Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Diabetologia ; 56(2): 423-33, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23188390

RESUMEN

AIMS/HYPOTHESIS: Type 2 diabetes mellitus is associated with reduced incretin effects. Although previous studies have shown that hyperglycaemia contributes to impaired incretin responses in beta cells, it is largely unknown how hyperlipidaemia, another feature of type 2 diabetes, contributes to impaired glucagon-like peptide 1 (GLP-1) response. Here, we investigated the effects of NEFA on incretin receptor signalling and examined the glucose-lowering efficacy of incretin-based drugs in combination with the lipid-lowering agent bezafibrate. METHODS: We used db/db mice to examine the in vivo efficacy of the treatment. Beta cell lines and mouse islets were used to examine GLP-1 and glucose-dependent insulinotropic peptide receptor signalling. RESULTS: Palmitate treatment decreased Glp1r expression in rodent insulinoma cell lines and isolated islets. This was associated with impairment of the following: GLP-1-stimulated cAMP production, phosphorylation of cAMP-responsive elements binding protein (CREB) and insulin secretion. In insulinoma cell lines, the expression of exogenous Glp1r restored cAMP production and the phosphorylation of CREB. Treatment with bezafibrate in combination with des-fluoro-sitagliptin or exendin-4 led to more robust glycaemic control, associated with improved islet morphology and beta cell mass in db/db mice. CONCLUSIONS/INTERPRETATION: Elevated NEFA contributes to impaired responsiveness to GLP-1, partially through downregulation of GLP-1 receptor signalling. Improvements in lipid control in mouse models of obesity and diabetes increase the efficacy of incretin-based therapy.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Incretinas/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Receptores de Glucagón/metabolismo , Animales , Receptor del Péptido 1 Similar al Glucagón , Masculino , Ratones , Ratones Endogámicos C57BL
2.
Diabetologia ; 55(11): 2999-3009, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22893028

RESUMEN

AIMS/HYPOTHESIS: Pancreatic beta cell destruction in type 1 diabetes may be mediated by cytokines such as IL-1ß, IFN-γ and TNF-α. Endoplasmic reticulum (ER) stress and nuclear factor-κB (NFκB) signalling are activated by cytokines, but their significance in beta cells remains unclear. Here, we investigated the role of cytokine-induced ER stress and NFκB signalling in beta cell destruction. METHODS: Isolated mouse islets and MIN6 beta cells were incubated with IL-1ß, IFN-γ and TNF-α. The chemical chaperone 4-phenylbutyric acid (PBA) was used to inhibit ER stress. Protein production and gene expression were assessed by western blot and real-time RT-PCR. RESULTS: We found in beta cells that inhibition of cytokine-induced ER stress with PBA unexpectedly potentiated cell death and NFκB-regulated gene expression. These responses were dependent on NFκB activation and were associated with a prolonged decrease in the inhibitor of κB-α (IκBα) protein, resulting from increased IκBα protein degradation. Cytokine-mediated NFκB-regulated gene expression was also potentiated after pre-induction of ER stress with thapsigargin, but not tunicamycin. Both PBA and thapsigargin treatments led to preferential upregulation of ER degradation genes over ER-resident chaperones as part of the adaptive unfolded protein response (UPR). In contrast, tunicamycin activated a balanced adaptive UPR in association with the maintenance of Xbp1 splicing. CONCLUSIONS/INTERPRETATION: These data suggest a novel mechanism by which cytokine-mediated ER stress interacts with NFκB signalling in beta cells, by regulating IκBα degradation. The cross-talk between the UPR and NFκB signalling pathways may be important in the regulation of cytokine-mediated beta cell death.


Asunto(s)
Muerte Celular/fisiología , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , FN-kappa B/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citocinas/farmacología , Diabetes Mellitus Tipo 1/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Inhibidores Enzimáticos/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas , Fenilbutiratos/farmacología , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tapsigargina/farmacología , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Tunicamicina/farmacología
3.
Diabetologia ; 54(7): 1766-76, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21472432

RESUMEN

AIMS/HYPOTHESIS: Pro-inflammatory cytokines such as IL-1ß, IFN-γ and TNF-α may contribute to pancreatic beta cell destruction in type 1 diabetes. A mechanism requiring nitric oxide, which is generated by inducible nitric oxide synthase (iNOS), in cytokine-induced endoplasmic reticulum (ER) stress and apoptosis has been proposed. Here, we tested the role of nitric oxide in cytokine-induced ER stress and the subsequent unfolded protein response (UPR) in beta cells. METHODS: Isolated islets from wild-type and iNos (also known as Nos2) knockout (iNos ( -/- )) mice, and MIN6 beta cells were incubated with IL-1ß, IFN-γ and TNF-α for 24-48 h. N (G)-methyl-L: -arginine was used to inhibit nitric oxide production in MIN6 cells. Protein levels and gene expression were assessed by western blot and real-time RT-PCR. RESULTS: In islets and MIN6 cells, inhibition of nitric oxide production had no effect on the generation of ER stress by cytokines, as evidenced by downregulation of Serca2b (also known as Atp2a2) mRNA and increased phosphorylation of PKR-like ER kinase, Jun N-terminal kinase (JNK) and eukaryotic translation initiation factor 2 α subunit. However, nitric oxide regulated the pattern of UPR signalling, which delineates the cellular decision to adapt to ER stress or to undergo apoptosis. Inhibition of nitric oxide production led to reduced expression of pro-apoptotic UPR markers, Chop (also known as Ddit3), Atf3 and Trib3. In contrast, adaptive UPR markers (chaperones, foldases and degradation enhancers) were increased. Further analysis of mouse islets showed that cytokine-induced Chop and Atf3 expression was also dependent on JNK activity. CONCLUSIONS/INTERPRETATION: The mechanism by which cytokines induce ER stress in mouse beta cells is independent of nitric oxide production. However, nitric oxide may regulate the switch between adaptive and apoptotic UPR signalling.


Asunto(s)
Citocinas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Óxido Nítrico/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Células Secretoras de Insulina/citología , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
4.
Diabetologia ; 54(2): 380-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21103982

RESUMEN

AIMS/HYPOTHESIS: Proinflammatory cytokines contribute to beta cell destruction in type 1 diabetes, but the mechanisms are incompletely understood. The aim of the current study was to address the role of the protein kinase C (PKC) isoform PKCδ, a diverse regulator of cell death, in cytokine-stimulated apoptosis in primary beta cells. METHODS: Islets isolated from wild-type or Prkcd(-/-) mice were treated with IL-1ß, TNF-α and IFNγ and assayed for apoptosis, nitric oxide (NO) generation and insulin secretion. Activation of signalling pathways, apoptosis and endoplasmic reticulum (ER) stress were determined by immunoblotting. Stabilisation of mRNA transcripts was measured by RT-PCR following transcriptional arrest. Mice were injected with multiple low doses of streptozotocin (MLD-STZ) and fasting blood glucose monitored. RESULTS: Deletion of Prkcd inhibited apoptosis and NO generation in islets stimulated ex vivo with cytokines. It also delayed the onset of hyperglycaemia in MLD-STZ-treated mice. Activation of ERK, p38, JNK, AKT1, the ER stress markers DDIT3 and phospho-EIF2α and the intrinsic apoptotic markers BCL2 and MCL1 was not different between genotypes. However, deletion of Prkcd destabilised mRNA transcripts for Nos2, and for multiple components of the toll-like receptor 2 (TLR2) signalling complex, which resulted in disrupted TLR2 signalling. CONCLUSIONS/INTERPRETATION: Loss of PKCδ partially protects against hyperglycaemia in the MLD-STZ model in vivo, and against cytokine-mediated apoptosis in vitro. This is accompanied by reduced NO generation and destabilisation of Nos2 and components of the TLR2 signalling pathway. The results highlight a mechanism for regulating proinflammatory gene expression in beta cells independently of transcription.


Asunto(s)
Apoptosis/efectos de los fármacos , Citocinas/farmacología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Isoenzimas/metabolismo , Proteína Quinasa C-delta/metabolismo , Animales , Apoptosis/genética , Western Blotting , Técnicas In Vitro , Células Secretoras de Insulina/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Islotes Pancreáticos/citología , Isoenzimas/genética , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Proteína Quinasa C-delta/genética , Factor de Necrosis Tumoral alfa/farmacología
5.
Diabetologia ; 52(11): 2369-2373, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19727664

RESUMEN

AIMS/HYPOTHESIS: Saturated fatty acids augment endoplasmic reticulum (ER) stress in pancreatic beta cells and this is implicated in the loss of beta cell mass that accompanies type 2 diabetes. However, the mechanisms underlying the induction of ER stress are unclear. Our aim was to establish whether saturated fatty acids cause defects in ER-to-Golgi protein trafficking, which may thereby contribute to ER stress via protein overload. METHODS: Cells of the mouse insulinoma cell line MIN6 were transfected with temperature-sensitive vesicular stomatitis virus G protein (VSVG) tagged with green fluorescent protein to quantify the rate of ER-to-Golgi protein trafficking. I14 antibody, which detects only correctly folded VSVG, was employed to probe the folding environment of the ER. ER stress markers were monitored by western blotting. RESULTS: Pretreatment with palmitate, but not oleate, significantly reduced the rate of ER-to-Golgi protein trafficking assessed using VSVG. This was not secondary to ER stress, since thapsigargin, which compromises chaperone function by depletion of ER calcium, markedly inhibited VSVG folding and promoted strong ER stress but only slightly reduced protein trafficking. Blockade of ER-to-Golgi protein trafficking with brefeldin A (BFA) was sufficient to trigger ER stress, but neither BFA nor palmitate compromised VSVG folding. CONCLUSIONS/INTERPRETATION: Reductions in ER-to-Golgi protein trafficking potentially contribute to ER stress during lipoapoptosis. In this case ER stress would be triggered by protein overload, rather than a disruption of the protein-folding capacity of the ER.


Asunto(s)
Retículo Endoplásmico/fisiología , Células Secretoras de Insulina/fisiología , Glicoproteínas de Membrana/metabolismo , Transporte de Proteínas/fisiología , Proteínas/metabolismo , Estrés Fisiológico/fisiología , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular Tumoral , Cicloheximida/farmacología , Genes Reporteros , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma , Ratones , Ácido Palmítico/farmacología , Transporte de Proteínas/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Transfección
6.
Mol Metab ; 13: 30-44, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29799418

RESUMEN

OBJECTIVE: The skeleton, which is strongly controlled by endocrine factors, has recently been shown to also play an active endocrine role itself, specifically influencing energy metabolism. However, much less is known about this role. Therefore, we sought to identify novel endocrine factors involved in the regulation of both bone mass and whole-body glucose homeostasis. METHODS: We used transcriptomic and proteomic analysis of Y1 receptor deficient osteoblasts combined with the generation of a novel osteoglycin deficient mouse model and performed comprehensive in vivo phenotype profiling, combined with osteoglycin administration in wildtype mice and human studies. RESULTS: Here we identify a novel role for osteoglycin, a secreted proteoglycan, in coordinating bone accretion with changes in energy balance. Using an osteoglycin knockout mouse model, we show that at a whole body level, osteoglycin acts to suppress bone formation and modulate whole body energy supplies by altering glucose uptake through changes in insulin secretion and sensitivity, as well as by altering food intake through central signaling. Examining humans following gastric surgery as a model of negative energy balance, we show that osteoglycin is associated with BMI and lean mass as well as changes in weight, BMI, and glucose levels. CONCLUSIONS: Thus, we identify osteoglycin as a novel factor involved in the regulation of energy homeostasis and identify a role for it in facilitating the matching of bone acquisition to alterations in energy status.


Asunto(s)
Huesos/metabolismo , Metabolismo Energético/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Adiposidad , Adulto , Animales , Peso Corporal , Metabolismo de los Hidratos de Carbono , Dieta Alta en Grasa , Femenino , Glucosa/metabolismo , Intolerancia a la Glucosa , Homeostasis/efectos de los fármacos , Humanos , Resistencia a la Insulina , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad , Osteoblastos/metabolismo , Osteoblastos/fisiología , Osteogénesis , Proteoma , Proteómica , Receptores de Neuropéptido Y , Transducción de Señal , Transcriptoma
7.
Mol Metab ; 10: 66-73, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29478918

RESUMEN

OBJECTIVES: Type 2 diabetes (T2D) is associated with chronic, low grade inflammation. Activation of the NLRP3 inflammasome and secretion of its target interleukin-1ß (IL-1ß) have been implicated in pancreatic ß cell failure in T2D. Specific targeting of the NLRP3 inflammasome to prevent pancreatic ß cell death could allow for selective T2D treatment without compromising all IL-1ß-associated immune responses. We hypothesized that treating a mouse model of T2D with MCC950, a compound that specifically inhibits NLRP3, would prevent pancreatic ß cell death, thereby preventing the onset of T2D. METHODS: Diabetic db/db mice were treated with MCC950 via drinking water for 8 weeks from 6 to 14 weeks of age, a period over which they developed pancreatic ß cell failure. We assessed metabolic parameters such as body composition, glucose tolerance, or insulin secretion over the course of the intervention. RESULTS: MCC950 was a potent inhibitor of NLRP3-induced IL-1ß in vitro and was detected at high levels in the plasma of treated db/db mice. Treatment of pre-diabetic db/db mice with MCC950, however, did not prevent pancreatic dysfunction and full onset of the T2D pathology. When examining the NLRP3 pathway in the pancreas of db/db mice, we could not detect an activation of this pathway nor increased levels of its target IL-1ß. CONCLUSIONS: NLRP3 driven-pancreatic IL-1ß inflammation does not play a key role in the pathogenesis of the db/db murine model of T2D.


Asunto(s)
Antiinflamatorios/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Células Secretoras de Insulina/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacología , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Furanos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Hipoglucemiantes/farmacología , Indenos , Células Secretoras de Insulina/efectos de los fármacos , Interleucina-1beta/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sulfonamidas , Sulfonas/farmacología , Sulfonas/uso terapéutico
8.
Diabetes ; 50 Suppl 1: S154-9, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11272180

RESUMEN

Inadequate beta-cell function is an essential component of all forms of diabetes. The most obvious problem is a failure to maintain sufficient beta-cell mass and function to cope with whatever insulin resistance is present. The most striking functional defect is a loss of acute glucose-induced insulin secretion (GIIS). This review discusses the ways in which beta-cells successfully adapt to increased demand and then decompensate as diabetes develops. Successful adaptation is achieved through increased beta-cell mass and increased insulin secretion. The hypothesis is explored that beta-cells exposed to the diabetic milieu lose their differentiation, which leads to loss of specialized functions such as GIIS. This concept has been strengthened by the finding of dedifferentiation of beta-cells in a rat model of partial pancreatectomy that includes a reduction of insulin gene expression, which may further contribute to decreased insulin production. Another finding was increased expression of c-Myc, which probably contributes to an increase in the expression of lactate dehydrogenase and the development of beta-cell hypertrophy. Arguments are developed that the beta-cell changes found in diabetes are better correlated with increased glucose levels than with non-esterified fatty acid levels, thus supporting the importance of glucose toxicity.


Asunto(s)
Diabetes Mellitus/fisiopatología , Islotes Pancreáticos/fisiopatología , Adaptación Fisiológica , Animales , División Celular , Tamaño de la Célula , Progresión de la Enfermedad , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Modelos Biológicos
9.
Diabetes ; 43(10): 1203-10, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7926289

RESUMEN

Thiazolidinediones offer promise as oral insulin-sensitizing agents. The effects of a new, high-potency compound (BRL 49653, SmithKline Beecham, Epsom, U.K.) were examined in insulin-resistant (high-fat-fed, HF) and control (high-starch-fed, HS) rats. The diet period was 3 weeks, with a BRL 49653 (10 mumol.kg-1.day-1) or vehicle gavage on the last 4 days. Then basal or euglycemic clamp studies were performed on animals in the conscious fasted state. In the basal state, BRL 49653 produced many similar metabolic responses in HF and HS rats (reduced insulin, glycerol, ketone body, and nonesterified fatty acid levels, reduced whole body glucose turnover, reduced brown adipose tissue glucose metabolism, and increased cardiac glucose metabolism and GLUT4 levels). In contrast, under euglycemic clamp conditions (500 pmol/l insulin), BRL 49653 only induced changes in the HF group (increased glucose infusion rate from 12.2 +/- 0.9 to 21.6 +/- 1.1 mg.kg-1.min-1 [P < 0.001], increased insulin suppressibility of hepatic glucose production [P < 0.01], and increased glucose uptake in muscle [P < 0.01]). BRL 49653 significantly reduced liver but not muscle triglyceride content in HF rats. We conclude that the agent has a general effect on lowering circulating lipid and insulin levels, manifested similarly in normal and insulin-resistant rats, but that enhancement of peripheral insulin action is confined to insulin-resistant rats. Therefore, the hypoinsulinemic action of the thiazolidinediones is probably not related simply to improved peripheral insulin sensitivity. The pattern of individual tissue response to BRL 49653 suggests that altered lipid availability is an important mediator of its effects on glucose metabolism.


Asunto(s)
Glucosa/metabolismo , Hipoglucemiantes/farmacología , Insulina/farmacología , Metabolismo de los Lípidos , Tiazoles/farmacología , Tiazolidinedionas , Ácido 3-Hidroxibutírico , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Disponibilidad Biológica , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Carbohidratos de la Dieta , Grasas de la Dieta , Ácidos Grasos no Esterificados/sangre , Técnica de Clampeo de la Glucosa , Glicerol/sangre , Corazón/efectos de los fármacos , Hidroxibutiratos/sangre , Insulina/sangre , Resistencia a la Insulina , Hígado/efectos de los fármacos , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Masculino , Músculos/efectos de los fármacos , Músculos/metabolismo , Miocardio/metabolismo , Ratas , Ratas Wistar , Rosiglitazona , Triglicéridos/metabolismo
10.
Adv Exp Med Biol ; 441: 263-70, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9781332

RESUMEN

Malonyl CoA is a regulator of carnitine palmitoyl transferase 1 (CPT1), the enzyme that controls the transfer of long chain fatty acyl CoA into mitochondria where it is oxidized. Recent studies indicate that in skeletal muscle the concentration of malonyl CoA is acutely (minutes) regulated by changes in its fuel supply and energy expenditure. In response to changes in fuel supply, regulation appears to be due to alterations in the cytosolic concentration of citrate, which is both an allosteric activator of acetyl CoA carboxylase (ACC), the enzyme that catalyzes malonyl CoA synthesis and a source of its precursor, cytosolic acetyl CoA. During exercise and immediately thereafter regulation by citrate appears to be lost and malonyl CoA levels diminish as the result of a decrease in ACC activity secondary to phosphorylation. Sustained increases in the concentration of malonyl CoA have been observed in muscle of a number of insulin-resistant rodents including the Zucker (fa/fa) and GK rats, KKAgy mice, glucose-infused rats and rats in which muscle has been made insulin resistant by denervation. Available data suggest that malonyl CoA could be linked to insulin resistance in these rodents by virtue of its effects on the cytosolic concentration of long chain fatty acyl CoA (LCFA CoA) and one or more protein kinase C isozymes. Whether similar alterations occur in other tissues and contribute to the pathophysiology of the insulin resistance syndrome remains to be determined.


Asunto(s)
Resistencia a la Insulina , Malonil Coenzima A/metabolismo , Músculo Esquelético/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Ratones , Ratas
11.
Diabetologia ; 50(10): 2117-25, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17641871

RESUMEN

AIMS/HYPOTHESIS: Hyperglycaemia can impair beta cell function after islet transplantation. Appropriate glucose-induced insulin secretion is dependent on a unique expression pattern of genes. Here we examined the effects of diabetes on gene expression in transplanted islets. MATERIALS AND METHODS: Streptozotocin-induced diabetic or control non-diabetic Lewis rats were transplanted under the kidney capsule with an insufficient number (2,000) of syngeneic islets to normalise blood glucose levels in diabetic rats. Eighteen days after transplantation, islet grafts were retrieved and RT-PCR used to assess expression of selected genes critical for beta cell function. Islet grafts from diabetic rats transplanted with a sufficient number of islets (3,000) to normalise hyperglycaemia were used to assess the effects of correcting blood glucose levels. Additionally, gene expression of transplanted islets from non-diabetic rats was compared with freshly isolated islets. RESULTS: In islet grafts from diabetic rats, mRNA levels of several transcription factors important for the maintenance of beta cell differentiation were reduced (pancreatic and duodenal homeobox 1 [Pdx1], neurogenic differentiation 1 [Neurod1], NK6 transcription factor related, locus 1 [Nkx6.1], paired box gene 6 [Pax6]), as were genes implicated in beta cell function (Glut2 [also known as solute carrier family 2 [facilitated glucose transporter], member 2 [Slc2a2], glucokinase, insulin, islet amyloid polypeptide [Iapp]). Conversely, mRNA levels of lactate dehydrogenase, which is normally suppressed in beta cells, were increased. The majority of the changes in gene expression were normalised after correction of hyperglycaemia, indicating that the severe loss of beta cell differentiation correlates with continuous exposure to diabetes. Even islet grafts from non-diabetic rats showed a few alterations in beta cell gene expression in comparison with fresh islets. CONCLUSIONS/INTERPRETATION: Chronic hyperglycaemia contributes to the deterioration of beta cell differentiation after islet transplantation.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/cirugía , Regulación de la Expresión Génica , Células Secretoras de Insulina/patología , Trasplante de Islotes Pancreáticos , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/genética , Modelos Animales de Enfermedad , Insulina/sangre , Masculino , Canales de Potasio de Rectificación Interna/genética , ARN Mensajero/genética , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Trasplante Homólogo
12.
Diabetologia ; 50(4): 752-63, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17268797

RESUMEN

AIMS/HYPOTHESIS: Increased lipid supply causes beta cell death, which may contribute to reduced beta cell mass in type 2 diabetes. We investigated whether endoplasmic reticulum (ER) stress is necessary for lipid-induced apoptosis in beta cells and also whether ER stress is present in islets of an animal model of diabetes and of humans with type 2 diabetes. METHODS: Expression of genes involved in ER stress was evaluated in insulin-secreting MIN6 cells exposed to elevated lipids, in islets isolated from db/db mice and in pancreas sections of humans with type 2 diabetes. Overproduction of the ER chaperone heat shock 70 kDa protein 5 (HSPA5, previously known as immunoglobulin heavy chain binding protein [BIP]) was performed to assess whether attenuation of ER stress affected lipid-induced apoptosis. RESULTS: We demonstrated that the pro-apoptotic fatty acid palmitate triggers a comprehensive ER stress response in MIN6 cells, which was virtually absent using non-apoptotic fatty acid oleate. Time-dependent increases in mRNA levels for activating transcription factor 4 (Atf4), DNA-damage inducible transcript 3 (Ddit3, previously known as C/EBP homologous protein [Chop]) and DnaJ homologue (HSP40) C3 (Dnajc3, previously known as p58) correlated with increased apoptosis in palmitate- but not in oleate-treated MIN6 cells. Attenuation of ER stress by overproduction of HSPA5 in MIN6 cells significantly protected against lipid-induced apoptosis. In islets of db/db mice, a variety of marker genes of ER stress were also upregulated. Increased processing (activation) of X-box binding protein 1 (Xbp1) mRNA was also observed, confirming the existence of ER stress. Finally, we observed increased islet protein production of HSPA5, DDIT3, DNAJC3 and BCL2-associated X protein in human pancreas sections of type 2 diabetes subjects. CONCLUSIONS/INTERPRETATION: Our results provide evidence that ER stress occurs in type 2 diabetes and is required for aspects of the underlying beta cell failure.


Asunto(s)
Apoptosis , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Animales , Western Blotting , Línea Celular , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiología , Humanos , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Chaperonas Moleculares/genética , Chaperonas Moleculares/fisiología , Estrés Oxidativo , ARN Mensajero/metabolismo
13.
Diabetologia ; 49(6): 1254-63, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16570159

RESUMEN

AIMS/HYPOTHESIS: We determined whether high-glucose-induced beta cell dysfunction is associated with oxidative stress in the DBA/2 mouse, a mouse strain susceptible to islet failure. MATERIALS AND METHODS: Glucose- and non-glucose-mediated insulin secretion from the islets of DBA/2 and control C57BL/6 mice was determined following a 48-h exposure to high glucose. Flux via the hexosamine biosynthesis pathway was assessed by determining O-glycosylated protein levels. Oxidative stress was determined by measuring hydrogen peroxide levels and the expression of anti-oxidant enzymes. RESULTS: Exposure to high glucose levels impaired glucose-stimulated insulin secretion in DBA/2 islets but not C57BL/6 islets, and this was associated with reduced islet insulin content and lower ATP levels than in C57BL/6 islets. Exposure of islets to glucosamine for 48 h mimicked the effects of high glucose on insulin secretion in the DBA/2 islets. High glucose exposure elevated O-glycosylated proteins; however, this occurred in islets from both strains, excluding a role for O-glycosylation in the impairment of DBA/2 insulin secretion. Additionally, both glucosamine and high glucose caused an increase in hydrogen peroxide in DBA/2 islets but not in C57BL/6 islets, an effect prevented by the antioxidant N-acetyl-L: -cysteine. Interestingly, while glutathione peroxidase and catalase expression was comparable between the two strains, the antioxidant enzyme manganese superoxide dismutase, which converts superoxide to hydrogen peroxide, was increased in DBA/2 islets, possibly explaining the increase in hydrogen peroxide levels. CONCLUSIONS/INTERPRETATION: Chronic high glucose culture caused an impairment in glucose-stimulated insulin secretion in DBA/2 islets, which have a genetic predisposition to failure, and this may be the result of oxidative stress.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Estrés Oxidativo/genética , Adenosina Trifosfato/metabolismo , Animales , Técnicas de Cultivo de Célula , Supervivencia Celular , Cartilla de ADN , Regulación de la Expresión Génica , Glucosa/farmacología , Glicosilación , Peróxido de Hidrógeno/análisis , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA/genética , Reacción en Cadena de la Polimerasa/métodos
14.
Am J Physiol ; 273(1 Pt 1): E1-9, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9252473

RESUMEN

Rats minimize hyperglycemia during chronic glucose infusion, but the metabolic processes are unclear. We investigated the tissues involved and the role of altered insulin sensitivity. Cannulated rats were infused with glucose (40 mg.kg-1.min-1) for 1 or 4 days or with saline (control). Hyperglycemia at 1 day (15.3 +/- 1.0 mM) was absent at 4 days (7.5 +/- 0.3 mM), but hyperinsulinemia persisted. Whole body glucose disposal was similarly elevated at 1 and 4 days, implying increased glucose clearance at 4 days (2-fold, P < 0.001). Muscle glucose uptake and glycogen content declined in glucose-infused rats from 1 to 4 days, whereas white adipose tissue (WAT) glucose uptake (6-fold, P < 0.001) and lipogenesis (3-fold, P < 0.001) increased. Muscle and liver triglyceride were doubled at both 1 and 4 days (P < 0.05 vs. control). Insulin sensitivity (assessed during euglycemic clamps) decreased in muscle to 34% of control at 1 and 4 days (P < 0.001 vs. control) and increased fivefold in WAT from 1 to 4 days (P < 0.05). Thus chronic glucose infusion results in a slow increase in efficiency of glucose clearance with enhanced WAT glucose uptake, lipogenesis, and insulin action. In contrast, the adaptation reduces glucose oversupply to muscle. Muscle shows sustained insulin resistance, with lipid accumulation a possible contributing factor.


Asunto(s)
Aclimatación/fisiología , Tejido Adiposo/metabolismo , Glucosa/metabolismo , Hiperglucemia/fisiopatología , Insulina/metabolismo , Músculo Esquelético/metabolismo , Animales , Desoxiglucosa/metabolismo , Ácidos Grasos no Esterificados/sangre , Glucosa/administración & dosificación , Técnica de Clampeo de la Glucosa , Hiperinsulinismo , Infusiones Intravenosas , Insulina/sangre , Secreción de Insulina , Lactatos/sangre , Masculino , Especificidad de Órganos , Ratas , Ratas Wistar , Factores de Tiempo , Triglicéridos/sangre
15.
Am J Physiol ; 273(3 Pt 2): H1309-16, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9321820

RESUMEN

The glucose transporter GLUT1 may play a more important role in cardiac than in skeletal muscle, but its regulation is unclear. During fasting, cardiac GLUT1 declines in the presence of low plasma insulin and glucose and high nonesterified fatty acid (NEFA) levels, whereas GLUT4 is unchanged. We investigated insulin, glucose, and NEFA levels as regulatory factors of cardiac GLUT content in chronically cannulated rats. Fasting rats were infused for 24 h with saline or insulin (2 rates) while plasma glucose was equalized by a glucose clamp; final transporter content was compared with a fed control group. There was a close association of GLUT1 content with insulin (r2 = 0.83, P < 0.001), with GLUT1 varying over a threefold range, under equivalent fasting glycemic conditions (plasma glucose, 5.1 +/- 0.1 mM). Maintenance of fed insulin levels during fasting prevented the GLUT1 fall (P < 0.01), whereas hyperinsulinemia (117 +/- 10 mU/l) led to significant overexpression of GLUT1 (155 +/- 12% of control, P < 0.01). When high glucose (7.6 +/- 0.1 mM) or high NEFA (0.76 +/- 0.05 mM) levels accompanied the hyperinsulinemia, upregulation of GLUT1 was blocked. GLUT1 content correlated with an estimate of cardiac glucose clearance across the groups. Cardiac GLUT4 content, hexokinase, and acyl-CoA synthase activities were unaffected by fasting, insulin, or substrate manipulation. In conclusion, insulin preferentially upregulates GLUT1 (but not GLUT4) in a dose-dependent manner in cardiac muscle in vivo, and substrate supply modulates this response, since upregulation can be effectively blocked by increased glucose or lipid availability. Therefore, both insulin exposure and energy status of cardiac muscle may be important determinants of cardiac GLUT1 expression.


Asunto(s)
Glucemia/fisiología , Ácidos Grasos no Esterificados/sangre , Corazón/efectos de los fármacos , Insulina/farmacología , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Musculares , Miocardio/metabolismo , Animales , Coenzima A Ligasas/metabolismo , Desoxiglucosa/metabolismo , Transportador de Glucosa de Tipo 1 , Transportador de Glucosa de Tipo 4 , Hexoquinasa/metabolismo , Homeostasis , Cinética , Masculino , Ratas , Ratas Wistar , Análisis de Regresión
16.
Am J Physiol ; 277(6): E1070-6, 1999 12.
Artículo en Inglés | MEDLINE | ID: mdl-10600797

RESUMEN

Chronic glucose infusion results in hyperinsulinemia and causes lipid accumulation and insulin resistance in rat muscle. To examine possible mechanisms for the insulin resistance, alterations in malonyl-CoA and long-chain acyl-CoA (LCA-CoA) concentration and the distribution of protein kinase C (PKC) isozymes, putative links between muscle lipids and insulin resistance, were determined. Cannulated rats were infused with glucose (40 mg. kg(-1). min(-1)) for 1 or 4 days. This increased red quadriceps muscle LCA-CoA content (sum of 6 species) by 1.3-fold at 1 day and 1.4-fold at 4 days vs. saline-infused controls (both P < 0.001 vs. control). The concentration of malonyl-CoA was also increased (1.7-fold at 1 day, P < 0.01, and 2.2-fold at 4 days, P < 0.001 vs. control), suggesting an even greater increase in cytosolic LCA-CoA. The ratio of membrane to cytosolic PKC-epsilon was increased twofold in the red gastrocnemius after both 1 and 4 days, suggesting chronic activation. No changes were observed for PKC-alpha, -delta, and -theta. We conclude that LCA-CoAs accumulate in muscle during chronic glucose infusion, consistent with a malonyl-CoA-induced inhibition of fatty acid oxidation (reverse glucose-fatty acid cycle). Accumulation of LCA-CoAs could play a role in the generation of muscle insulin resistance by glucose oversupply, either directly or via chronic activation of PKC-epsilon.


Asunto(s)
Glucosa/farmacología , Resistencia a la Insulina/fisiología , Isoenzimas/metabolismo , Metabolismo de los Lípidos , Músculo Esquelético/enzimología , Proteína Quinasa C/metabolismo , Acilcoenzima A/metabolismo , Animales , Glucemia , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Insulina/sangre , Isoenzimas/análisis , Masculino , Malonil Coenzima A/metabolismo , Proteína Quinasa C/análisis , Proteína Quinasa C-alfa , Proteína Quinasa C-delta , Proteína Quinasa C-epsilon , Proteína Quinasa C-theta , Ratas , Ratas Wistar , Fracciones Subcelulares/enzimología
17.
Am J Physiol ; 276(6): E1030-7, 1999 06.
Artículo en Inglés | MEDLINE | ID: mdl-10362615

RESUMEN

In liver, insulin and glucose acutely increase the concentration of malonyl-CoA by dephosphorylating and activating acetyl-CoA carboxylase (ACC). In contrast, in incubated rat skeletal muscle, they appear to act by increasing the cytosolic concentration of citrate, an allosteric activator of ACC, as reflected by increases in the whole cell concentrations of citrate and malate [Saha, A. K., D. Vavvas, T. G. Kurowski, A. Apazidis, L. A. Witters, E. Shafrir, and N. B. Ruderman. Am. J. Physiol. 272 (Endocrinol. Metab. 35): E641-E648, 1997]. We report here that sustained increases in plasma insulin and glucose may also increase the concentration of malonyl-CoA in rat skeletal muscle in vivo by this mechanism. Thus 70 and 125% increases in malonyl-CoA induced in skeletal muscle by infusions of glucose for 1 and 4 days, respectively, and a twofold increase in its concentration during a 90-min euglycemic-hyperinsulinemic clamp were all associated with significant increases in the sum of whole cell concentrations of citrate and/or malate. Similar correlations were observed in muscle of the hyperinsulinemic fa/fa rat, in denervated muscle, and in muscle of rats infused with insulin for 5 h. In muscle of 48-h-starved rats 3 and 24 h after refeeding, increases in malonyl-CoA were not accompanied by consistent increases in the concentrations of malate or citrate. However, they were associated with a decrease in the whole cell concentration of long-chain fatty acyl-CoA (LCFA-CoA), an allosteric inhibitor of ACC. The results suggest that increases in the concentration of malonyl-CoA, caused in rat muscle in vivo by sustained increases in plasma insulin and glucose or denervation, may be due to increases in the cytosolic concentration of citrate. In contrast, during refeeding after starvation, the increase in malonyl-CoA in muscle is probably due to another mechanism.


Asunto(s)
Ácido Cítrico/metabolismo , Citosol/metabolismo , Malonil Coenzima A/metabolismo , Músculo Esquelético/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Alimentos , Insulina/farmacología , Malatos/metabolismo , Masculino , Desnervación Muscular , Músculo Esquelético/efectos de los fármacos , Obesidad/genética , Obesidad/metabolismo , Concentración Osmolar , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Inanición/metabolismo
18.
J Biol Chem ; 276(38): 35375-81, 2001 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-11457846

RESUMEN

Glucose-induced insulin secretion from hyperglycemic 90% pancreatectomized rats is markedly impaired, possibly because of loss of beta cell differentiation. Association of these changes with beta cell hypertrophy, increased mRNA levels of the transcription factor c-Myc, and their complete normalization by phlorizin treatment suggested a link between chronic hyperglycemia, increased c-Myc expression, and altered beta cell function. In this study, we tested the effect of hyperglycemia on rat pancreatic islet c-Myc expression both in vivo and in vitro. Elevation of plasma glucose for 1-4 days (glucose infusion/clamp) was followed by parallel increases in islet mRNA levels (relative to TATA-binding protein) of c-Myc and two of its target genes, ornithine decarboxylase and lactate dehydrogenase A. Similar changes were observed in vitro upon stimulation of cultured islets or purified beta cells with 20 and 30 mmol.liter(-1) glucose for 18 h. These effects of high glucose were reproduced by high potassium-induced depolarization or dibutyryl-cAMP and were inhibited by agents decreasing cytosolic Ca(2+) or cAMP concentrations. In conclusion, the expression of the early response gene c-Myc in rat pancreatic beta cells is stimulated by high glucose in a Ca(2+)-dependent manner and by cAMP. c-Myc could therefore participate to the regulation of beta cell growth, apoptosis, and differentiation under physiological or pathophysiological conditions.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Genes myc , Glucosa/farmacología , Islotes Pancreáticos/efectos de los fármacos , Animales , Secuencia de Bases , Calcio/metabolismo , Células Cultivadas , Clonidina/farmacología , AMP Cíclico/farmacología , Citosol/metabolismo , Cartilla de ADN , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA