Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 1.088
Filtrar
Más filtros

Publication year range
1.
Biotechnol Bioeng ; 116(8): 1868-1877, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30982957

RESUMEN

Sensitive detection of protein aggregates is important for evaluating the quality of biopharmaceuticals and detecting misfolded proteins in several neurodegenerative diseases. However, it is challenging to detect extremely low concentrations (<10 ppm) of aggregated protein in the presence of high concentrations of soluble protein. Glucagon, a peptide hormone used in the treatment of extreme hypoglycemia, is aggregation-prone and forms amyloid fibrils. Detection of glucagon fibrils using conformation-specific antibodies is an attractive approach for identifying such aggregates during process and formulation development. Therefore, we have used yeast surface display and magnetic-activated cell sorting to sort single-chain antibody libraries to identify antibody variants with high conformational specificity for glucagon fibrils. Notably, we find several high-affinity antibodies that display excellent selectivity for glucagon fibrils, and we have integrated these antibodies into a sensitive immunoassay. Surprisingly, the sensitivity of our assay-which involves direct (nonantibody mediated) glucagon immobilization in microtiter plates-can be significantly enhanced by pretreating the microtiter plates with various types of globular proteins before glucagon immobilization. Moreover, increased total concentrations of glucagon peptide also significantly improve the sensitivity of our assay, which appears to be due to the strong seeding activity of immobilized fibrils at high glucagon concentrations. Our final assay is highly sensitive (fibril detection limit of ~0.5-1 ppm) and is >20 times more sensitive than detection using a conventional, amyloid-specific fluorescent dye (Thioflavin T). We expect that this type of sensitive immunoassay can be readily integrated into the drug development process to improve the generation of safe and potent peptide therapeutics.


Asunto(s)
Amiloide/análisis , Anticuerpos/química , Glucagón/análisis , Amiloide/ultraestructura , Ensayo de Inmunoadsorción Enzimática/métodos , Células HEK293 , Humanos , Agregado de Proteínas , Solubilidad
2.
Analyst ; 144(8): 2541-2549, 2019 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-30864587

RESUMEN

In this work, we demonstrate the potential use of SPRi for secretion-monitoring of pancreatic islets, small micro-organs that regulate glucose homeostasis in the body. In the islets, somatostatin works as a paracrine inhibitor of insulin and glucagon secretion. However, this inhibitory effect is lost in diabetic individuals and little is known about its contribution to the pathology of diabetes. Thus, the simultaneous detection of insulin, glucagon and somatostatin could help understand such communications. Previously, the authors introduced an SPRi biosensor to simultaneously monitor insulin, glucagon and somatostatin using an indirect competitive immunoassay. However, our sensor achieved a relatively high LOD for somatostatin detection (246 nM), the smallest of the three hormones. For this reason, the present work aimed to improve the performance of our SPRi biosensor using gold nanoparticles (GNPs) as a means of ensuring somatostatin detection from a small group of islets. Although GNP amplification is frequently reported in the literature for individual detection of analytes using SPR, studies regarding the optimal strategy in a multiplexed SPR setup are missing. Therefore, with the aim of finding the optimal GNP amplification strategies for multiplex sensing we compared three architectures: (1) GNPs immobilized on the sensor surface, (2) GNPs conjugated with primary antibodies (GNP-Ab1) and (3) GNPs conjugated with a secondary antibody (GNP-Ab2). Among these strategies an immunoassay using GNP-Ab2 conjugates was able to achieve multiplex detection of the three hormones without cross-reactivity and with 9-fold LOD improvement for insulin, 10-fold for glucagon and 200-fold for somatostatin when compared to the SPRi biosensor without GNPs. The present work denotes the first report of the simultaneous detection of such hormones with a sensitivity level comparable to ELISA assays, particularly for somatostatin.


Asunto(s)
Glucagón/análisis , Oro/química , Insulina/análisis , Nanopartículas del Metal/química , Somatostatina/análisis , Anticuerpos Monoclonales/inmunología , Técnicas Biosensibles/métodos , Calibración , Glucagón/inmunología , Humanos , Inmunoensayo/métodos , Insulina/inmunología , Límite de Detección , Somatostatina/inmunología
3.
Int J Mol Sci ; 20(21)2019 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-31671667

RESUMEN

Glucagon circulates in concentrations in the low picomolar range, which is demanding regarding the sensitivity of the methods for quantification applied. In addition, the differential and tissue specific proteolytic processing of the glucagon precursor and the presence in of several glucagon-like sequences, not only in the precursor of glucagon, but also in a number of other peptides of the glucagon-secretin family of peptides, put special demands on the specificity of the assays. Finally, experience has shown that unspecific interference of plasma components has presented additional problems. All of these problems have resulted in a lot of diverging results concerning measured and reported glucagon responses in both humans and experimental animals that have and still are causing considerable debate and controversy. There is very solid evidence that glucagon is an important hormone in human and mammalian metabolism, but its precise physiological role in glucose and lipid metabolism and in metabolic disease has been difficult to establish, not least because of these difficulties. It was our purpose with this review to discuss the methods of glucagon quantification and discuss pitfalls and sources of error. We also reviewed some of the dogmas regarding glucagon secretion in the light of the methodological difficulties.


Asunto(s)
Glucagón/análisis , Plasma/química , Animales , Glucagón/sangre , Guías como Asunto , Humanos , Metabolismo de los Lípidos , Sensibilidad y Especificidad
4.
Anal Chem ; 90(5): 3132-3139, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29378126

RESUMEN

Diabetes arises from secretory defects in vascularized micro-organs known as the islets of Langerhans. Recent studies indicated that furthering our understanding of the paracrine effect of somatostatin on glucose-induced insulin secretion could represent a novel therapeutic avenue for diabetes. While many research groups are interested in insulin and glucagon secretion, few are particularly focused on studying the paracrine interaction in islets' cells, and none on monitoring a secretory fingerprint that contemplates more than two hormones. Surface plasmon resonance imaging can achieve high-throughput and multiplexed biomolecule quantification, making it an ideal candidate for detection of multiple islet's secretion products if arrays of hormones can be properly implemented on the sensing surface. In this study, we introduced a multiplex surface plasmon resonance imaging-based biosensor for simultaneous quantification of insulin, glucagon, and somatostatin. Performing this multiplex biosensing of hormones was mainly the result of the design of an antifouling sensing surface comprised by a mixed self-assembly monolayer of CH3O-PEG-SH and 16-mercaptohexadecanoic acid, which allowed it to operate in a complex matrix such as an islet secretome. The limit of detection in multiplex mode was 1 nM for insulin, 4 nM for glucagon, and 246 nM for somatostatin with a total analysis time of 21 min per point, making our approach the first reporting a label-free and multiplex measurement of such a combination of human hormones. This biosensor holds the promise of providing us with a mean for the further understanding of the paracrine effect of somatostatin on glucose-induced insulin secretion and consequently help develop novel therapeutic agents for diabetes.


Asunto(s)
Técnicas Biosensibles/métodos , Glucagón/análisis , Insulina/análisis , Somatostatina/análisis , Resonancia por Plasmón de Superficie/métodos , Animales , Anticuerpos/inmunología , Incrustaciones Biológicas/prevención & control , Bovinos , Glucagón/inmunología , Humanos , Inmunoensayo/métodos , Insulina/inmunología , Límite de Detección , Muramidasa/química , Ácidos Palmíticos/química , Polietilenglicoles/química , Albúmina Sérica Bovina/química , Somatostatina/inmunología
5.
Diabetologia ; 60(7): 1344-1353, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28364253

RESUMEN

AIMS/HYPOTHESIS: We investigated whether a reduced incretin effect, as observed in patients with type 2 diabetes, can be detected in high-risk individuals, such as women with prior gestational diabetes mellitus (pGDM). METHODS: In this cross-sectional study, 102 women without diabetes with pGDM and 15 control participants without pGDM and with normal glucose tolerance (NGT) underwent a 4 h 75 g OGTT and an isoglycaemic i.v. glucose infusion (IIGI). Women with pGDM were classified as having NGT or prediabetes (impaired fasting glucose and/or impaired glucose tolerance). Insulin sensitivity was assessed using the Matsuda index and HOMA2-IR and the incretin effect was calculated from insulin responses during the study (100% × [AUCinsulin,OGTT - AUCinsulin,IIGI]/AUCinsulin,OGTT). RESULTS: Sixty-three of the 102 women with pGDM (62%) had prediabetes (median [interquartile range]: age, 38.3 [6.5] years; BMI, 32.1 [5.8] kg/m2) and 39 women (38%) had NGT (age, 39.5 [5.6] years; BMI, 31.0 [6.7] kg/m2). Control participants (n = 15) were not significantly different from the pGDM group with regards to age (39.2 [7.4] years) and BMI (28.8 [9.2] kg/m2). Compared with women with NGT and control participants, women with prediabetes had lower insulin sensitivity, as measured by the Matsuda index (3.0 [2.4] vs 5.0 [2.6] vs 1.5 [1.8], respectively; p < 0.001). The incretin effect was 55.3% [27.8], 73.8% [19.0] and 76.7% [24.6] in women with prediabetes, women with normal glucose tolerance and control participants, respectively (p < 0.01). CONCLUSION/INTERPRETATION: Prediabetes was highly prevalent in women with pGDM, and alterations in the incretin effect were detected in this group before the development of type 2 diabetes. TRIAL REGISTRATION: clinicaltrialsregister.eu 2012-001371-37-DK.


Asunto(s)
Diabetes Gestacional/sangre , Diabetes Gestacional/fisiopatología , Incretinas/sangre , Estado Prediabético/sangre , Estado Prediabético/fisiopatología , Adulto , Área Bajo la Curva , Glucemia/análisis , Índice de Masa Corporal , Estudios de Casos y Controles , Estudios Transversales , Dinamarca , Diabetes Mellitus Tipo 2 , Método Doble Ciego , Femenino , Glucagón/análisis , Péptido 1 Similar al Glucagón/análisis , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Persona de Mediana Edad , Análisis Multivariante , Embarazo , Prevalencia
6.
Lab Invest ; 97(5): 577-590, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28218739

RESUMEN

During progression to type 1 diabetes, insulin-producing ß-cells are lost through an autoimmune attack resulting in unrestrained glucagon expression and secretion, activation of glycogenolysis, and escalating hyperglycemia. We recently identified a protein, designated islet homeostasis protein (IHoP), which specifically co-localizes within glucagon-positive α-cells and is overexpressed in the islets of both post-onset non-obese diabetic (NOD) mice and type 1 diabetes patients. Here we report that in the αTC1.9 mouse α-cell line, IHoP was released in response to high-glucose challenge and was found to regulate secretion of glucagon. We also show that in NOD mice with diabetes, major histocompatibility complex class II was upregulated in islets. In addition hyperglycemia was modulated in NOD mice via suppression of IHoP utilizing small interfering RNA (IHoP-siRNA) constructs/approaches. Suppression of IHoP in the pre-diabetes setting maintained normoglycemia, glyconeolysis, and fostered ß-cell restoration in NOD mice 35 weeks post treatment. Furthermore, we performed adoptive transfer experiments using splenocytes from IHoP-siRNA-treated NOD/ShiLtJ mice, which thwarted the development of hyperglycemia and the extent of insulitis seen in recipient mice. Last, IHoP can be detected in the serum of human type 1 diabetes patients and could potentially serve as an early novel biomarker for type 1 diabetes in patients.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas/metabolismo , Animales , Línea Celular , Femenino , Glucagón/análisis , Glucagón/metabolismo , Antígenos HLA-D/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Hiperglucemia/metabolismo , Islotes Pancreáticos/química , Masculino , Ratones , Ratones Endogámicos NOD , Proteínas/análisis , Proteínas/antagonistas & inhibidores , Transactivadores/metabolismo
7.
Eur J Nutr ; 56(2): 591-601, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26593435

RESUMEN

PURPOSE: The present study was undertaken to explore the possible anti-diabetic mechanism(s) of Emblica officinalis (EO) and its active constituent, ellagic acid (EA), in vitro and in vivo. METHOD: Neonatal streptozotocin-induced non-obese type 2 diabetic rats were treated with a methanolic extract of EO (250 or 500 mg/kg) for 28 days, and blood glucose, serum insulin, and plasma antioxidant status were measured. Insulin and glucagon immunostaining and morphometry were performed in pancreatic section, and liver TBARS and GSH levels were measured. Additionally, EA was tested for glucose-stimulated insulin secretion and glucose tolerance test. RESULTS: Treatment with EO extract resulted in a significant decrease in the fasting blood glucose in a dose- and time-dependent manner in the diabetic rats. It significantly increased serum insulin in the diabetic rats in a dose-dependent manner. Insulin-to-glucose ratio was also increased by EO treatment. Immunostaining of pancreas showed that EO250 increased ß-cell size, but EO500 increased ß-cells number in diabetic rats. EO significantly increased plasma total antioxidants and liver GSH and decreased liver TBARS. EA stimulated glucose-stimulated insulin secretion from isolated islets and decreased glucose intolerance in diabetic rats. CONCLUSION: Ellagic acid in EO exerts anti-diabetic activity through the action on ß-cells of pancreas that stimulates insulin secretion and decreases glucose intolerance.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ácido Elágico/administración & dosificación , Hipoglucemiantes , Células Secretoras de Insulina/efectos de los fármacos , Phyllanthus emblica/química , Animales , Antioxidantes , Glucemia/análisis , Frutas/química , Glucagón/análisis , Glutatión/análisis , Insulina/análisis , Insulina/sangre , Células Secretoras de Insulina/química , Células Secretoras de Insulina/citología , Hígado/química , Hígado/efectos de los fármacos , Fitoterapia , Extractos Vegetales/administración & dosificación , Ratas , Sustancias Reactivas al Ácido Tiobarbitúrico/análisis
8.
Am J Physiol Endocrinol Metab ; 311(2): E302-9, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27245336

RESUMEN

Glucagon is a metabolically important hormone, but many aspects of its physiology remain obscure, because glucagon secretion is difficult to measure in mice and rats due to methodological inadequacies. Here, we introduce and validate a low-volume, enzyme-linked immunosorbent glucagon assay according to current analytical guidelines, including tests of sensitivity, specificity, and accuracy, and compare it, using the Bland-Altman algorithm and size-exclusion chromatography, with three other widely cited assays. After demonstrating adequate performance of the assay, we measured glucagon secretion in response to intravenous glucose and arginine in anesthetized mice (isoflurane) and rats (Hypnorm/midazolam). Glucose caused a long-lasting suppression to very low values (1-2 pmol/l) within 2 min in both species. Arginine stimulated secretion 8- to 10-fold in both species, peaking at 1-2 min and returning to basal levels at 6 min (mice) and 12 min (rats). d-Mannitol (osmotic control) was without effect. Ketamine/xylazine anesthesia in mice strongly attenuated (P < 0.01) α-cell responses. Chromatography of pooled plasma samples confirmed the accuracy of the assay. In conclusion, dynamic analysis of glucagon secretion in rats and mice with the novel accurate sandwich enzyme-linked immunosorbent assay revealed extremely rapid and short-lived responses to arginine and rapid and profound suppression by glucose.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Anestésicos Disociativos/farmacología , Animales , Arginina/farmacología , Cromatografía en Gel , Ensayo de Inmunoadsorción Enzimática , Femenino , Glucagón/análisis , Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/efectos de los fármacos , Glucosa/farmacología , Hipnóticos y Sedantes/farmacología , Ketamina/farmacología , Masculino , Manitol/farmacología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Edulcorantes/farmacología , Xilazina/farmacología
9.
Anal Chem ; 88(16): 7910-5, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27440478

RESUMEN

In this report, a method to multiplex fluorescence anisotropy measurements is described using frequency encoding. As a demonstration of the method, simultaneous competitive immunoassays for insulin and glucagon were performed by measuring the ratio of bound and free Cy5-insulin and FITC-glucagon in the presence of their respective antibodies. A vertically polarized 635 nm laser was pulsed at 73 Hz and used to excite Cy5-insulin, while a vertically polarized 488 nm laser pulsed at 137 Hz excited FITC-glucagon. The total emission was split into parallel and perpendicular polarizations and collected onto separate photomultiplier tubes. The signals from each channel were demodulated using a fast Fourier transform, resolving the contributions from each fluorophore. Anisotropy calculations were carried out using the magnitude of the peaks in the frequency domain. The method produced the expected shape of the calibration curves with limits of detection of 0.6 and 5 nM for insulin and glucagon, respectively. This methodology could readily be expanded to other biological systems and further multiplexed to monitor increased numbers of analytes.


Asunto(s)
Polarización de Fluorescencia , Glucagón/análisis , Inmunoensayo , Insulina/análisis , Colorantes Fluorescentes/química , Rayos Láser
10.
Phys Biol ; 13(2): 025004, 2016 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-27063927

RESUMEN

Plasma glucose in mammals is regulated by hormones secreted by the islets of Langerhans embedded in the exocrine pancreas. Islets consist of endocrine cells, primarily α, ß, and δ cells, which secrete glucagon, insulin, and somatostatin, respectively. ß cells form irregular locally connected clusters within islets that act in concert to secrete insulin upon glucose stimulation. Varying demands and available nutrients during development produce changes in the local connectivity of ß cells in an islet. We showed in earlier work that graph theory provides a framework for the quantification of the seemingly stochastic cyto-architecture of ß cells in an islet. To quantify the dynamics of endocrine connectivity during development requires a framework for characterizing changes in the probability distribution on the space of possible graphs, essentially a Fokker-Planck formalism on graphs. With large-scale imaging data for hundreds of thousands of islets containing millions of cells from human specimens, we show that this dynamics can be determined quantitatively. Requiring that rearrangement and cell addition processes match the observed dynamic developmental changes in quantitative topological graph characteristics strongly constrained possible processes. Our results suggest that there is a transient shift in preferred connectivity for ß cells between 1-35 weeks and 12-24 months.


Asunto(s)
Islotes Pancreáticos/citología , Islotes Pancreáticos/crecimiento & desarrollo , Recuento de Células , Preescolar , Gráficos por Computador , Simulación por Computador , Glucagón/análisis , Glucagón/metabolismo , Humanos , Lactante , Recién Nacido , Insulina/análisis , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Modelos Biológicos , Procesos Estocásticos
12.
Histochem Cell Biol ; 143(5): 497-504, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25361590

RESUMEN

Fibroblast activation protein (FAP, seprase, EC 3.4.21.B28) and dipeptidyl peptidase-IV (DPP-IV, CD26, EC 3.4.14.5) are homologous serine proteases implicated in the modulation of the bioavailability and thus the function of a number of biologically active peptides. In spite of their generally nonoverlapping expression patterns, DPP-IV and FAP are co-expressed and probably co-regulated in certain cell types suggesting that for some biological processes their functional synergy is essential. By an in situ enzymatic activity assay, we show an abundant DPP-IV-like enzymatic activity sensitive to a highly specific DPP-IV inhibitor sitagliptin and corresponding DPP-IV immunoreactivity in the adult human islets of Langerhans. Moreover, the homologous protease FAP was present in the human endocrine pancreas and was co-expressed with DPP-IV. DPP-IV and FAP were found in the pancreatic alpha cells as determined by the co-localization with glucagon immunoreactivity. In summary, we show abundant enzymatic activity of the canonical DPP-IV (CD26) in Langerhans islets in the natural tissue context and demonstrate for the first time the co-expression of FAP and DPP-IV in pancreatic alpha cells in adult humans. Given their ability to proteolytically modify several biologically active peptides, both proteases have the potential to modulate the paracrine signaling in the human Langerhans islets.


Asunto(s)
Dipeptidil Peptidasa 4/análisis , Gelatinasas/análisis , Islotes Pancreáticos/enzimología , Proteínas de la Membrana/análisis , Serina Endopeptidasas/análisis , Adulto , Endopeptidasas , Glucagón/análisis , Células Secretoras de Glucagón/enzimología , Humanos , Inmunohistoquímica , Islotes Pancreáticos/citología , Microscopía Confocal
13.
Digestion ; 91(2): 174-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25765455

RESUMEN

BACKGROUND/AIMS: The source of insulin-secreting cells from adult duct system is attractive, but its clinical practice remains poorly understood. Here, we aimed at identifying the distribution of secreted hormone reactive cells in adult ducts. METHODS: Consecutive pancreatic slices from nondiabetic subjects were subjected to immunohistochemistry and immunofluorescence to screen islet hormones (insulin; glucagon, Glu; somatostatin, Som; pancreatic polypeptide, PP) and exocrine biomarkers (cytokeratin 19, CK19; chromogranin A, CgA; amylase). All pancreatic sections were imaged using an optical or confocal microscope. RESULTS: Immunostaining results showed that insulin was expressed in adult ducts, in which the cell count was more than other islet hormone immunoactive cells. CK19-positive cells are mainly distributed in the ducts, whereas CgA-labeled cells are localized in endocrine cells. The duct branches visibly exhibited cell populations that co-expressed islet hormones in exocrine cell populations. CONCLUSIONS: In this report, our findings demonstrate that adult ductal cells that produce insulin may contribute to beta-cell proliferation.


Asunto(s)
Islotes Pancreáticos/química , Conductos Pancreáticos/citología , Hormonas Pancreáticas/análisis , Anciano , Amilasas/análisis , Biomarcadores/análisis , Recuento de Células , Cromogranina A/análisis , Femenino , Glucagón/análisis , Humanos , Inmunohistoquímica , Insulina/análisis , Queratina-19/análisis , Masculino , Persona de Mediana Edad , Páncreas Exocrino/citología , Polipéptido Pancreático/análisis , Somatostatina/análisis
14.
Endocr J ; 62(4): 329-37, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25740613

RESUMEN

Pancreatic beta-cell mass contributes to glucose tolerance. The aim of this study was to evaluate the relationships between human beta-cell mass and various clinical parameters, including insulin secretory capacity. The study included 32 Japanese patients who underwent pancreatectomy and were naive to oral hypoglycemic agents and insulin. They were classified into those with normal glucose tolerance (n=13), impaired glucose tolerance (n=9) and diabetes (n=10), and their insulin secretory capacity and insulin resistance were evaluated. Immunohistochemistry was used to determine relative beta-cell area (%) which represented the proportion of insulin-positive cell area to whole pancreatic section. Increment of C-peptide immunoreactivity level by glucagon test (ΔC-peptide, increment of serum C-peptide [nmol/L] at 6 min after intravenous injection of 1-mg glucagon; r=0.64, p=0.002), homeostasis model assessment of beta-cell function (HOMA-beta, fasting immunoreactive insulin [µIU/mL] x 20 / (fasting plasma glucose [mmol/L] - 3.5); r=0.50, p=0.003), C-peptide index (CPI, fasting C-peptide [nmol/L] / fasting plasma glucose [mmol/L]; r=0.36, p=0.042), and fasting immunoreactive insulin (F-IRI [pmol/L]; r=0.36, p=0.044) correlated significantly and positively with the relative beta-cell area. The area under the curve of plasma glucose level from 0 to 120 min by 75 g-OGTT (AUC0-120) also correlated significantly and inversely with the relative beta-cell area (r=-0.36, p=0.045). Stepwise multiple regression analysis identified ΔC-peptide as the only independent and significant determinant of the relative beta-cell area. We conclude that ΔC-peptide, HOMA-beta, CPI, F-IRI and AUC0-120 correlated closely with the relative beta-cell area, and ΔC-peptide was the most valuable index for the prediction of the area.


Asunto(s)
Péptido C/sangre , Glucagón/sangre , Células Secretoras de Insulina/citología , Anciano , Glucemia/análisis , Recuento de Células , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/patología , Femenino , Glucagón/análisis , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Persona de Mediana Edad
15.
J Pineal Res ; 56(1): 82-96, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24117965

RESUMEN

The pineal hormone melatonin is known to influence insulin secretion via the G-protein-coupled receptor isoforms MT1 and MT2. The present study was aimed to further elucide the impact of melatonin on blood glucose regulation. To this end, mouse lines were used, in which one of the two or both melatonin receptors were deleted. In comparison with wild-type mice of the same age (8-12 months old), increased plasma insulin and melatonin levels and decreased blood glucose levels and body weights were detected in the MT1- and double-knockout lines. The elimination of melatonin receptor signalling also altered blood glucose concentrations, body weight and melatonin and insulin levels when comparing wild-type and receptor knockout mice of different ages (6 wk and 8-12 months old); such changes, however, were dependent on the type of receptor deleted. Furthermore, reverse transcription polymerase chain reaction results provided evidence that melatonin receptor deficiency has an impact on transcript levels of pancreatic islet hormones as well as on pancreatic and hepatic glucose transporters (Glut1 and 2). Under stimulated insulin secretion in the presence of melatonin in the rat insulinoma ß-cells INS-1, the Glut1 transcript level was decreased. In conclusion, the present findings demonstrate that melatonin receptor knockout types affect blood glucose levels, body weight, plasma levels of melatonin and insulin, as well as pancreatic hormone and Glut1 expression in significantly different manners.


Asunto(s)
Glucemia/metabolismo , Receptor de Melatonina MT1/metabolismo , Receptor de Melatonina MT2/metabolismo , Análisis de Varianza , Animales , Glucemia/genética , Peso Corporal/genética , Línea Celular Tumoral , Femenino , Glucagón/análisis , Glucagón/genética , Glucagón/metabolismo , Transportador de Glucosa de Tipo 1/análisis , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Insulina/sangre , Masculino , Melatonina/sangre , Ratones , Ratones Noqueados , Especificidad de Órganos , ARN Mensajero/análisis , ARN Mensajero/genética , Receptor de Melatonina MT1/genética , Receptor de Melatonina MT2/genética , Somatostatina/análisis , Somatostatina/genética , Somatostatina/metabolismo
16.
PLoS One ; 19(5): e0304086, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38771849

RESUMEN

Recently, the first generic glucagon for injection was approved for the treatment of severe hypoglycemia. Unlike its brand name recombinant glucagon, the generic glucagon is synthetic. Since glucagon has a high propensity to form aggregates in solution, it is essential to assess the aggregation profile of the synthetic glucagon compared to the recombinant glucagon. In this study, two robust separation methods, size-exclusion chromatography (SEC-HPLC) and field-flow fractionation coupled with a multi-angle light scattering detector (FFF-MALS), were employed to characterize generic and brand glucagon aggregation in six lots (three newly released, three expired). The presence of aggregation in samples was determined from the generated chromatograms and analyzed. The study showed that both products have comparable aggregation profiles. The SEC-HPLC demonstrated that in both glucagon versions, the expired lots had a higher percentage of dimers than the newly released lots, but even at expiration, the amount was negligible (∼0.1%). The FFF-MALS method did not detect any dimers or higher molecular weight aggregates. Further evaluation of the detection limit found that FFF-MALS was unable to detect aggregates at amounts lower than 0.5% of total glucagon. The negligible amounts of dimer detected in the generic and brand glucagon indicate that both versions are physically stable and are not prone to aggregation under clinically relevant conditions.


Asunto(s)
Cromatografía en Gel , Glucagón , Agregado de Proteínas , Glucagón/química , Glucagón/análisis , Cromatografía Líquida de Alta Presión/métodos , Cromatografía en Gel/métodos , Dispersión de Radiación , Humanos , Luz
17.
Pharmacol Res ; 76: 58-66, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23891763

RESUMEN

The ultimate treatment goal of diabetes is to preserve and restore islet cell function. Treatment of certain diabetic animal models with incretins has been reported to preserve and possibly enhance islet function and promote islet cell growth. The studies reported here detail islet cell anatomy in animals chronically treated with the incretin analog, liraglutide. Our aim was to quantitatively and qualitatively analyze islet cells from diabetic animals treated with vehicle (control) or liraglutide to determine whether normal islet cell anatomy is maintained or enhanced with pharmaceutical treatment. We harvested pancreata from liraglutide and vehicle-treated Zucker Diabetic Fatty (ZDF) rats to examine islet structure and function and obtain isolated islets. Twelve-week-old male rats were assigned to 3 groups: (1) liraglutide-treated diabetic, (2) vehicle-treated diabetic, and (3) lean non-diabetic. Liraglutide was given SC twice daily for 9 weeks. As expected, liraglutide treatment reduced body weight by 15% compared to the vehicle-treated animals, eventually to levels that were not different from lean controls. At the termination of the study, blood glucose was significantly less in the liraglutide-treated rats compared to vehicle treated controls (485.8±22.5 and 547.2±33.1mg/dl, respectively). Insulin content/islet (measured by immunohistochemistry) was 34.2±0.7 pixel units in vehicle-treated rats, and 54.9±0.6 in the liraglutide-treated animals. Glucose-stimulated insulin secretion from isolated islets (measured as the stimulation index) was maintained in the liraglutide-treated rats, but not in the vehicle-treated. However, liraglutide did not preserve normal islet architecture. There was a decrease in the glucagon-positive area/islet and in the α-cell numbers/area with liraglutide treatment (6.5 cells/field), compared to vehicle (17.9 cells/field). There was an increase in ß-cell numbers, the ß- to α-cell ratio that was statistically higher in the liraglutide-treated rats (24.3±4.4) compared to vehicle (9.1±2.8). Disrupted mitochondria were more commonly observed in the α-cells (51.9±10.3% of cells) than in the ß-cells (27.2±4.4%) in the liraglutide-treated group. While liraglutide enhanced or maintained growth and function of certain islet cells, the overall ratio of α- to ß-cells was decreased and there was an absolute reduction in islet α-cell content. There was selective disruption of intracellular α-cell organelles, representing an uncoupling of the bihormonal islet signaling that is required for normal metabolic regulation. The relevance of the findings to long-term liraglutide treatment in people with diabetes is unknown and should be investigated in appropriately designed clinical studies.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Péptido 1 Similar al Glucagón/análogos & derivados , Células Secretoras de Glucagón/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Animales , Glucemia/análisis , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Glucagón/análisis , Glucagón/metabolismo , Péptido 1 Similar al Glucagón/administración & dosificación , Péptido 1 Similar al Glucagón/uso terapéutico , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Hipoglucemiantes/administración & dosificación , Insulina/análisis , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Liraglutida , Masculino , Ratas , Ratas Zucker
18.
Pediatr Diabetes ; 13(1): 51-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22118630

RESUMEN

OBJECTIVE: Proinsulin is a marker of beta-cell distress and dysfunction in type 2 diabetes and transplanted islets. Proinsulin levels are elevated in patients newly diagnosed with type 1 diabetes. Our aim was to assess the relationship between proinsulin, insulin dose-adjusted haemoglobin A1c (IDAA1C), glucagon-like peptide-1 (GLP-1), glucagon, and remission status the first year after diagnosis of type 1 diabetes. METHODS: Juvenile patients (n = 275) were followed 1, 6, and 12 months after diagnosis. At each visit, partial remission was defined as IDAA1C ≤ 9%. The patients had a liquid meal test at the 1-, 6-, and 12-month visits, which included measurement of C-peptide, proinsulin, GLP-1, glucagon, and insulin antibodies (IA). RESULTS: Patients in remission at 6 and 12 months had significantly higher levels of proinsulin compared to non-remitting patients (p < 0.0001, p = 0.0002). An inverse association between proinsulin and IDAA1C was found at 1 and 6 months (p = 0.0008, p = 0.0022). Proinsulin was positively associated with C-peptide (p < 0.0001) and IA (p = 0.0024, p = 0.0068, p < 0.0001) at 1, 6, and 12 months. Glucagon (p < 0.0001 and p < 0.02) as well as GLP-1 (p = 0.0001 and p = 0.002) were significantly lower in remitters than in non-remitters at 6 and 12 months. Proinsulin associated positively with GLP-1 at 1 month (p = 0.004) and negatively at 6 (p = 0.002) and 12 months (p = 0.0002). CONCLUSIONS: In type 1 diabetes, patients in partial remission have higher levels of proinsulin together with lower levels of GLP-1 and glucagon compared to patients not in remission. In new onset type 1 diabetes proinsulin level may be a sign of better residual beta-cell function.


Asunto(s)
Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/epidemiología , Péptido 1 Similar al Glucagón/sangre , Glucagón/sangre , Proinsulina/sangre , Adolescente , Edad de Inicio , Glucemia/análisis , Péptido C/análisis , Péptido C/sangre , Niño , Preescolar , Estudios de Cohortes , Diabetes Mellitus Tipo 1/diagnóstico , Femenino , Estudios de Seguimiento , Glucagón/análisis , Péptido 1 Similar al Glucagón/análisis , Humanos , Lactante , Recién Nacido , Masculino , Proinsulina/análisis , Remisión Espontánea
19.
Anal Bioanal Chem ; 402(9): 2797-803, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22286080

RESUMEN

A rapid microfluidic based capillary electrophoresis immunoassay (CEIA) was developed for on-line monitoring of glucagon secretion from pancreatic islets of Langerhans. In the device, a cell chamber containing living islets was perfused with buffers containing either high or low glucose concentration. Perfusate was continuously sampled by electroosmosis through a separate channel on the chip. The perfusate was mixed on-line with fluorescein isothiocyanate-labeled glucagon (FITC-glucagon) and monoclonal anti-glucagon antibody. To minimize sample dilution, the on-chip mixing ratio of sampled perfusate to reagents was maximized by allowing reagents to only be added by diffusion. Every 6 s, the reaction mixture was injected onto a 1.5-cm separation channel where free FITC-glucagon and the FITC-glucagon-antibody complex were separated under an electric field of 700 V cm(-1). The immunoassay had a detection limit of 1 nM. Groups of islets were quantitatively monitored for changes in glucagon secretion as the glucose concentration was decreased from 15 to 1 mM in the perfusate revealing a pulse of glucagon secretion during a step change. The highly automated system should be enable studies of the regulation of glucagon and its potential role in diabetes and obesity. The method also further demonstrates the potential of rapid CEIA on microfluidic systems for monitoring cellular function.


Asunto(s)
Glucagón/análisis , Glucagón/metabolismo , Inmunoensayo/métodos , Islotes Pancreáticos/metabolismo , Microfluídica/métodos , Animales , Técnicas In Vitro , Islotes Pancreáticos/química , Microfluídica/instrumentación
20.
Proc Natl Acad Sci U S A ; 105(16): 6139-44, 2008 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-18413598

RESUMEN

To determine whether adipocyte storage capacity influences the onset and severity of type 2 diabetes and other components of the metabolic syndrome, we made normal and db/db mice resistant to obesity by overexpressing leptin receptor-b on the aP2-Lepr-b promoter. On a 4% diet, these mice have no phenotype, but on a 60% fat diet, they resist diet-induced obesity because constitutive adipocyte-specific overexpression of Lepr-b prevents obesity via the antilipogenic autocrine/paracrine action of leptin on adipocytes. After 8 months on the same 60% fat diet, body fat of transgenic mice was 70% below WT controls. Cardiac and liver fat was elevated in the transgenics, and their hyperinsulinemia was more marked, suggesting greater insulin resistance. The aP2-Lepr-b transgene also prevented obesity in db/db mice; at 10 weeks of age their body fat was half that of the db/db mice. This lack of obesity was attributable to reduced expression of sterol regulatory element binding protein-1c and its target lipogenic enzymes in adipose tissue and a 6-fold increase in Pref-1 mRNA. Severe diabetes was present in transgenics at 4 weeks of age, 10 weeks before db/db controls. Echocardiographic evidence of cardiomyopathy appeared at 10 weeks, weeks before the db/db mice. Histologically, loss of beta cells and myocardial fibrosis was present in the transgenic group at least 6 weeks before the db/db mice. These results suggest that the expression level of genes that regulate the adipogenic response to overnutrition profoundly influences the age of onset and severity of diet-induced type 2 diabetes and co-morbidities.


Asunto(s)
Adipogénesis/genética , Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad/genética , Síndrome Metabólico/genética , Obesidad/genética , Receptores de Leptina/genética , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Proteínas de Unión al Calcio , Cardiomiopatías/genética , Cardiomiopatías/patología , Glucagón/análisis , Glucagón/metabolismo , Insulina/análisis , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Transgénicos , Páncreas/química , Páncreas/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transgenes
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda