or Nuc) contains an intriguing combination of DNA- and calcium-binding motifs, a trait that it shares with the protein nucleobindin 2 (NUCB2; also known as nesfatin). NUCB2 has been implicated in several aspects of metabolic control and has been identified in a number of endocrine organs. No such comprehensive mapping of NUCB1 has been presented. We have explored the expression and distribution of NUCB1 in tissues and cells of the mouse endocrine system, with particular focus on the endocrine pancreas. Using reverse transcription plus the polymerase chain reaction (RT-PCR) and Western blot, we demonstrate that NUCB1 is present in the endocrine islets of Langerhans but absent from the exocrine acinar cells. Immunofluorescence studies have revealed that all islet cell types contain NUCB1, including the NUCB2-expressing beta cells. RT-PCR, Western blot and immunofluorescence have shown that NUCB1 is expressed in the pituitary, thyroid, parathyroid, gastrointestinal tract, adrenals and gonads. However, within these tissues, NUCB1 expression is not ubiquitous. For example, in the testis, NUCB1 occurs in the seminiferous tubules but not in the Leydig-cell-containing interstitial tissue. Similarly, the lamina propria of the duodenum lacks NUCB1, despite its presence in enterocytes. Where present, NUCB1 consistently appears to be associated with the Golgi apparatus. Thus, NUCB1 is broadly, but not ubiquitously, expressed in cells of the mouse endocrine system. Together with its location in the Golgi apparatus and its putative Ca(2+)-binding ability, this distribution suggests a role for NUCB1 in Ca(2+) handling/sensing in secretory cells.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Especificidad de Órganos , Animales , Western Blotting , Proteínas de Unión al Calcio/genética , Proteínas de Unión al ADN/genética , Femenino , Técnica del Anticuerpo Fluorescente , Islotes Pancreáticos/citología , Masculino , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Nucleobindinas , Transporte de Proteínas , Fracciones Subcelulares/metabolismo
15.
Br J Nutr ; 111 Suppl 1: S23-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24382120

RESUMEN

In taste cells, taste receptors, their coupled G proteins and downstream signalling elements mediate the detection and transduction of sweet, bitter and umami compounds. In some intestinal endocrine cells, taste receptors and gustducin contribute to the release of glucagon-like peptide 1 (GLP-1) and other gut hormones in response to glucose and non-energetic sweeteners. Conversely, taste cells have been found to express multiple hormones typically found in intestinal endocrine cells, e.g. GLP-1, glucagon, somatostatin and ghrelin. In the present study, by immunohistochemistry, multiple subsets of taste cells were found to express GLP-1. The release of GLP-1 from 'endocrine taste cells' into the bloodstream was examined. In wild-type mice, even after oesophagectomy and vagotomy, oral stimulation with glucose induced an elevation of GLP-1 levels in the bloodstream within 10 min. Stimulation of taste cell explants from wild-type mice with glucose led to the release of GLP-1 into the medium. Knocking out of the Tas1r3 gene did not eliminate glucose-stimulated GLP-1 release from taste cells in vivo. The present results indicate that a portion of the cephalic-phase rise in circulating GLP-1 levels is mediated by the direct release of GLP-1 from taste cells into the bloodstream.


Asunto(s)
Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/sangre , Glucosa/metabolismo , Mucosa Intestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Papilas Gustativas/metabolismo , Gusto , Animales , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Transducción de Señal
16.
Proc Natl Acad Sci U S A ; 110(51): 20581-6, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24248353

RESUMEN

The islets of Langerhans constitute the endocrine part of the pancreas and are responsible for maintenance of blood glucose homeostasis. They are deeply embedded in the exocrine pancreas, limiting their accessibility for functional studies. Understanding regulation of function and survival and assessing the clinical outcomes of individual treatment strategies for diabetes requires a monitoring system that continuously reports on the endocrine pancreas. We describe the application of a natural body window that successfully reports on the properties of in situ pancreatic islets. As proof of principle, we transplanted "reporter islets" into the anterior chamber of the eye of leptin-deficient mice. These islets displayed obesity-induced growth and vascularization patterns that were reversed by leptin treatment. Hence, reporter islets serve as optically accessible indicators of islet function in the pancreas, and also reflect the efficacy of specific treatment regimens aimed at regulating islet plasticity in vivo.


Asunto(s)
Ojo/metabolismo , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Leptina/metabolismo , Aloinjertos , Animales , Ojo/citología , Femenino , Islotes Pancreáticos/citología , Leptina/genética , Ratones , Ratones Obesos
17.
Mol Metab ; 2(1): 1-2, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24024130
18.
Diabetologia ; 56(12): 2669-78, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23963325

RESUMEN

AIMS/HYPOTHESIS: The aim of this study was to visualise the dynamics and interactions of the cells involved in autoimmune-driven inflammation in type 1 diabetes. METHODS: We adopted the anterior chamber of the eye (ACE) transplantation model to perform non-invasive imaging of leucocytes infiltrating the endocrine pancreas during initiation and progression of insulitis in the NOD mouse. Individual, ACE-transplanted islets of Langerhans were longitudinally and repetitively imaged by stereomicroscopy and two-photon microscopy to follow fluorescently labelled leucocyte subsets. RESULTS: We demonstrate that, in spite of the immune privileged status of the eye, the ACE-transplanted islets develop infiltration and beta cell destruction, recapitulating the autoimmune insulitis of the pancreas, and exemplify this by analysing reporter cell populations expressing green fluorescent protein under the Cd11c or Foxp3 promoters. We also provide evidence that differences in morphological appearance of subpopulations of infiltrating leucocytes can be correlated to their distinct dynamic behaviour. CONCLUSIONS/INTERPRETATION: Together, these findings demonstrate that the kinetics and dynamics of these key cellular components of autoimmune diabetes can be elucidated using this imaging platform for single cell resolution, non-invasive and repetitive monitoring of the individual islets of Langerhans during the natural development of autoimmune diabetes.


Asunto(s)
Cámara Anterior/patología , Autoinmunidad , Antígeno CD11c/inmunología , Diabetes Mellitus Tipo 1/patología , Factores de Transcripción Forkhead/inmunología , Inflamación/patología , Islotes Pancreáticos/patología , Estado Prediabético/patología , Animales , Cámara Anterior/inmunología , Autoanticuerpos/sangre , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inmunología , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Inflamación/inmunología , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Microscopía Fluorescente , Estado Prediabético/inmunología
19.
Diabetes ; 60(10): 2571-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21873551

RESUMEN

OBJECTIVE: Freshly isolated pancreatic islets contain, in contrast to cultured islets, intraislet endothelial cells (ECs), which can contribute to the formation of functional blood vessels after transplantation. We have characterized how donor islet endothelial cells (DIECs) may contribute to the revascularization rate, vascular density, and endocrine graft function after transplantation of freshly isolated and cultured islets. RESEARCH DESIGN AND METHODS: Freshly isolated and cultured islets were transplanted under the kidney capsule and into the anterior chamber of the eye. Intravital laser scanning microscopy was used to monitor the revascularization process and DIECs in intact grafts. The grafts' metabolic function was examined by reversal of diabetes, and the ultrastructural morphology by transmission electron microscopy. RESULTS: DIECs significantly contributed to the vasculature of fresh islet grafts, assessed up to 5 months after transplantation, but were hardly detected in cultured islet grafts. Early participation of DIECs in the revascularization process correlated with a higher revascularization rate of freshly isolated islets compared with cultured islets. However, after complete revascularization, the vascular density was similar in the two groups, and host ECs gained morphological features resembling the endogenous islet vasculature. Surprisingly, grafts originating from cultured islets reversed diabetes more rapidly than those originating from fresh islets. CONCLUSIONS: In summary, DIECs contributed to the revascularization of fresh, but not cultured, islets by participating in early processes of vessel formation and persisting in the vasculature over long periods of time. However, the DIECs did not increase the vascular density or improve the endocrine function of the grafts.


Asunto(s)
Células Endoteliales/fisiología , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/irrigación sanguínea , Neovascularización Fisiológica , Animales , Cámara Anterior , Supervivencia Celular , Células Endoteliales/trasplante , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Islotes Pancreáticos/ultraestructura , Riñón , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Receptores de Interleucina-1 , Factores de Tiempo , Trasplante Heterotópico
20.
J Neurosci ; 30(41): 13774-83, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20943918

RESUMEN

Heterologously expressed sensory receptors generally do not achieve the ligand sensitivity observed in vivo, and may require specific accessory proteins to ensure optimal function. We searched for taste cell-expressed receptor transporting protein (RTP) and receptor expression enhancing protein (REEP) family members that might serve as accessory molecules to enhance gustatory receptor function. We determined that REEP2 is an integral membrane protein expressed in taste cells, physically associates with both subunits of the type 1 taste receptor 2 and type 1 taste receptor 3 sweet receptor and specifically enhances responses to tastants of heterologously expressed sweet and bitter taste receptors. Downregulation of endogenously expressed REEP2 in the chemosensory enteroendocrine GLUTag cell line dramatically reduced sensitivity of endogenous sweet receptors. In contrast to the observation that RTP1, RTP2, and REEP1 enhance function of olfactory receptors by promoting their transit to the cell surface, we found that REEP2 does not increase cell surface expression of sweet receptors but instead alters their spatial organization. REEP2 recruits sweet receptors into lipid raft microdomains localized near the taste cell's apical region, thereby improving G-protein-coupled receptor signaling and promoting receptor access to tastants arriving through the apical taste pore.


Asunto(s)
Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Papilas Gustativas/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Inmunohistoquímica , Microdominios de Membrana/genética , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Microscopía Electrónica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...