Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 254(1): 31-45, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33527355

RESUMEN

Maturity-onset diabetes of the young type 5 (MODY5) is due to heterozygous mutations or deletion of HNF1B. No mouse models are currently available to recapitulate the human MODY5 disease. Here, we investigate the pancreatic phenotype of a unique MODY5 mouse model generated by heterozygous insertion of a human HNF1B splicing mutation at the intron-2 splice donor site in the mouse genome. This Hnf1bsp2/+ model generated with targeted mutation of Hnf1b mimicking the c.544+1G>T (T) mutation identified in humans, results in alternative transcripts and a 38% decrease of native Hnf1b transcript levels. As a clinical feature of MODY5 patients, the hypomorphic mouse model Hnf1bsp2/+ displays glucose intolerance. Whereas Hnf1bsp2/+ isolated islets showed no altered insulin secretion, we found a 65% decrease in pancreatic insulin content associated with a 30% decrease in total large islet volume and a 20% decrease in total ß-cell volume. These defects were associated with a 30% decrease in expression of the pro-endocrine gene Neurog3 that we previously identified as a direct target of Hnf1b, showing a developmental etiology. As another clinical feature of MODY5 patients, the Hnf1bsp2/+ pancreases display exocrine dysfunction with hypoplasia. We observed chronic pancreatitis with loss of acinar cells, acinar-to-ductal metaplasia, and lipomatosis, with upregulation of signaling pathways and impaired acinar cell regeneration. This was associated with ductal cell deficiency characterized by shortened primary cilia. Importantly, the Hnf1bsp2/+ mouse model reproduces the pancreatic features of the human MODY5/HNF1B disease, providing a unique in vivo tool for molecular studies of the endocrine and exocrine defects and to advance basic and translational research. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/fisiopatología , Esmalte Dental/anomalías , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Factor Nuclear 1-beta del Hepatocito/genética , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/fisiopatología , Páncreas/fisiopatología , Animales , Enfermedades del Sistema Nervioso Central/patología , Esmalte Dental/patología , Esmalte Dental/fisiopatología , Diabetes Mellitus Tipo 2/patología , Humanos , Enfermedades Renales Quísticas/patología , Ratones , Ratones Transgénicos , Mutación , Páncreas/patología , Fenotipo
2.
Mol Pharm ; 18(12): 4428-4436, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34649437

RESUMEN

Type 1 diabetes develops in childhood and adolescence, with peak incidence in the early teenage years. There is an urgent need for an accurate method to detect insulin-producing ß-cells in patients that is not affected by alterations in ß-cell function. As part of our research program to design specific probes to measure ß-cell mass, we recently developed a novel insulin-binding peptide probe (IBPP) for the detection of ß-cells in vivo. Here, we applied our innovative method to show specific labeling of this IBPP to human and mouse fixed ß-cells in pancreatic islets. Importantly, we showed staining of human and mouse islets in culture without any negative functional or cell viability impact. Moreover, the IBPP-stained mouse islets after tail vein injection in vivo, albeit with batch differences in staining efficiency. In conclusion, we provide evidence showing that the IBPP can be used for future accurate detection of ß-cell mass in a variety of preclinical models of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/diagnóstico por imagen , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Células Cultivadas , Humanos , Insulina/análisis , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Coloración y Etiquetado
3.
Diabetologia ; 61(4): 896-905, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29209740

RESUMEN

AIMS/HYPOTHESIS: Genetic studies show coupling of genes affecting beta cell function to type 1 diabetes, but hitherto no studies on whether beta cell dysfunction could precede insulitis and clinical onset of type 1 diabetes are available. METHODS: We used 40-day-old BioBreeding (BB) DRLyp/Lyp rats (a model of spontaneous autoimmune type 1 diabetes) and diabetes-resistant DRLyp/+ and DR+/+ littermates (controls) to investigate beta cell function in vivo, and insulin and glucagon secretion in vitro. Beta cell mass was assessed by optical projection tomography (OPT) and morphometry. Additionally, measurements of intra-islet blood flow were performed using microsphere injections. We also assessed immune cell infiltration, cytokine expression in islets (by immunohistochemistry and qPCR), as well as islet Glut2 expression and ATP/ADP ratio to determine effects on glucose uptake and metabolism in beta cells. RESULTS: DRLyp/Lyp rats were normoglycaemic and without traces of immune cell infiltrates. However, IVGTTs revealed a significant decrease in the acute insulin response to glucose compared with control rats (1685.3 ± 121.3 vs 633.3 ± 148.7; p < 0.0001). In agreement, insulin secretion was severely perturbed in isolated islets, and both first- and second-phase insulin release were lowered compared with control rats, while glucagon secretion was similar in both groups. Interestingly, after 5-7 days of culture of islets from DRLyp/Lyp rats in normal media, glucose-stimulated insulin secretion (GSIS) was improved; although, a significant decrease in GSIS was still evident compared with islets from control rats at this time (7393.9 ± 1593.7 vs 4416.8 ± 1230.5 pg islet-1 h-1; p < 0.0001). Compared with controls, OPT of whole pancreas from DRLyp/Lyp rats revealed significant reductions in medium (4.1 × 109 ± 9.5 × 107 vs 3.8 × 109 ± 5.8 × 107 µm3; p = 0.044) and small sized islets (1.6 × 109 ± 5.1 × 107 vs 1.4 × 109 ± 4.5 × 107 µm3; p = 0.035). Finally, we found lower intra-islet blood perfusion in vivo (113.1 ± 16.8 vs 76.9 ± 11.8 µl min-1 [g pancreas]-1; p = 0.023) and alterations in the beta cell ATP/ADP ratio in DRLyp/Lyp rats vs control rats. CONCLUSIONS/INTERPRETATION: The present study identifies a deterioration of beta cell function and mass, and intra-islet blood flow that precedes insulitis and diabetes development in animals prone to autoimmune type 1 diabetes. These underlying changes in islet function may be previously unrecognised factors of importance in type 1 diabetes development.


Asunto(s)
Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Células Secretoras de Insulina/citología , Insulina/metabolismo , Adenosina Difosfato/química , Adenosina Trifosfato/química , Animales , Glucemia/metabolismo , Femenino , Genotipo , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Células de Langerhans/metabolismo , Masculino , Páncreas/metabolismo , Perfusión , Ratas , Ratas Endogámicas BB , Ratas Wistar
4.
Surg Endosc ; 30(2): 532-542, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26065537

RESUMEN

BACKGROUND: In type 2 diabetes mellitus, there is a progressive loss of beta-cell mass. Bariatric surgery has in recent investigations showed promising results in terms of diabetes remission, but little is established regarding the effect of surgery on the survival or regeneration of pancreatic beta-cells. In this study, we aim to explore how bariatric surgery with its subsequent hormonal alterations affects the islets of Langerhans. METHODS: Twenty-four Goto-Kakizaki rats were operated with duodenojejunostomy (DJ), sleeve gastrectomy (SG) or sham operation. From the 38th week after surgery, body weight, fasting blood glucose, glycosylated hemoglobin, mixed meal tolerance with repeated measures of insulin, glucagon-like peptide 1, gastrin and total ghrelin were evaluated. Forty-six weeks after surgery, the animals were euthanized and the total beta-cell mass in all animals was examined by three-dimensional volume quantification by optical projection tomography based on the signal from insulin-specific antibody staining. RESULTS: Body weight did not differ between groups (P(g) = 0.37). SG showed lower fasting blood glucose compared to DJ and sham (P(g) = 0.037); HbA1c levels in SG were lower compared to DJ only (p < 0.05). GLP-1 levels were elevated for DJ compared to SG and sham (P(g) = 0.001), whereas gastrin levels were higher in SG compared to the two other groups (P(g) = 0.002). Beta-cell mass was significantly greater in animals operated with SG compared to both DJ and sham (p = 0.036). CONCLUSION: Sleeve gastrectomy is superior to duodenojejunostomy and sham operation when comparing the preservation of beta-cell mass 46 weeks after surgery in Goto-Kakizaki rats. This could be related to both the increased gastrin levels and the long-term improvement in glycemic parameters observed after this procedure.


Asunto(s)
Cirugía Bariátrica/métodos , Diabetes Mellitus Tipo 2/cirugía , Duodeno/cirugía , Gastrectomía/métodos , Células Secretoras de Insulina/patología , Yeyuno/cirugía , Anastomosis Quirúrgica , Animales , Diabetes Mellitus Tipo 2/patología , Imagenología Tridimensional , Masculino , Tamaño de los Órganos , Ratas , Ratas Endogámicas , Tomografía Óptica/métodos , Resultado del Tratamiento
5.
Nat Commun ; 15(1): 3318, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38632302

RESUMEN

Pancreatic islets of Langerhans play a pivotal role in regulating blood glucose homeostasis, but critical information regarding their mass, distribution and composition is lacking within a whole organ context. Here, we apply a 3D imaging pipeline to generate a complete account of the insulin-producing islets throughout the human pancreas at a microscopic resolution and within a maintained spatial 3D context. These data show that human islets are far more heterogenous than previously accounted for with regards to their size distribution and cellular make up. By deep tissue 3D imaging, this in-depth study demonstrates that 50% of the human insulin-expressing islets are virtually devoid of glucagon-producing α-cells, an observation with significant implications for both experimental and clinical research.


Asunto(s)
Células Secretoras de Glucagón , Islotes Pancreáticos , Humanos , Páncreas/metabolismo , Islotes Pancreáticos/metabolismo , Insulina/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucemia/metabolismo , Secreción de Insulina
6.
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.

7.
Phys Biol ; 10(3): 036009, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23629025

RESUMEN

The islets of Langerhans, responsible for controlling blood glucose levels, are dispersed within the pancreas. A universal power law governing the fractal spatial distribution of islets in two-dimensional pancreatic sections has been reported. However, the fractal geometry in the actual three-dimensional pancreas volume, and the developmental process that gives rise to such a self-similar structure, has not been investigated. Here, we examined the three-dimensional spatial distribution of islets in intact mouse pancreata using optical projection tomography and found a power law with a fractal dimension of 2.1. Furthermore, based on two-dimensional pancreatic sections of human autopsies, we found that the distribution of human islets also follows a universal power law with a fractal dimension of 1.5 in adult pancreata, which agrees with the value previously reported in smaller mammalian pancreas sections. Finally, we developed a self-avoiding growth model for the development of the islet distribution and found that the fractal nature of the spatial islet distribution may be associated with the self-avoidance in the branching process of vascularization in the pancreas.


Asunto(s)
Fractales , Islotes Pancreáticos/crecimiento & desarrollo , Modelos Anatómicos , Modelos Biológicos , Adulto , Animales , Femenino , Humanos , Islotes Pancreáticos/anatomía & histología , Ratones , Ratones Endogámicos C57BL , Tomografía Óptica
8.
Opt Express ; 21(14): 16584-604, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23938510

RESUMEN

We demonstrate a technique to improve structural data obtained from Optical Projection Tomography (OPT) using Image Fusion (IF) and contrast normalization. This enables the visualization of molecular expression patterns in biological specimens with highly variable contrast values. In the approach, termed IF-OPT, different exposures are fused by assigning weighted contrasts to each. When applied to projection images from mouse organs and digital phantoms our results demonstrate the capability of IF-OPT to reveal high and low signal intensity details in challenging specimens. We further provide measurements to highlight the benefits of the new algorithm in comparison to other similar methods.


Asunto(s)
Algoritmos , Aumento de la Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Imagen Molecular/métodos , Reconocimiento de Normas Patrones Automatizadas/métodos , Técnica de Sustracción , Tomografía Óptica/métodos , Animales , Ratones
9.
Methods Mol Biol ; 2592: 1-19, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36507982

RESUMEN

The rodent pancreas is the prevalent model system for preclinical diabetes research. However, due to the compound endocrine-exocrine organization of the gland, with the endocrine islets of Langerhans scattered by the thousands throughout the much greater exocrine parenchyma, stereological assessments of endocrine cell mass, commonly insulin-producing ß-cells, are exceedingly challenging. In recent years, optical mesoscopic imaging techniques such as optical projection tomography (OPT) and light sheet fluorescence microscopy (LSFM) have seen dramatic developments, enabling 3D visualization of fluorescently labeled cells in mm- to cm-sized tissues with µm resolution. Here we present a protocol for 3D visualization and "absolute" quantitative assessments of, for example, islet mass throughout the volume of rodent pancreata with maintained spatial context.


Asunto(s)
Islotes Pancreáticos , Tomografía Óptica , Animales , Roedores , Tomografía Óptica/métodos , Páncreas/diagnóstico por imagen , Microscopía Fluorescente , Imagen Molecular , Islotes Pancreáticos/diagnóstico por imagen , Imagenología Tridimensional/métodos
10.
Nat Commun ; 14(1): 2007, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37037810

RESUMEN

Viral tropism within the brain and the role(s) of vertebrate immune response to neurotropic flaviviruses infection is largely understudied. We combine multimodal imaging (cm-nm scale) with single nuclei RNA-sequencing to study Langat virus in wildtype and interferon alpha/beta receptor knockout (Ifnar-/-) mice to visualize viral pathogenesis and define molecular mechanisms. Whole brain viral infection is imaged by Optical Projection Tomography coregistered to ex vivo MRI. Infection is limited to grey matter of sensory systems in wildtype mice, but extends into white matter, meninges and choroid plexus in Ifnar-/- mice. Cells in wildtype display strong type I and II IFN responses, likely due to Ifnb expressing astrocytes, infiltration of macrophages and Ifng-expressing CD8+ NK cells, whereas in Ifnar-/-, the absence of this response contributes to a shift in cellular tropism towards non-activated resident microglia. Multimodal imaging-transcriptomics exemplifies a powerful way to characterize mechanisms of viral pathogenesis and tropism.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Interferón Tipo I , Garrapatas , Ratones , Animales , Interferón Tipo I/metabolismo , Neuronas/metabolismo , Ratones Noqueados , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Tropismo , Garrapatas/metabolismo , Ratones Endogámicos C57BL
11.
Dev Biol ; 352(2): 267-77, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21281624

RESUMEN

Endoderm development is dependent on inductive signals from different structures in close vicinity, including the notochord, lateral plate mesoderm and endothelial cells. Recently, we demonstrated that a functional vascular system is necessary for proper pancreas development, and that sphingosine-1-phosphate (S1P) exhibits the traits of a blood vessel-derived molecule involved in early pancreas morphogenesis. To examine whether S1P(1)-signaling plays a more general role in endoderm development, S1P(1)-deficient mice were analyzed. S1P(1) ablation results in compromised growth of several foregut-derived organs, including the stomach, dorsal and ventral pancreas and liver. Within the developing pancreas the reduction in organ size was due to deficient proliferation of Pdx1(+) pancreatic progenitors, whereas endocrine cell differentiation was unaffected. Ablation of endothelial cells in vitro did not mimic the S1P(1) phenotype, instead, increased organ size and hyperbranching were observed. Consistent with a negative role for endothelial cells in endoderm organ expansion, excessive vasculature was discovered in S1P(1)-deficient embryos. Altogether, our results show that endothelial cell hyperplasia negatively influences organ development in several foregut-derived organs.


Asunto(s)
Endodermo/embriología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Desarrollo Embrionario , Endodermo/citología , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/genética , Morfogénesis/fisiología , Páncreas/irrigación sanguínea , Páncreas/citología , Páncreas/embriología , Páncreas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/deficiencia , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato , Transactivadores/metabolismo
12.
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
13.
Front Endocrinol (Lausanne) ; 12: 633063, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746904

RESUMEN

The exocrine-endocrine multipart organization of the pancreas makes it an exceedingly challenging organ to analyze, quantitatively and spatially. Both in rodents and humans, estimates of the pancreatic cellular composition, including beta-cell mass, has been largely relying on the extrapolation of 2D stereological data originating from limited sample volumes. Alternatively, they have been obtained by low resolution non-invasive imaging techniques providing little detail regarding the anatomical organization of the pancreas and its cellular and/or molecular make up. In this mini-review, the state of the art and the future potential of currently existing and emerging high-resolution optical imaging techniques working in the mm-cm range with µm resolution, here referred to as mesoscopic imaging approaches, will be discussed regarding their contribution toward a better understanding of pancreatic anatomy both in normal conditions and in the diabetic setting. In particular, optical projection tomography (OPT) and light sheet fluorescence microscopy (LSFM) imaging of the pancreas and their associated tissue processing and computational analysis protocols will be discussed in the light of their current capabilities and future potential to obtain more detailed 3D-spatial, quantitative, and molecular information of the pancreas.


Asunto(s)
Microscopía Fluorescente/métodos , Imagen Óptica/métodos , Páncreas/diagnóstico por imagen , Tomografía Óptica/métodos , Humanos , Imagenología Tridimensional/métodos
14.
Commun Biol ; 4(1): 1063, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34508173

RESUMEN

The possibility to quantitatively study specific molecular/cellular features of complete human organs with preserved spatial 3D context would have widespread implications for pre-clinical and clinical medicine. Whereas optical 3D imaging approaches have experienced a formidable revolution, they have remained limited due to current incapacities in obtaining specific labelling within large tissue volumes. We present a simple approach enabling reconstruction of antibody labeled cells within entire human organs with preserved organ context. We demonstrate the utility of the approach by providing volumetric data and 3D distribution of hundreds of thousands of islets of Langerhans within the human pancreas. By assessments of pancreata from non-diabetic and type 2 diabetic individuals, we display previously unrecognized features of the human islet mass distribution and pathology. As such, this method may contribute not only in unraveling new information of the pancreatic anatomy/pathophysiology, but it may be translated to essentially any antibody marker or organ system.


Asunto(s)
Imagenología Tridimensional , Islotes Pancreáticos/citología , Anciano , Humanos , Masculino
15.
eNeuro ; 8(5)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34417283

RESUMEN

Opsin 3 (Opn3) is highly expressed in the adult brain, however, information for spatial and temporal expression patterns during embryogenesis is significantly lacking. Here, an Opn3-eGFP reporter mouse line was used to monitor cell body expression and axonal projections during embryonic and early postnatal to adult stages. By applying 2D and 3D fluorescence imaging techniques, we have identified the onset of Opn3 expression, which predominantly occurred during embryonic stages, in various structures during brain/head development. In addition, this study defines over twenty Opn3-eGFP-positive neural structures never reported before. Opn3-eGFP was first observed at E9.5 in neural regions, including the ganglia that will ultimately form the trigeminal, facial and vestibulocochlear cranial nerves (CNs). As development proceeds, expanded Opn3-eGFP expression coincided with the formation and maturation of critical components of the central and peripheral nervous systems (CNS, PNS), including various motor-sensory tracts, such as the dorsal column-medial lemniscus (DCML) sensory tract, and olfactory, acoustic, and optic tracts. The widespread, yet distinct, detection of Opn3-eGFP already at early embryonic stages suggests that Opn3 might play important functional roles in the developing brain and spinal cord to regulate multiple motor and sensory circuitry systems, including proprioception, nociception, ocular movement, and olfaction, as well as memory, mood, and emotion. This study presents a crucial blueprint from which to investigate autonomic and cognitive opsin-dependent neural development and resultant behaviors under physiological and pathophysiological conditions.


Asunto(s)
Opsinas , Opsinas de Bastones , Animales , Embrión de Mamíferos , Desarrollo Embrionario , Ratones , Médula Espinal
16.
Mol Metab ; 53: 101264, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34091063

RESUMEN

OBJECTIVE: Early postnatal life is a critical period for the establishment of the functional ß-cell mass that will sustain whole-body glucose homeostasis during the lifetime. ß cells are formed from progenitors during embryonic development but undergo significant expansion in quantity and attain functional maturity after birth. The signals and pathways involved in these processes are not fully elucidated. Cyclic adenosine monophosphate (cAMP) is an intracellular signaling molecule that is known to regulate insulin secretion, gene expression, proliferation, and survival of adult ß cells. The heterotrimeric G protein Gs stimulates the cAMP-dependent pathway by activating adenylyl cyclase. In this study, we sought to explore the role of Gs-dependent signaling in postnatal ß-cell development. METHODS: To study Gs-dependent signaling, we generated conditional knockout mice in which the α subunit of the Gs protein (Gsα) was ablated from ß-cells using the Cre deleter line Ins1Cre. Mice were characterized in terms of glucose homeostasis, including in vivo glucose tolerance, glucose-induced insulin secretion, and insulin sensitivity. ß-cell mass was studied using histomorphometric analysis and optical projection tomography. ß-cell proliferation was studied by ki67 and phospho-histone H3 immunostatining, and apoptosis was assessed by TUNEL assay. Gene expression was determined in isolated islets and sorted ß cells by qPCR. Intracellular cAMP was studied in isolated islets using HTRF-based technology. The activation status of the cAMP and insulin-signaling pathways was determined by immunoblot analysis of the relevant components of these pathways in isolated islets. In vitro proliferation of dissociated islet cells was assessed by BrdU incorporation. RESULTS: Elimination of Gsα in ß cells led to reduced ß-cell mass, deficient insulin secretion, and severe glucose intolerance. These defects were evident by weaning and were associated with decreased proliferation and inadequate expression of key ß-cell identity and maturation genes in postnatal ß-cells. Additionally, loss of Gsα caused a broad multilevel disruption of the insulin transduction pathway that resulted in the specific abrogation of the islet proliferative response to insulin. CONCLUSION: We conclude that Gsα is required for ß-cell growth and maturation in the early postnatal stage and propose that this is partly mediated via its crosstalk with insulin signaling. Our findings disclose a tight connection between these two pathways in postnatal ß cells, which may have implications for using cAMP-raising agents to promote ß-cell regeneration and maturation in diabetes.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Subunidades alfa de la Proteína de Unión al GTP Gs/deficiencia , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal
17.
Adv Exp Med Biol ; 654: 39-57, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20217493

RESUMEN

The establishment of improved technologies for imaging of the pancreas is a key element in addressing several aspects of diabetes pathogenesis. In this respect, the development of a protocol that allows for non-invasive scoring of human islets, or islet beta-cells, is of particular importance. The development of such a technology would have profound impact on both clinical and experimental medicine, ranging from early diagnosis of diabetes to the evaluation of therapeutic regimes. Another important task is the development of modalities for high-resolution imaging of experimental animal models for diabetes. Rodent models for diabetes research have for decades been instrumental to the diabetes research community. The ability to image, and to accurately quantify, key players of diabetogenic processes with molecular specificity will be of great importance for elucidating mechanistic aspects of the disease. This chapter aims to overview current progress within these research areas.


Asunto(s)
Diabetes Mellitus/patología , Diagnóstico por Imagen/métodos , Islotes Pancreáticos/fisiología , Animales , Humanos , Procesamiento de Imagen Asistido por Computador , Células Secretoras de Insulina/citología , Luminiscencia , Espectroscopía de Resonancia Magnética , Ratones , Modelos Biológicos , Óptica y Fotónica/métodos , Páncreas/metabolismo , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tomografía Óptica/métodos
18.
Differentiation ; 77(4): 386-94, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19275872

RESUMEN

The lens of the eye is derived from the non-neural ectoderm situated next to the optic vesicle. Fibroblast growth factor (FGF) signals play a major role at various stages of vertebrate lens development ranging from induction and proliferation to differentiation. Less is however known about the identity of genes that are induced by FGF activity within the lens. We have isolated and characterized mouse cytoplasmic activation/proliferation-associated protein-2 (Caprin2), with domains belonging to both the Caprin family and the C1q and tumour necrosis factor (TNF) super-family. Here we show that Caprin2 is expressed in the developing vertebrate lens in mouse and chick, and that Caprin2 expression is up-regulated in primary lens fiber cells, after the induction of crystallins the earliest known markers for differentiated lens fiber cells. Caprin2 is subsequently down-regulated in the centre of the lens at the time and at the position of the first fiber cell denucleation and terminal differentiation. In vitro analyses of lens fiber cell differentiation provide evidence that FGF activity emanating from neighboring prospective retinal cells is required and that FGF8 activity is sufficient to induce Caprin2 in lens fiber cells. These results not only provide evidence that FGF signals induce the newly characterized protein Caprin2 in the lens, but also support the general idea that FGF signals are required for lens fiber cell differentiation.


Asunto(s)
Proteínas del Ojo/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Cristalino/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Biomarcadores/metabolismo , Northern Blotting , Western Blotting , Diferenciación Celular , Embrión de Pollo , Clonación Molecular , Proteínas del Ojo/química , Proteínas del Ojo/genética , Proteínas del Ojo/aislamiento & purificación , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Cristalino/citología , Cristalino/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Proteínas de Unión al ARN , Retina/citología , Regulación hacia Arriba/efectos de los fármacos
19.
Trends Endocrinol Metab ; 31(12): 905-917, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33160815

RESUMEN

A common feature in the pathophysiology of different types of diabetes is the reduction of ß cell mass and/or impairment of ß cell function. Diagnosis and treatment of type 1 and type 2 diabetes is currently hampered by a lack of reliable techniques to restore ß cell survival, to improve insulin secretion, and to quantify ß cell mass in patients. Current new approaches may allow us to precisely and specifically visualize ß cells in vivo and provide viable therapeutic strategies to preserve, recover, and regenerate ß cells. In this review, we discuss recent protective approaches for ß cells and the advantages and limitations of current imaging probes in the field.


Asunto(s)
Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Animales , Humanos , Células Secretoras de Insulina/fisiología
20.
Sci Rep ; 10(1): 18246, 2020 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-33106532

RESUMEN

The possibility to assess pancreatic anatomy with microscopic resolution in three dimensions (3D) would significantly add to pathological analyses of disease processes. Pancreatic ductal adenocarcinoma (PDAC) has a bleak prognosis with over 90% of the patients dying within 5 years after diagnosis. Cure can be achieved by surgical resection, but the efficiency remains drearily low. Here we demonstrate a method that without prior immunohistochemical labelling provides insight into the 3D microenvironment and spread of PDAC and premalignant cysts in intact surgical biopsies. The method is based solely on the autofluorescent properties of the investigated tissues using optical projection tomography and/or light-sheet fluorescence microscopy. It does not interfere with subsequent histopathological analysis and may facilitate identification of tumor-free resection margins within hours. We further demonstrate how the developed approach can be used to assess individual volumes and numbers of the islets of Langerhans in unprecedently large biopsies of human pancreatic tissue, thus providing a new means by which remaining islet mass may be assessed in settings of diabetes. Generally, the method may provide a fast approach to provide new anatomical insight into pancreatic pathophysiology.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Islotes Pancreáticos/patología , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/diagnóstico por imagen , Humanos , Imagenología Tridimensional/métodos , Islotes Pancreáticos/diagnóstico por imagen , Imagen Óptica/métodos , Neoplasias Pancreáticas/diagnóstico por imagen , Tomografía Óptica/métodos , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA