Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Transplant ; 33: 9636897241241995, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38554052

RESUMEN

The parathyroid cell is a vital regulator of extracellular calcium levels, operating through the secretion of parathyroid hormone (PTH). Despite its importance, the regulation of PTH secretion remains complex and not fully understood, representing a unique interplay between extracellular and intracellular calcium, and hormone secretion. One significant challenge in parathyroid research has been the difficulty in maintaining cells ex vivo for in-depth cellular investigations. To address this issue, we introduce a novel platform for parathyroid cell transplantation and noninvasive in vivo imaging using the anterior chamber of the eye as a transplantation site. We found that parathyroid adenoma tissue transplanted into the mouse eye engrafted onto the iris, became vascularized, and retained cellular composition. Transplanted animals exhibited elevated PTH levels, indicating a functional graft. With in vivo confocal microscopy, we were able to repetitively monitor parathyroid graft morphology and vascularization. In summary, there is a pressing need for new methods to study complex cellular processes in parathyroid cells. Our study provides a novel approach for noninvasive in vivo investigations that can be applied to understand parathyroid physiology and pathology under physiological and pathological conditions. This innovative strategy can deepen our knowledge on parathyroid function and disease.


Asunto(s)
Calcio , Neoplasias de las Paratiroides , Ratones , Animales , Glándulas Paratiroides/diagnóstico por imagen , Glándulas Paratiroides/patología , Hormona Paratiroidea , Neoplasias de las Paratiroides/diagnóstico por imagen , Neoplasias de las Paratiroides/patología
2.
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
3.
Adipocyte ; 11(1): 315-324, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35531859

RESUMEN

Oncostatin M (OSM) is a member of the glycoprotein 130 cytokine family that is involved in chronic inflammation and increased in adipose tissue under obesity and insulin resistance. OSM was shown to inhibit adipogenesis, suppress browning, and contribute to insulin resistance in cultured white adipocytes. In contrast, OSM may have a metabolically favourable role on adipocytes in mouse models of obesity and insulin resistance. However, a putative role of OSM in modulating lipolysis has not been investigated in detail to date. To address this, cultured white adipocytes of mouse or human origin were exposed to 10 or 100 ng/ml of OSM for various time periods. In murine 3T3-L1 cells, OSM stimulation directly activated hormone-sensitive lipase (HSL) and other players of the lipolytic machinery, and dose-dependently increased free fatty acid and glycerol release. In parallel, OSM attenuated insulin-mediated suppression of lipolysis and induced phosphorylation of serine-residues on the insulin receptor substrate-1 (IRS1) protein. Key experiments were verified in a second murine and a human adipocyte cell line. Inhibiton of extracellular signal-regulated kinase (ERK)-1/2 activation, abolished OSM-mediated HSL phosphorylation and lipolysis. In conclusion, OSM signalling directly promotes lipolysis in white adipocytes in an ERK1/2-dependent manner.


Asunto(s)
Adipocitos Blancos , Oncostatina M , Células 3T3-L1 , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Ácidos Grasos/metabolismo , Glicerol/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Lipólisis , Ratones , Obesidad/metabolismo , Oncostatina M/farmacología
4.
Mol Metab ; 54: 101341, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34547509

RESUMEN

OBJECTIVE: Obesity is associated with low-grade adipose tissue inflammation and locally elevated levels of several glycoprotein 130 (gp130) cytokines. The conversion of white into brown-like adipocytes (browning) may increase energy expenditure and revert the positive energy balance that underlies obesity. Although different gp130 cytokines and their downstream targets were shown to regulate expression of the key browning marker uncoupling protein 1 (Ucp1), it remains largely unknown how this contributes to the development and maintenance of obesity. Herein, we aim to study the role of gp130 cytokine signaling in white adipose tissue (WAT) browning in the obese state. METHODS: Protein and gene expression levels of UCP1 and other thermogenic markers were assessed in a subcutaneous adipocyte cell line, adipose tissue depots from control or adipocyte-specific gp130 knockout (gp130Δadipo) mice fed either chow or a high-fat diet (HFD), or subcutaneous WAT biopsies from a human cohort of lean and obese subjects. WAT browning was modeled in vitro by exposing mature adipocytes to isoproterenol after stimulation with gp130 cytokines. ERK and JAK-STAT signaling were blocked using the inhibitors U0126 and Tofacitinib, respectively. RESULTS: Inguinal WAT of HFD-fed gp130Δadipo mice exhibited significantly elevated levels of UCP1 and other browning markers such as Cidea and Pgc-1α. In vitro, treatment with the gp130 cytokine oncostatin M (OSM) lowered isoproterenol-induced UCP1 protein and gene expression levels in a dose-dependent manner. Mechanistically, OSM mediated the inhibition of Ucp1 via the JAK-STAT but not the ERK pathway. As with mouse data, OSM gene expression in human WAT positively correlated with BMI (r = 0.284, p = 0.021, n = 66) and negatively with UCP1 expression (r = -0.413, p < 0.001, n = 66). CONCLUSIONS: Our data support the notion that OSM negatively regulates thermogenesis in WAT and thus may be an attractive target for treating obesity.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Oncostatina M/metabolismo , Factor de Transcripción STAT3/metabolismo , Células 3T3-L1 , Adipocitos Blancos/metabolismo , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Oncostatina M/genética
5.
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
6.
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
7.
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
8.
Diabetologia ; 59(11): 2387-2392, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27443307

RESUMEN

AIMS/HYPOTHESIS: Dynamic processes in pancreatic tissue are difficult to study. We aimed to develop an intravital imaging method to longitudinally examine engraftment, vascularisation, expansion and differentiation in mature islets or embryonic pancreases transplanted under the kidney capsule. METHODS: Isolated pancreatic islets from adult mice and murine embryonic day (E)12.5 pancreases containing fluorescent biomarkers were transplanted under the kidney capsule of immunodeficient recipient mice. Human islet cells were dispersed, transduced with a lentivirus expressing a fluorescent label and reaggregated before transplantation. Graft-containing kidneys were positioned subcutaneously and an imaging window was fitted into the skin on top of the kidney. Intravital imaging using multiphoton microscopy was performed for up to 2 weeks. Volumes of fluorescently labelled cells were determined as a measure of development and survival. RESULTS: Transplanted islets and embryonic pancreases showed good engraftment and remained viable. Engraftment and vascularisation could be longitudinally examined in murine and human islet cells. Murine islet beta cell volume was unchanged over time. Transplanted embryonic pancreases increased to up to 6.1 times of their original volume and beta cell volume increased 90 times during 2 weeks. CONCLUSIONS/INTERPRETATION: This method allows for repeated intravital imaging of grafts containing various sources of pancreatic tissue transplanted under the kidney capsule. Using fluorescent markers, dynamic information concerning engraftment or differentiation can be visualised and measured.


Asunto(s)
Islotes Pancreáticos/citología , Páncreas/citología , Páncreas/embriología , Animales , Femenino , Humanos , Técnicas In Vitro , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Riñón/citología , Riñón/metabolismo , Lentivirus/genética , Ratones , Páncreas/metabolismo , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...