Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Am J Pathol ; 192(1): 130-145, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34619135

RESUMEN

High circulating lipids occurring in obese individuals and insulin-resistant patients are considered a contributing factor to type 2 diabetes. Exposure to high lipid concentration is proposed to both protect and damage beta-cells under different circumstances. Here, by feeding mice a high-fat diet (HFD) for 2 weeks to up to 14 months, the study showed that HFD initially causes the beta-cells to expand in population, whereas long-term exposure to HFD is associated with failure of beta-cells and the inability of animals to respond to glucose challenge. To prevent the failure of beta-cells and the development of type 2 diabetes, the molecular mechanisms that underlie this biphasic response of beta-cells to lipid exposure were explored. Using palmitic acid (PA) in cultured beta-cells and islets, the study demonstrated that chronic exposure to lipids leads to reduced viability and inhibition of cell cycle progression concurrent with down-regulation of a pro-growth/survival kinase AKT, independent of glucose. This AKT down-regulation by PA is correlated with the induction of mTOR/S6K activity. Inhibiting mTOR activity with rapamycin induced Raptor and restored AKT activity, allowing beta-cells to gain proliferation capacity that was lost after HFD exposure. In summary, a novel mechanism in which lipid exposure may cause the dipole effects on beta-cell growth was elucidated, where mTOR acts as a lipid sensor. These mechanisms can be novel targets for future therapeutic developments.


Asunto(s)
Regulación hacia Abajo , Células Secretoras de Insulina/enzimología , Ácido Palmítico/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina D2/metabolismo , Dieta Alta en Grasa , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Fosfohidrolasa PTEN/metabolismo , Fosforilación/efectos de los fármacos , Proteína Reguladora Asociada a mTOR/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
2.
Biologicals ; 78: 17-26, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35840492

RESUMEN

This online workshop Accelerating Global Deletion of the Abnormal Toxicity Test for vaccines and biologicals. Planning common next steps was organized on October 14th, 2021, by the Animal Free Safety Assessment Collaboration (AFSA), the Humane Society International (HSI), the European Federation of Pharmaceutical Industries and Associations (EFPIA), in collaboration with the International Alliance of Biological Standardization (IABS). The workshop saw a participation of over a hundred representatives from international organizations, pharmaceutical industries and associations, and regulatory authorities of 28 countries. Participants reported on country- and region-specific regulatory requirements and, where present, on the perspectives on the waiving and elimination of the Abnormal Toxicity Test. With AFSA, HSI, EFPIA and IABS representatives as facilitators, the participants also discussed specific country/global actions to further secure the deletion of ATT from all regulatory requirements worldwide.


Asunto(s)
Pruebas de Toxicidad , Vacunas , Industria Farmacéutica , Humanos , Estándares de Referencia , Vacunas/efectos adversos
3.
Diabetologia ; 57(2): 352-61, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24162585

RESUMEN

AIMS/HYPOTHESIS: Adult beta cells have a diminished ability to proliferate. Phosphatase and tensin homologue (PTEN) is a lipid phosphatase that antagonises the function of the mitogenic phosphatidylinositol 3-kinase (PI3K) pathway. The objective of this study was to understand the role of PTEN and PI3K signalling in the maintenance of beta cells postnatally. METHODS: We developed a Pten (lox/lox); Rosa26 (lacZ); RIP-CreER (+) model that permitted us to induce Pten deletion by treatment with tamoxifen in mature animals. We evaluated islet mass and function as well as beta cell proliferation in 3- and 12-month-old mice treated with tamoxifen (Pten deleted) vs mice treated with vehicle (Pten control). RESULTS: Deletion of Pten in juvenile (3-month-old) beta cells significantly induced their proliferation and increased islet mass. The expansion of islet mass occurred concomitantly with the enhanced ability of the Pten-deleted mice to maintain euglycaemia in response to streptozotocin treatment. In older mice (>12 months of age), deletion of Pten similarly increased islet mass and beta cell proliferation. This novel finding suggests that PTEN-regulated mechanisms may override the age-onset diminished ability of beta cells to respond to mitogenic stimulation. We also found that proteins regulating G1/S cell-cycle transition, such as cyclin D1, cyclin D2, p27 and p16, were altered when PTEN was lost, suggesting that they may play a role in PTEN/PI3K-regulated beta cell proliferation in adult tissue. CONCLUSIONS/INTERPRETATION: The signals regulated by the PTEN/PI3K pathway are important for postnatal maintenance of beta cells and regulation of their proliferation in adult tissues.


Asunto(s)
Envejecimiento/patología , Diabetes Mellitus Experimental/patología , Células Secretoras de Insulina/metabolismo , Fosfohidrolasa PTEN/metabolismo , Animales , Ciclo Celular , Muerte Celular , Proliferación Celular , Metilación de ADN , Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo/genética , Eliminación de Gen , Homeostasis , Masculino , Ratones , Ratones Mutantes , Fosfohidrolasa PTEN/deficiencia , Transducción de Señal , Regulación hacia Arriba
4.
Mol Cell Biol ; 40(11)2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32179553

RESUMEN

Isoforms of protein kinase B (also known as AKT) play important roles in mediating insulin and growth factor signals. Previous studies have suggested that the AKT2 isoform is critical for insulin-regulated glucose metabolism, while the role of the AKT1 isoform remains less clear. This study focuses on the effects of AKT1 on the adaptive response of pancreatic ß cells. Using a mouse model with inducible ß-cell-specific deletion of the Akt1 gene (ßA1KO mice), we showed that AKT1 is involved in high-fat-diet (HFD)-induced growth and survival of ß cells but is unnecessary for them to maintain a population in the absence of metabolic stress. When unchallenged, ßA1KO mice presented the same metabolic profile and ß-cell phenotype as the control mice with an intact Akt1 gene. When metabolic stress was induced by HFD, ß cells in control mice with intact Akt1 proliferated as a compensatory mechanism for metabolic overload. Similar effects were not observed in ßA1KO mice. We further demonstrated that AKT1 protein deficiency caused endoplasmic reticulum (ER) stress and potentiated ß cells to undergo apoptosis. Our results revealed that AKT1 protein loss led to the induction of eukaryotic initiation factor 2 α subunit (eIF2α) signaling and ER stress markers under normal-chow-fed conditions, indicating chronic low-level ER stress. Together, these data established a role for AKT1 as a growth and survival factor for adaptive ß-cell response and suggest that ER stress induction is responsible for this effect of AKT1.


Asunto(s)
Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Células Secretoras de Insulina/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/genética
5.
Artículo en Inglés | MEDLINE | ID: mdl-27307790

RESUMEN

BACKGROUND: Hepatic fibrosis is a prominent pathological feature associated with chronic liver disease including non-alcoholic hepatosteatosis (NASH), and a precursor for liver cancer development. We previously reported that PTEN loss in the liver, which leads to hyperactivated liver insulin signaling results in NASH development. Here we used the same mouse model to study the progression from steatosis to fibrosis. RESULTS: The Pten null livers develop progressive liver fibrosis as indicated by Sirius Red staining and increased expression of collagen I, Timp 1, SMAα, and p75NTR. Consistently, hepatic stellate cells (HSCs) isolated from Pten null livers are readily activated when compared with that from mice with intact PTEN. Deletion of AKT2, the downstream target of PTEN signal, blocked NASH development, and alleviated fibrosis. HSCs from the Pten/Akt2 double null mice are quiescent like those isolated from the control livers. Our analysis shows that the activation of HSCs does not depend on the intrinsic signals regulated by PI3K/AKT, the target of PTEN, but does depend on steatosis and injury to the liver. During the progression of liver fibrosis in the Pten null model, Wnt ligands and signaling receptor are induced, concurrent with the reduction of sFRP5, a Wnt antagonist. We showed that treatment of HSCs with Wnt receptor antagonist blocks the observed morphological changes when HSCs undergo activation in culture. This signal appears to be mediated by ß-catenin, as manipulating ß-catenin signaling alters marker gene expressions of HSC activation. CONCLUSIONS: Wnt/ß-catenin activation serves as an important mediator for fibrosis development resulting from NASH using a mouse model where NASH is mimicked by PTEN loss.

6.
Pancreas ; 44(7): 1111-20, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26333110

RESUMEN

OBJECTIVES: In injury conditions, myofibroblasts are induced to lay down matrix proteins and support the repair process. In this study, we investigated the role of myofibroblasts, particularly stellate cells, in the growth and regeneration of pancreatic ß cells. METHODS: We used both in vitro and in vivo approaches to address whether stellate cells may promote the growth of ß cells. RESULTS: Our experiments demonstrated that activated stellate cells support the proliferation of ß cells in vitro. In vivo, mesenchymals surrounding the pancreatic islets are activated (induced to proliferate) in the islet regeneration model of Pten null mice. These mesenchymals display markers of pancreatic stellate cells, such as desmin and to a lesser extent, smooth muscle actin α. We have shown previously that targeted ß-cell deletion of Pten lead to a significant increase in total islet mass. This phenotype was accompanied by an increase in peri-islet mitotic activity, particularly in islets injured by streptozotocin, a ß cell-specific toxin. CONCLUSIONS: Together with the in vitro observations, our data, here, suggest that that these mesenchymal cells may support the regeneration of the islets. Identifying how the communication occurs may provide clinically relevant mechanism for inducing ß-cell regeneration.


Asunto(s)
Células Secretoras de Insulina/fisiología , Miofibroblastos/fisiología , Páncreas/fisiopatología , Células Estrelladas Pancreáticas/fisiología , Actinas/metabolismo , Animales , Comunicación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Desmina/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Inmunohistoquímica , Células Secretoras de Insulina/citología , Masculino , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Ratones Noqueados , Ratones Transgénicos , Miofibroblastos/citología , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Páncreas/lesiones , Páncreas/metabolismo , Células Estrelladas Pancreáticas/química , Células Estrelladas Pancreáticas/citología , Ratas , Regeneración/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA