Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Diabetologia ; 59(6): 1231-41, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27003683

RESUMEN

AIMS/HYPOTHESIS: Understanding the developmental biology of beta cell regeneration is critical for developing new diabetes therapies. Obesity is a potent but poorly understood stimulus for beta cell expansion. Current models of obesity are complicated by developmental compensation or concurrent diabetes, limiting their usefulness for identifying the lineage mechanism(s) of beta cell expansion. We aimed to determine whether acute inducible obesity stimulates beta cell expansion and to determine the lineage mechanism of beta cell growth in obesity. METHODS: We created whole-body tamoxifen-inducible leptin receptor (LepR)-deficient mice (Ubc-Cre (ERT2) LepR (loxP/loxP) ) as a novel model of acute obesity. Beta cell mass and proliferation were quantified after short-term LepR deletion. Clonal analysis of beta cell expansion using the Brainbow2.1 reporter was performed 6 months post tamoxifen initiation. RESULTS: LepR deficiency induced a doubling of body mass within 3 weeks, with moderate glucose intolerance (unlike typical LepR mutant mice [db/db], which have frank diabetes). Beta cell mass expanded threefold through increased beta cell proliferation, without evidence for contribution from specialised progenitors or stem cells (via sequential thymidine labelling and Brainbow2.1 reporter). Thus, self-renewal is the primary lineage mechanism in obesity-induced beta cell expansion. However, even the rapid beta cell proliferation could not exceed the restrictions of the replication refractory period. CONCLUSIONS/INTERPRETATION: In summary, we created a novel model of inducible obesity demonstrating that even extreme metabolic demand does not alter beta cell lineage.


Asunto(s)
Células Secretoras de Insulina/patología , Obesidad/patología , Animales , Proliferación Celular/genética , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Obesidad/genética , Receptores de Leptina/deficiencia , Receptores de Leptina/genética
2.
Dev Cell ; 12(5): 817-26, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17488631

RESUMEN

Cellular progenitors remain poorly characterized in many adult tissues, limited in part by the lack of unbiased techniques to identify progenitors and their progeny. To address this fundamental problem, we developed a novel DNA analog-based lineage-tracing technique to detect multiple rounds of cell division in vivo. Here, we apply this technique to determine the adult lineage mechanism of the insulin-secreting beta cells of pancreatic islets, an important unresolved question in diabetes research. As expected, gastrointestinal and skin epithelia involve specialized progenitors that repeatedly divide to give rise to postmitotic cells. In contrast, specialized progenitors do not contribute to adult beta cells, not even during acute beta cell regeneration. Instead, beta cells are the products of uniform self-renewal, slowed by a replication refractory period that prevents beta cells from immediately redividing. Our approach provides unbiased resolution of previously inaccessible developmental niches and can elucidate lineage mechanisms without candidate markers.


Asunto(s)
Células Secretoras de Insulina/citología , Regeneración , Animales , Linaje de la Célula , Proliferación Celular , Segregación Cromosómica , Replicación del ADN , Células Epiteliales/citología , Femenino , Tracto Gastrointestinal/citología , Idoxuridina/metabolismo , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Mitosis , Piel/citología , Células Madre/citología , Timidina/análogos & derivados
3.
J Biol Chem ; 285(51): 40050-9, 2010 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-20943662

RESUMEN

The calcium-regulated phosphatase calcineurin intersects with both calcium and cAMP-mediated signaling pathways in the pancreatic ß-cell. Pharmacologic calcineurin inhibition, necessary to prevent rejection in the setting of organ transplantation, is associated with post-transplant ß-cell failure. We sought to determine the effect of calcineurin inhibition on ß-cell replication and survival in rodents and in isolated human islets. Further, we assessed whether the GLP-1 receptor agonist and cAMP stimulus, exendin-4 (Ex-4), could rescue ß-cell replication and survival following calcineurin inhibition. Following treatment with the calcineurin inhibitor tacrolimus, human ß-cell apoptosis was significantly increased. Although we detected no human ß-cell replication, tacrolimus significantly decreased rodent ß-cell replication. Ex-4 nearly normalized both human ß-cell survival and rodent ß-cell replication when co-administered with tacrolimus. We found that tacrolimus decreased Akt phosphorylation, suggesting that calcineurin could regulate replication and survival via the PI3K/Akt pathway. We identify insulin receptor substrate-2 (Irs2), a known cAMP-responsive element-binding protein target and upstream regulator of the PI3K/Akt pathway, as a novel calcineurin target in ß-cells. Irs2 mRNA and protein are decreased by calcineurin inhibition in both rodent and human islets. The effect of calcineurin on Irs2 expression is mediated at least in part through the nuclear factor of activated T-cells (NFAT), as NFAT occupied the Irs2 promoter in a calcineurin-sensitive manner. Ex-4 restored Irs2 expression in tacrolimus-treated rodent and human islets nearly to baseline. These findings reveal calcineurin as a regulator of human ß-cell survival in part through regulation of Irs2, with implications for the pathogenesis and treatment of diabetes following organ transplantation.


Asunto(s)
Calcineurina/farmacología , Proliferación Celular/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Calcineurina/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Diabetes Mellitus/etiología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Exenatida , Regulación de la Expresión Génica/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón , Humanos , Hipoglucemiantes/farmacología , Inmunosupresores/farmacología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Factores de Transcripción NFATC/metabolismo , Trasplante de Órganos/efectos adversos , Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Tacrolimus/farmacología , Ponzoñas/farmacología
4.
Front Endocrinol (Lausanne) ; 12: 652628, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054727

RESUMEN

The glucagon-like peptide-1 receptor (GLP-1R) is a G-protein-coupled receptor (GPCR) whose activation results in suppression of food intake and improvement of glucose metabolism. Several receptor interacting proteins regulate the signaling of GLP-1R such as G protein-coupled receptor kinases (GRK) and ß-arrestins. Here we evaluated the physiological and pharmacological impact of GRK inhibition on GLP-1R activity leveraging small molecule inhibitors of GRK2 and GRK3. We demonstrated that inhibition of GRK: i) inhibited GLP-1-mediated ß-arrestin recruitment, ii) enhanced GLP-1-induced insulin secretion in isolated islets and iii) has additive effect with dipeptidyl peptidase 4 in mediating suppression of glucose excursion in mice. These findings highlight the importance of GRK to modulate GLP-1R function in vitro and in vivo. GRK inhibition is a potential therapeutic approach to enhance endogenous and pharmacologically stimulated GLP-1R signaling.


Asunto(s)
Quinasa 1 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Péptido 1 Similar al Glucagón/metabolismo , Amidas/química , Animales , Células CHO , Calcio/metabolismo , Cricetulus , Diabetes Mellitus/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Ingestión de Alimentos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Obesidad/metabolismo , Fosforilación , Receptores de Glucagón/metabolismo , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , beta-Arrestinas/metabolismo
5.
PLoS One ; 14(9): e0214829, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31490929

RESUMEN

Tamoxifen is a mixed agonist/antagonist estrogen analogue that is frequently used to induce conditional gene deletion in mice using Cre-loxP mediated gene recombination. Tamoxifen is routinely employed in extremely high-doses relative to typical human doses to induce efficient gene deletion in mice. Although tamoxifen has been widely assumed to have no influence upon ß-cells, the acute developmental and functional consequences of high-dose tamoxifen upon glucose homeostasis and adult ß-cells are largely unknown. We tested if tamoxifen influences glucose homeostasis in male mice of various genetic backgrounds. We then carried out detailed histomorphometry studies of mouse pancreata. We also performed gene expression studies with islets of tamoxifen-treated mice and controls. Tamoxifen had modest effects upon glucose homeostasis of mixed genetic background (F1 B6129SF1/J) mice, with fasting hyperglycemia and improved glucose tolerance but without overt effects on fed glucose levels or insulin sensitivity. Tamoxifen inhibited proliferation of ß-cells in a dose-dependent manner, with dramatic reductions in ß-cell turnover at the highest dose (decreased by 66%). In sharp contrast, tamoxifen did not reduce proliferation of pancreatic acinar cells. ß-cell proliferation was unchanged by tamoxifen in 129S2 mice but was reduced in C57Bl6 genetic background mice (decreased by 59%). Gene expression studies revealed suppression of RNA for cyclins D1 and D2 within islets of tamoxifen-treated mice. Tamoxifen has a cytostatic effect on ß-cells, independent of changes in glucose homeostasis, in mixed genetic background and also in C57Bl6 mice. Tamoxifen should be used judiciously to inducibly inactivate genes in studies of glucose homeostasis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Tamoxifeno/farmacología , Células Acinares/efectos de los fármacos , Células Acinares/fisiología , Animales , Células Cultivadas , Ciclina D/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Diabetes ; 68(5): 963-974, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30833466

RESUMEN

Glucagon-containing α-cells potently regulate glucose homeostasis, but the developmental biology of α-cells in adults remains poorly understood. Although glucagon receptor antagonists (GRAs) have great potential as antidiabetic therapies, murine and human studies have raised concerns that GRAs might cause uncontrolled α-cell growth. Surprisingly, previous rodent GRA studies were only performed in young mice, implying that the potential impact of GRAs to drive α-cell expansion in adult patients is unclear. We assessed adaptive α-cell turnover and adaptive proliferation, administering a novel GRA (JNJ-46207382) to both young and aged mice. Basal α-cell proliferation rapidly declined soon after birth and continued to drop to very low levels in aged mice. GRA drove a 2.4-fold increase in α-cell proliferation in young mice. In contrast, GRA-induced α-cell proliferation was severely reduced in aged mice, although still present at 3.2-fold the very low basal rate of aged controls. To interrogate the lineage of GRA-induced α-cells, we sequentially administered thymidine analogs and quantified their incorporation into α-cells. Similar to previous studies of ß-cells, α-cells only divided once in both basal and stimulated conditions. Lack of contribution from highly proliferative "transit-amplifying" cells supports a model whereby α-cells expand by self-renewal and not via specialized progenitors.


Asunto(s)
Envejecimiento/fisiología , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/metabolismo , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/metabolismo , Animales , Células Secretoras de Glucagón/citología , Hipoglucemiantes/efectos adversos , Masculino , Ratones , Timidina/efectos adversos , Timidina/análogos & derivados
7.
Artículo en Inglés | MEDLINE | ID: mdl-29883889

RESUMEN

Sulprostone is a potent prostaglandin E2 (PGE2) analogue and one of the first identified selective G-protein-coupled receptor 3 (EP3) agonists. It has been investigated as a potential antiulcer agent and frequently used in the research of EP3 antagonist. To assist pharmacokinetic and pharmacodynamic studies, a rapid and sensitive LC-MS/MS method was developed and qualified for the quantitation of sulprostone in monkey plasma. Using electrospray ionization mass spectrometry, an ammonium adduct in positive mode was chosen for analysis which had seven times of the sensitivity of the depronated ion in negative mode. Latanoprost, a prostaglandin F2α analogue, was used as the internal standard while good sensitivity and chromatography were obtained on a 2.6 µm core-shell column with pentafluorophenyl stationary phase. An assay dynamic range of 2 to 4000 ng/mL was achieved with a sample volume of 25 µL plasma on a Sciex API4000 instrument with simple protein precipitation. Several esterase inhibitors including sodium fluoride (NaF), phenylmethanesulfonyl fluoride (PMSF), diisopropylfluorophosphate (DFP), paraoxon and dichlorvos as well as wet ice conditions were explored for the stabilization of sulprostone in monkey plasma. The developed method was successfully applied for the evaluation of pharmacokinetics of sulprostone after intravenous administration of 0.5 mg/kg to cynomolgus monkey.


Asunto(s)
Cromatografía Liquida/métodos , Dinoprostona/análogos & derivados , Espectrometría de Masas en Tándem/métodos , Animales , Dinoprostona/sangre , Dinoprostona/química , Dinoprostona/farmacocinética , Estabilidad de Medicamentos , Modelos Lineales , Macaca fascicularis , Masculino , Subtipo EP3 de Receptores de Prostaglandina E/agonistas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
8.
Diabetes ; 67(4): 674-686, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29326366

RESUMEN

The proliferative response of non-ß islet endocrine cells in response to type 1 diabetes (T1D) remains undefined. We quantified islet endocrine cell proliferation in a large collection of nondiabetic control and T1D human pancreata across a wide range of ages. Surprisingly, islet endocrine cells with abundant proliferation were present in many adolescent and young-adult T1D pancreata. But the proliferative islet endocrine cells were also present in similar abundance within control samples. We queried the proliferating islet cells with antisera against various islet hormones. Although pancreatic polypeptide, somatostatin, and ghrelin cells did not exhibit frequent proliferation, glucagon-expressing α-cells were highly proliferative in many adolescent and young-adult samples. Notably, α-cells only comprised a fraction (∼1/3) of the proliferative islet cells within those samples; most proliferative cells did not express islet hormones. The proliferative hormone-negative cells uniformly contained immunoreactivity for ARX (indicating α-cell fate) and cytoplasmic Sox9 (Sox9Cyt). These hormone-negative cells represented the majority of islet endocrine Ki67+ nuclei and were conserved from infancy through young adulthood. Our studies reveal a novel population of highly proliferative ARX+ Sox9Cyt hormone-negative cells and suggest the possibility of previously unrecognized islet development and/or lineage plasticity within adolescent and adult human pancreata.


Asunto(s)
Proliferación Celular , Diabetes Mellitus Tipo 1 , Células Secretoras de Glucagón/citología , Adolescente , Adulto , Estudios de Casos y Controles , Linaje de la Célula , Plasticidad de la Célula , Niño , Preescolar , Femenino , Ghrelina/metabolismo , Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Lactante , Recién Nacido , Islotes Pancreáticos/citología , Antígeno Ki-67/metabolismo , Masculino , Polipéptido Pancreático/metabolismo , Factor de Transcripción SOX9/metabolismo , Somatostatina/metabolismo , Factores de Transcripción/metabolismo , Adulto Joven
9.
J Clin Endocrinol Metab ; 102(8): 2647-2659, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28323930

RESUMEN

Context: The cellular basis of persistent ß-cell function in type 1 diabetes (T1D) remains enigmatic. No extensive quantitative ß-cell studies of T1D pancreata have been performed to test for ongoing ß-cell regeneration or neogenesis. Objective: We sought to determine the mechanism of ß-cell persistence in T1D pancreata. Design: We studied T1D (n = 47) and nondiabetic control (n = 59) pancreata over a wide range of ages from the Juvenile Diabetes Research Foundation Network of Pancreatic Organ Donors with Diabetes via high-throughput microscopy. Intervention and Main Outcome Measures: We quantified ß-cell mass, ß-cell turnover [via Ki-67 and terminal deoxynucleotide transferase-mediated dUTP nick end labeling (TUNEL)], islet ductal association, and insulin/glucagon coexpression in T1D and control pancreata. Results: Residual insulin-producing ß cells were detected in some (but not all) T1D cases of varying disease duration. Several T1D pancreata had substantial numbers of ß cells. Although ß-cell proliferation was prominent early in life, it dramatically declined after infancy in both nondiabetic controls and T1D individuals. However, ß-cell proliferation was equivalent in control and T1D pancreata. ß-cell death (assessed by TUNEL) was extremely rare in control and T1D pancreata. Thus, ß-cell turnover was not increased in T1D. Furthermore, we found no evidence of small islet/ductal neogenesis or α-cell to ß-cell transdifferentiation in T1D pancreata, regardless of disease duration. Conclusion: Longstanding ß-cell function in patients with T1D appears to be largely a result of ß cells that persist, without any evidence of attempted ß-cell regeneration, small islet/ductal neogenesis, or transdifferentiation from other islet endocrine cell types.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Transdiferenciación Celular , Niño , Preescolar , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Glucagón/metabolismo , Células Secretoras de Glucagón , Humanos , Etiquetado Corte-Fin in Situ , Lactante , Recién Nacido , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Páncreas/citología , Páncreas/metabolismo , Regeneración , Factores de Tiempo , Adulto Joven
10.
Endocrinology ; 158(6): 1701-1714, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28323942

RESUMEN

The impact of incretins upon pancreatic ß-cell expansion remains extremely controversial. Multiple studies indicate that incretin-based therapies can increase ß-cell proliferation, and incretins have been hypothesized to expand ß-cell mass. However, disagreement exists on whether incretins increase ß-cell mass. Moreover, some reports indicate that incretins may cause metaplastic changes in pancreatic histology. To resolve these questions, we treated a large cohort of mice with incretin-based therapies and carried out a rigorous analysis of ß-cell turnover and pancreatic histology using high-throughput imaging. Young mice received exenatide via osmotic pump, des-fluoro-sitagliptin, or glipizide compounded in diet for 2 weeks (short-term) on a low-fat diet (LFD) or 4.5 months (long-term) on a LFD or high-fat diet (HFD). Pancreata were quantified for ß-cell turnover and mass. Slides were examined for gross anatomical and microscopic changes to exocrine pancreas. Short-term des-fluoro-sitagliptin increased serum insulin and induced modest ß-cell proliferation but no change in ß-cell mass. Long-term incretin therapy in HFD-fed mice resulted in reduced weight gain, improved glucose homeostasis, and abrogated ß-cell mass expansion. No evidence for rapidly dividing progenitor cells was found in islets or pancreatic parenchyma, indicating that incretins do not induce islet neogenesis or pancreatic metaplasia. Contrasting prior reports, we found no evidence of ß-cell mass expansion after acute or chronic incretin therapy. Chronic incretin administration was not associated with histological abnormalities in pancreatic parenchyma; mice did not develop tumors, pancreatitis, or ductal hyperplasia. We conclude that incretin therapies do not generate ß-cells or alter pancreatic histology in young mice.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Incretinas/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Páncreas/efectos de los fármacos , Páncreas/patología , Animales , Recuento de Células , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Exenatida , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos/uso terapéutico , Fosfato de Sitagliptina/uso terapéutico , Ponzoñas/uso terapéutico
11.
Diabetes ; 54(9): 2557-67, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16123343

RESUMEN

Although many signaling pathways have been shown to promote beta-cell growth, surprisingly little is known about the normal life cycle of preexisting beta-cells or the signaling pathways required for beta-cell survival. Adult beta-cells have been speculated to have a finite life span, with ongoing adult beta-cell replication throughout life to replace lost cells. However, little solid evidence supports this idea. To more accurately measure adult beta-cell turnover, we performed continuous long-term labeling of proliferating cells with the DNA precursor analog 5-bromo-2-deoxyuridine (BrdU) in 1-year-old mice. We show that beta-cells of aged adult mice have extremely low rates of replication, with minimal evidence of turnover. Although some pancreatic components acquired BrdU label in a linear fashion, only 1 in approximately 1,400 adult beta-cells were found to undergo replication per day. We conclude that adult beta-cells are very long lived.


Asunto(s)
Envejecimiento/fisiología , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Animales , Bromouracilo/análogos & derivados , Muerte Celular , Proliferación Celular/efectos de los fármacos , Femenino , Glucagón/metabolismo , Masculino , Ratones , Polipéptido Pancreático/metabolismo , Somatostatina/metabolismo , Estreptozocina/farmacología , Uridina/análogos & derivados
12.
PLoS One ; 10(6): e0129809, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26057531

RESUMEN

The pathophysiology of canine diabetes remains poorly understood, in part due to enigmatic clinical features and the lack of detailed histopathology studies. Canine diabetes, similar to human type 1 diabetes, is frequently associated with diabetic ketoacidosis at onset or after insulin omission. However, notable differences exist. Whereas human type 1 diabetes often occurs in children, canine diabetes is typically described in middle age to elderly dogs. Many competing theories have been proposed regarding the underlying cause of canine diabetes, from pancreatic atrophy to chronic pancreatitis to autoimmune mediated ß-cell destruction. It remains unclear to what extent ß-cell loss contributes to canine diabetes, as precise quantifications of islet morphometry have not been performed. We used high-throughput microscopy and automated image processing to characterize islet histology in a large collection of pancreata of diabetic dogs. Diabetic pancreata displayed a profound reduction in ß-cells and islet endocrine cells. Unlike humans, canine non-diabetic islets are largely comprised of ß-cells. Very few ß-cells remained in islets of diabetic dogs, even in pancreata from new onset cases. Similarly, total islet endocrine cell number was sharply reduced in diabetic dogs. No compensatory proliferation or lymphocyte infiltration was detected. The majority of pancreata had no evidence of pancreatitis. Thus, canine diabetes is associated with extreme ß-cell deficiency in both new and longstanding disease. The ß-cell predominant composition of canine islets and the near-total absence of ß-cells in new onset elderly diabetic dogs strongly implies that similar to human type 1 diabetes, ß-cell loss underlies the pathophysiology of canine diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/veterinaria , Células Secretoras de Insulina/patología , Animales , Proliferación Celular , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/patología , Perros , Femenino , Glucagón/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/patología , Hiperglucemia/veterinaria , Insulina/metabolismo , Cetosis/complicaciones , Cetosis/patología , Cetosis/veterinaria , Linfocitos/inmunología , Masculino , Tamaño de los Órganos
13.
Mol Endocrinol ; 28(1): 28-39, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24284823

RESUMEN

Pancreatic ß-cell survival remains poorly understood despite decades of research. GATA transcription factors broadly regulate embryogenesis and influence survival of several cell types, but their role in adult ß-cells remains undefined. To investigate the role of GATA factors in adult ß-cells, we derived ß-cell-inducible Gata4- and Gata6-knockout mice, along with whole-body inducible Gata4 knockouts. ß-Cell Gata4 deletion modestly increased the proportion of dying ß-cells in situ with ultrastructural abnormalities suggesting endoplasmic reticulum (ER) stress. Notably, glucose homeostasis was not grossly altered in Gata4- and Gata6-knockout mice, suggesting that GATA factors do not have essential roles in ß-cells. Several ER stress signals were up-regulated in Gata4 and Gata6 knockouts, most notably CHOP, a known regulator of ER stress-induced apoptosis. However, ER stress signals were not elevated to levels observed after acute thapsigargin administration, suggesting that GATA deficiency only caused mild ER stress. Simultaneous deletion of Gata4 and CHOP partially restored ß-cell survival. In contrast, whole-body inducible Gata4 knockouts displayed no evidence of ER stress in other GATA4-enriched tissues, such as heart. Indeed, distinct GATA transcriptional targets were differentially expressed in islets compared with heart. Such ß-cell-specific findings prompted study of a large meta-analysis dataset to investigate single nucleotide polymorphisms harbored within the human GATA4 locus, revealing several variants significantly associated with type 1 diabetes mellitus. We conclude that GATA factors have important but nonessential roles to promote ER integrity and ß-cell survival in a tissue-specific manner and that GATA factors likely contribute to type 1 diabetes mellitus pathogenesis.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Retículo Endoplásmico/fisiología , Factor de Transcripción GATA4/fisiología , Células Secretoras de Insulina/fisiología , Animales , Apoptosis , Estudios de Casos y Controles , Supervivencia Celular , Células Cultivadas , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Retículo Endoplásmico/patología , Factor de Transcripción GATA6/fisiología , Regulación de la Expresión Génica , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Ratones Noqueados , Páncreas/patología , Polimorfismo de Nucleótido Simple , Riesgo , Factor de Transcripción CHOP/metabolismo , Transcripción Genética
14.
Diabetes ; 62(5): 1634-45, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23349489

RESUMEN

The existence of adult ß-cell progenitors remains the most controversial developmental biology topic in diabetes research. It has been reported that ß-cell progenitors can be activated by ductal ligation-induced injury of adult mouse pancreas and apparently act in a cell-autonomous manner to double the functional ß-cell mass within a week by differentiation and proliferation. Here, we demonstrate that pancreatic duct ligation (PDL) does not activate progenitors to contribute to ß-cell mass expansion. Rather, PDL stimulates massive pancreatic injury, which alters pancreatic composition and thus complicates accurate measurement of ß-cell content via traditional morphometry methodologies that superficially sample the pancreas. To overcome this potential bias, we quantified ß-cells from the entire pancreas and observed that ß-cell mass and insulin content are totally unchanged by PDL-induced injury. Lineage-tracing studies using sequential administration of thymidine analogs, rat insulin 2 promoter-driven cre-lox, and low-frequency ubiquitous cre-lox reveal that PDL does not convert progenitors to the ß-cell lineage. Thus, we conclude that ß-cells are not generated in injured adult mouse pancreas.


Asunto(s)
Células Madre Adultas/citología , Diferenciación Celular , Células Secretoras de Insulina/citología , Células Madre Adultas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores , Linaje de la Célula , Proliferación Celular , Cruzamientos Genéticos , Regulación de la Expresión Génica , Genes Reporteros , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ligadura , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Páncreas/citología , Páncreas/metabolismo , Conductos Pancreáticos/cirugía , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo
15.
Islets ; 2(6): 345-52, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21099336

RESUMEN

The role of aging in the pathogenesis of type 2 diabetes remains poorly understood. In the past adult ß-cells were assumed to undergo frequent turnover. However, we find that ß-cell turnover declines to very low levels in middle-aged mice. We therefore hypothesized that aged islets could exhibit a distinct gene expression program. We compared gene expression in islets from young mice to islets from aged mice under basal conditions. Aging was associated with differential expression of many genes in islets, including mRNAs encoding for chromatin remodeling components, RNA binding proteins, and pancreatic endocrine transcription factors. We previously observed that cell cycle entry of ß-cells is severely restricted by middle age, with minimal of ß-cell proliferation in response to regenerative stimuli such as 50% partial pancreatectomy. To characterize the effect of age in adaptive ß-cell proliferation, we measured gene expression in islets from young mice after pancreatectomy. As expected, partial pancreatectomy induced differential expression of many genes, including those encoding Reg (regenerating) proteins. Surprisingly, partial pancreatectomy also induced expression of Reg genes in islets from aged mice, which have greatly reduced capacity for adaptive ß-cell proliferation. However, there was little overlap (besides the Reg genes) in between the partial pancreatectomy induced islet genes in young mice versus old mice. Thus, partial pancreatectomy does not induce the same gene expression program in young mice vs old mice. Taken together, our results reveal that aged islets exhibit a unique gene expression signature that could contribute to the limited regenerative capacity of mature ß-cells.


Asunto(s)
Envejecimiento/metabolismo , Regulación del Desarrollo de la Expresión Génica , Islotes Pancreáticos/metabolismo , Animales , Proliferación Celular , Ensamble y Desensamble de Cromatina , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Páncreas/fisiología , Pancreatectomía , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Regeneración , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
J Vis Exp ; (46)2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21178965

RESUMEN

Accurate measurement of cell division is a fundamental challenge in experimental biology that becomes increasingly complex when slowly dividing cells are analyzed. Established methods to detect cell division include direct visualization by continuous microscopy in cell culture, dilution of vital dyes such as carboxyfluorescein di-aetate succinimidyl ester (CFSE), immuno-detection of mitogenic antigens such as ki67 or PCNA, and thymidine analogues. Thymidine analogues can be detected by a variety of methods including radio-detection for tritiated thymidine, immuno-detection for bromo-deoxyuridine (BrdU), chloro-deoxyuridine (CldU) and iodo-deoxyuridine (IdU), and chemical detection for ethinyl-deoxyuridine (EdU). We have derived a strategy to detect sequential incorporation of different thymidine analogues (CldU and IdU) into tissues of adult mice. Our method allows investigators to accurately quantify two successive rounds of cell division. By optimizing immunostaining protocols our approach can detect very low dose thymidine analogues administered via the drinking water, safe to administer to mice for prolonged periods of time. Consequently, our technique can be used to detect cell turnover in very long-lived tissues. Optimal immunofluoresent staining results can be achieved in multiple tissue types, including pancreas, skin, gut, liver, adrenal, testis, ovary, thyroid, lymph node, and brain. We have also applied this technique to identify oncogenic transformation within tissues. We have further applied this technique to determine if transit-amplifying cells contribute to growth or renewal of tissues. In this sense, sequential administration of thymidine analogues represents a novel approach for studying the origins and survival of cells involved in tissue homeostasis.


Asunto(s)
Desoxiuridina/análogos & derivados , Idoxuridina/análisis , Microscopía Fluorescente/métodos , Timidina/análogos & derivados , Animales , Desoxiuridina/análisis , Femenino , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Páncreas/química , Timidina/análisis
17.
Diabetes ; 58(6): 1365-72, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19265026

RESUMEN

OBJECTIVE: Regeneration of the insulin-secreting beta-cells is a fundamental research goal that could benefit patients with either type 1 or type 2 diabetes. beta-Cell proliferation can be acutely stimulated by a variety of stimuli in young rodents. However, it is unknown whether this adaptive beta-cell regeneration capacity is retained into old age. RESEARCH DESIGN AND METHODS: We assessed adaptive beta-cell proliferation capacity in adult mice across a wide range of ages with a variety of stimuli: partial pancreatectomy, low-dose administration of the beta-cell toxin streptozotocin, and exendin-4, a glucagon-like peptide 1 (GLP-1) agonist. beta-Cell proliferation was measured by administration of 5-bromo-2'-deoxyuridine (BrdU) in the drinking water. RESULTS: Basal beta-cell proliferation was severely decreased with advanced age. Partial pancreatectomy greatly stimulated beta-cell proliferation in young mice but failed to increase beta-cell replication in old mice. Streptozotocin stimulated beta-cell replication in young mice but had little effect in old mice. Moreover, administration of GLP-1 agonist exendin-4 stimulated beta-cell proliferation in young but not in old mice. Surprisingly, adaptive beta-cell proliferation capacity was minimal after 12 months of age, which is early middle age for the adult mouse life span. CONCLUSIONS: Adaptive beta-cell proliferation is severely restricted with advanced age in mice, whether stimulated by partial pancreatectomy, low-dose streptozotocin, or exendin-4. Thus, beta-cells in middle-aged mice appear to be largely postmitotic. Young rodents may not faithfully model the regenerative capacity of beta-cells in mature adult mice.


Asunto(s)
Envejecimiento/fisiología , División Celular/fisiología , Células Secretoras de Insulina/citología , Aclimatación , Animales , Femenino , Células Secretoras de Insulina/fisiología , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Pancreatectomía
18.
Mol Endocrinol ; 23(11): 1865-75, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19628581

RESUMEN

The molecular determinants of beta-cell mass expansion remain poorly understood. Cyclin D2 is the major D-type cyclin expressed in beta-cells, essential for adult beta-cell growth. We hypothesized that cyclin D2 could be actively regulated in beta-cells, which could allow mitogenic stimuli to influence beta-cell expansion. Cyclin D2 protein was sharply increased after partial pancreatectomy, but cyclin D2 mRNA was unchanged, suggesting posttranscriptional regulatory mechanisms influence cyclin D2 expression in beta-cells. Consistent with this hypothesis, cyclin D2 protein stability is powerfully regulated in fibroblasts. Threonine 280 of cyclin D2 is phosphorylated, and this residue critically limits D2 stability. We derived transgenic (tg) mice with threonine 280 of cyclin D2 mutated to alanine (T280A) or wild-type cyclin D2 under the control of the insulin promoter. Cyclin D2 T280A protein was expressed at much higher levels than wild-type cyclin D2 protein in beta-cells, despite equivalent expression of tg mRNAs. Cyclin D2 T280A tg mice exhibited a constitutively nuclear cyclin D2 localization in beta-cells, and increased cyclin D2 stability in islets. Interestingly, threonine 280-mutant cyclin D2 tg mice had greatly reduced beta-cell apoptosis, with suppressed expression of proapoptotic genes. Suppressed beta-cell apoptosis in threonine 280-mutant cyclin D2 tg mice resulted in greatly increased beta-cell area in aged mice. Taken together, these data indicate that cyclin D2 is regulated by protein stability in pancreatic beta-cells, that signals that act upon threonine 280 limit cyclin D2 stability in beta-cells, and that threonine 280-mutant cyclin D2 overexpression prolongs beta-cell survival and augments beta-cell mass expansion.


Asunto(s)
Ciclina D2/fisiología , Regulación de la Expresión Génica , Células Secretoras de Insulina/citología , Alanina/genética , Animales , Apoptosis , Cruzamientos Genéticos , Ciclina D2/metabolismo , Exenatida , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Mutación , Péptidos/metabolismo , ARN Mensajero/metabolismo , Treonina/genética , Ponzoñas/metabolismo
19.
J Biol Chem ; 280(47): 39388-93, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16170201

RESUMEN

The Irs2 branch of the insulin/insulin-like growth factor signaling cascade activates the phosphatidylinositol 3-kinase --> Akt --> Foxo1 cascade in many tissues, including hepatocytes and pancreatic beta-cells. The 3'-lipid phosphatase Pten ordinarily attenuates this cascade; however, its influence on beta-cell growth or function is unknown. To determine whether decreased Pten expression could restore beta-cell function and prevent diabetes in Irs2(-/-) mice, we generated wild type or Irs2 knock-out mice that were haploinsufficient for Pten (Irs2(-/-)::Pten(+/-)). Irs2(-/-) mice develop diabetes by 3 months of age as beta-cell mass declined progressively until insulin production was lost. Pten insufficiency increased peripheral insulin sensitivity in wild type and Irs2(-/-) mice and increased Akt and Foxo1 phosphorylation in the islets. Glucose tolerance improved in the Pten(+/-) mice, although beta-cell mass and circulating insulin levels decreased. Compared with Irs2(-/-) mice, the Irs2(-/-)::Pten(+/-) mice displayed nearly normal glucose tolerance and survived without diabetes, because normal but small islets produced sufficient insulin until the mice died of lymphoproliferative disease at 12 months age. Thus, steps to enhance phosphatidylinositol 3-kinase signaling can promote beta-cell growth, function, and survival without the Irs2 branch of the insulin/insulin-like growth factor signaling cascade.


Asunto(s)
Glucosa/metabolismo , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Fosfohidrolasa PTEN/fisiología , Animales , Proliferación Celular , Femenino , Homeostasis , Proteínas Sustrato del Receptor de Insulina , Resistencia a la Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosfoproteínas/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA