Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(8): 105065, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37468098

RESUMEN

Pancreatic beta cells maintain glucose homeostasis by secreting pulses of insulin in response to a rise in plasma glucose. Pulsatile insulin secretion occurs as a result of glucose-induced oscillations in beta-cell cytosolic Ca2+. The endoplasmic reticulum (ER) helps regulate beta-cell cytosolic Ca2+, and ER stress can lead to ER Ca2+ reduction, beta-cell dysfunction, and an increased risk of type 2 diabetes. However, the mechanistic effects of ER stress on individual calcium channels are not well understood. To determine the effects of tunicamycin-induced ER stress on ER inositol 1,4,5-triphosphate receptors (IP3Rs) and ryanodine receptors (RyRs) and their involvement in subsequent Ca2+ dysregulation, we treated INS-1 832/13 cells and primary mouse islets with ER stress inducer tunicamycin (TM). We showed TM treatment increased RyR1 mRNA without affecting RyR2 mRNA and decreased both IP3R1 and IP3R3 mRNA. Furthermore, we found stress reduced ER Ca2+ levels, triggered oscillations in cytosolic Ca2+ under subthreshold glucose conditions, and increased apoptosis and that these changes were prevented by cotreatment with the RyR1 inhibitor dantrolene. In addition, we demonstrated silencing RyR1-suppressed TM-induced subthreshold cytosolic Ca2+ oscillations, but silencing RyR2 did not affect these oscillations. In contrast, inhibiting IP3Rs with xestospongin-C failed to suppress the TM-induced cytosolic Ca2+ oscillations and did not protect beta cells from TM-induced apoptosis although xestospongin-C inclusion did prevent ER Ca2+ reduction. Taken together, these results show changes in RyR1 play a critical role in ER stress-induced Ca2+ dysfunction and beta-cell apoptosis.


Asunto(s)
Señalización del Calcio , Estrés del Retículo Endoplásmico , Células Secretoras de Insulina , Canal Liberador de Calcio Receptor de Rianodina , Animales , Ratones , Apoptosis , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Homeostasis , Células Secretoras de Insulina/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Tunicamicina , Ratas , Línea Celular
2.
Pharmacol Rev ; 73(3): 1001-1015, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34193595

RESUMEN

Both type 1 and type 2 diabetes mellitus are advancing at exponential rates, placing significant burdens on health care networks worldwide. Although traditional pharmacologic therapies such as insulin and oral antidiabetic stalwarts like metformin and the sulfonylureas continue to be used, newer drugs are now on the market targeting novel blood glucose-lowering pathways. Furthermore, exciting new developments in the understanding of beta cell and islet biology are driving the potential for treatments targeting incretin action, islet transplantation with new methods for immunologic protection, and the generation of functional beta cells from stem cells. Here we discuss the mechanistic details underlying past, present, and future diabetes therapies and evaluate their potential to treat and possibly reverse type 1 and 2 diabetes in humans. SIGNIFICANCE STATEMENT: Diabetes mellitus has reached epidemic proportions in the developed and developing world alike. As the last several years have seen many new developments in the field, a new and up to date review of these advances and their careful evaluation will help both clinical and research diabetologists to better understand where the field is currently heading.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Hipoglucemiantes/uso terapéutico , Insulina
3.
J Physiol ; 601(24): 5655-5667, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37983196

RESUMEN

Pancreatic beta cells secrete insulin in response to plasma glucose. The ATP-sensitive potassium channel (KATP ) links glucose metabolism to islet electrical activity in these cells by responding to increased cytosolic [ATP]/[ADP]. It was recently proposed that pyruvate kinase (PK) in close proximity to beta cell KATP locally produces the ATP that inhibits KATP activity. This proposal was largely based on the observation that applying phosphoenolpyruvate (PEP) and ADP to the cytoplasmic side of excised inside-out patches inhibited KATP . To test the relative contributions of local vs. mitochondrial ATP production, we recorded KATP activity using mouse beta cells and INS-1 832/13 cells. In contrast to prior reports, we could not replicate inhibition of KATP activity by PEP + ADP. However, when the pH of the PEP solutions was not corrected for the addition of PEP, strong channel inhibition was observed as a result of the well-known action of protons to inhibit KATP . In cell-attached recordings, perifusing either a PK activator or an inhibitor had little or no effect on KATP channel closure by glucose, further suggesting that PK is not an important regulator of KATP . In contrast, addition of mitochondrial inhibitors robustly increased KATP activity. Finally, by measuring the [ATP]/[ADP] responses to imposed calcium oscillations in mouse beta cells, we found that oxidative phosphorylation could raise [ATP]/[ADP] even when ADP was at its nadir during the burst silent phase, in agreement with our mathematical model. These results indicate that ATP produced by mitochondrial oxidative phosphorylation is the primary controller of KATP in pancreatic beta cells. KEY POINTS: Phosphoenolpyruvate (PEP) plus adenosine diphosphate does not inhibit KATP activity in excised patches. PEP solutions only inhibit KATP activity if the pH is unbalanced. Modulating pyruvate kinase has minimal effects on KATP activity. Mitochondrial inhibition, in contrast, robustly potentiates KATP activity in cell-attached patches. Although the ADP level falls during the silent phase of calcium oscillations, mitochondria can still produce enough ATP via oxidative phosphorylation to close KATP . Mitochondrial oxidative phosphorylation is therefore the main source of the ATP that inhibits the KATP activity of pancreatic beta cells.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Ratones , Animales , Células Secretoras de Insulina/metabolismo , Adenosina Trifosfato/farmacología , Adenosina Trifosfato/metabolismo , Fosfoenolpiruvato/metabolismo , Fosfoenolpiruvato/farmacología , Piruvato Quinasa/metabolismo , Piruvato Quinasa/farmacología , Adenosina Difosfato/farmacología , Adenosina Difosfato/metabolismo , Mitocondrias/metabolismo
4.
Am J Physiol Endocrinol Metab ; 324(6): E477-E487, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37074988

RESUMEN

The standard model for Ca2+ oscillations in insulin-secreting pancreatic ß cells centers on Ca2+ entry through voltage-activated Ca2+ channels. These work in combination with ATP-dependent K+ channels, which are the bridge between the metabolic state of the cells and plasma membrane potential. This partnership underlies the ability of the ß cells to secrete insulin appropriately on a minute-to-minute time scale to control whole body plasma glucose. Though this model, developed over more than 40 years through many cycles of experimentation and mathematical modeling, has been very successful, it has been challenged by a hypothesis that calcium-induced calcium release from the endoplasmic reticulum through ryanodine or inositol trisphosphate (IP3) receptors is instead the key driver of islet oscillations. We show here that the alternative model is in fact incompatible with a large body of established experimental data and that the new observations offered in support of it can be better explained by the standard model.


Asunto(s)
Células Secretoras de Insulina , Células Secretoras de Insulina/metabolismo , Calcio/metabolismo , Insulina/metabolismo , Señalización del Calcio , Secreción de Insulina
5.
Biophys J ; 121(5): 692-704, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35131294

RESUMEN

Pulsatile insulin secretion by pancreatic beta cells is necessary for tight glucose control in the body. Glycolytic oscillations have been proposed as the mechanism for generating the electrical oscillations underlying pulsatile insulin secretion. The glycolytic enzyme 6-phosphofructokinase-1 (PFK) synthesizes fructose-1,6-bisphosphate (FBP) from fructose-6-phosphate. It has been proposed that the slow electrical and Ca2+ oscillations (periods of 3-5 min) observed in islets result from allosteric feedback activation of PFKM by FBP. Pancreatic beta cells express three PFK isozymes: PFKL, PFKM, and PFKP. A prior study of mice that were engineered to lack PFKM using a gene-trap strategy to delete Pfkm produced a mosaic reduction in global Pfkm expression, but the islets isolated from the mice still exhibited slow Ca2+ oscillations. However, these islets still expressed residual PFKM protein. Thus, to more fully test the hypothesis that beta cell PFKM is responsible for slow islet oscillations, we made a beta-cell-specific knockout mouse that completely lacked PFKM. While PFKM deletion resulted in subtle metabolic changes in vivo, islets that were isolated from these mice continued to exhibit slow oscillations in electrical activity, beta cell Ca2+ concentrations, and glycolysis, as measured using PKAR, an FBP reporter/biosensor. Furthermore, simulations obtained with a mathematical model of beta cell activity shows that slow oscillations can persist despite PFKM loss provided that one of the other PFK isoforms, such as PFKP, is present, even if its level of expression is unchanged. Thus, while we believe that PFKM may be the main regulator of slow oscillations in wild-type islets, PFKP can provide functional redundancy. Our model also suggests that PFKM likely dominates, in vivo, because it outcompetes PFKP with its higher FBP affinity and lower ATP affinity. We thus propose that isoform redundancy may rescue key physiological processes of the beta cell in the absence of certain critical genes.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Fosfofructoquinasa-1 , Animales , Calcio/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Fosfofructoquinasa-1/genética , Fosfofructoquinasa-1/metabolismo
6.
Biophys J ; 121(8): 1449-1464, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35300967

RESUMEN

ATP-sensitive K+ (K(ATP)) channels were first reported in the ß-cells of pancreatic islets in 1984, and it was soon established that they are the primary means by which the blood glucose level is transduced to cellular electrical activity and consequently insulin secretion. However, the role that the K(ATP) channels play in driving the bursting electrical activity of islet ß-cells, which drives pulsatile insulin secretion, remains unclear. One difficulty is that bursting is abolished when several different ion channel types are blocked pharmacologically or genetically, making it challenging to distinguish causation from correlation. Here, we demonstrate a means for determining whether activity-dependent oscillations in K(ATP) conductance play the primary role in driving electrical bursting in ß-cells. We use mathematical models to predict that if K(ATP) is the driver, then contrary to intuition, the mean, peak, and nadir levels of ATP/ADP should be invariant to changes in glucose within the concentration range that supports bursting. We test this in islets using Perceval-HR to image oscillations in ATP/ADP. We find that mean, peak, and nadir levels are indeed approximately invariant, supporting the hypothesis that oscillations in K(ATP) conductance are the main drivers of the slow bursting oscillations typically seen at stimulatory glucose levels in mouse islets. In conclusion, we provide, for the first time to our knowledge, causal evidence for the role of K(ATP) channels not only as the primary target for glucose regulation but also for their role in driving bursting electrical activity and pulsatile insulin secretion.


Asunto(s)
Señalización del Calcio , Islotes Pancreáticos , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/fisiología , Glucosa/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Potenciales de la Membrana/fisiología , Ratones
7.
J Biol Chem ; 295(17): 5685-5700, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32179650

RESUMEN

Type 2 diabetes mellitus (T2DM) is characterized by impaired glucose-stimulated insulin secretion and increased peripheral insulin resistance. Unremitting endoplasmic reticulum (ER) stress can lead to beta-cell apoptosis and has been linked to type 2 diabetes. Although many studies have attempted to link ER stress and T2DM, the specific effects of ER stress on beta-cell function remain incompletely understood. To determine the interrelationship between ER stress and beta-cell function, here we treated insulin-secreting INS-1(832/13) cells or isolated mouse islets with the ER stress-inducer tunicamycin (TM). TM induced ER stress as expected, as evidenced by activation of the unfolded protein response. Beta cells treated with TM also exhibited concomitant alterations in their electrical activity and cytosolic free Ca2+ oscillations. As ER stress is known to reduce ER Ca2+ levels, we tested the hypothesis that the observed increase in Ca2+ oscillations occurred because of reduced ER Ca2+ levels and, in turn, increased store-operated Ca2+ entry. TM-induced cytosolic Ca2+ and membrane electrical oscillations were acutely inhibited by YM58483, which blocks store-operated Ca2+ channels. Significantly, TM-treated cells secreted increased insulin under conditions normally associated with only minimal release, e.g. 5 mm glucose, and YM58483 blocked this secretion. Taken together, these results support a critical role for ER Ca2+ depletion-activated Ca2+ current in mediating Ca2+-induced insulin secretion in response to ER stress.


Asunto(s)
Calcio/metabolismo , Estrés del Retículo Endoplásmico , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Animales , Señalización del Calcio , Cationes Bivalentes/metabolismo , Línea Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Masculino , Ratones , Ratas
8.
Prog Mol Subcell Biol ; 59: 145-162, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34050865

RESUMEN

The endoplasmic reticulum (ER) is an organelle that mediates the proper folding and assembly of proteins destined for the cell surface, the extracellular space and subcellular compartments such as the lysosomes. The ER contains a wide range of molecular chaperones to handle the folding requirements of a diverse set of proteins that traffic through this compartment. The lectin-like chaperones calreticulin and calnexin are an important class of structurally-related chaperones relevant for the folding and assembly of many N-linked glycoproteins. Despite the conserved mechanism of action of these two chaperones in nascent protein recognition and folding, calreticulin has unique functions in cellular calcium signaling and in the immune response. The ER-related functions of calreticulin in the assembly of major histocompatibility complex (MHC) class I molecules are well-studied and provide many insights into the modes of substrate and co-chaperone recognition by calreticulin. Calreticulin is also detectable on the cell surface under some conditions, where it induces the phagocytosis of apoptotic cells. Furthermore, mutations of calreticulin induce cell transformation in myeloproliferative neoplasms (MPN). Studies of the functions of the mutant calreticulin in cell transformation and immunity have provided many insights into the normal biology of calreticulin, which are discussed.


Asunto(s)
Señalización del Calcio , Señalización del Calcio/genética , Calnexina/genética , Calnexina/metabolismo , Calreticulina/genética , Calreticulina/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Sistema Inmunológico , Pliegue de Proteína
9.
J Biol Chem ; 292(9): 3841-3853, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28100774

RESUMEN

The failure of pancreatic islet ß-cells is a major contributor to the etiology of type 2 diabetes. ß-Cell dysfunction and declining ß-cell mass are two mechanisms that contribute to this failure, although it is unclear whether they are molecularly linked. Here, we show that the cell cycle regulator, cyclin-dependent kinase 2 (CDK2), couples primary ß-cell dysfunction to the progressive deterioration of ß-cell mass in diabetes. Mice with pancreas-specific deletion of Cdk2 are glucose-intolerant, primarily due to defects in glucose-stimulated insulin secretion. Accompanying this loss of secretion are defects in ß-cell metabolism and perturbed mitochondrial structure. Persistent insulin secretion defects culminate in progressive deficits in ß-cell proliferation, reduced ß-cell mass, and diabetes. These outcomes may be mediated directly by the loss of CDK2, which binds to and phosphorylates the transcription factor FOXO1 in a glucose-dependent manner. Further, we identified a requirement for CDK2 in the compensatory increases in ß-cell mass that occur in response to age- and diet-induced stress. Thus, CDK2 serves as an important nexus linking primary ß-cell dysfunction to progressive ß-cell mass deterioration in diabetes.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Diabetes Mellitus Experimental/patología , Células Secretoras de Insulina/patología , Páncreas/patología , Animales , Peso Corporal , Proliferación Celular , Quinasa 2 Dependiente de la Ciclina/genética , Dieta Alta en Grasa , Progresión de la Enfermedad , Femenino , Genotipo , Glucosa/química , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Noqueados , Microscopía Fluorescente , Fenotipo , Fosforilación
10.
PLoS Comput Biol ; 13(7): e1005686, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28749940

RESUMEN

Plasma insulin oscillations are known to have physiological importance in the regulation of blood glucose. In insulin-secreting ß-cells of pancreatic islets, K(ATP) channels play a key role in regulating glucose-dependent insulin secretion. In addition, they convey oscillations in cellular metabolism to the membrane by sensing adenine nucleotides, and are thus instrumental in mediating pulsatile insulin secretion. Blocking K(ATP) channels pharmacologically depolarizes the ß-cell plasma membrane and terminates islet oscillations. Surprisingly, when K(ATP) channels are genetically knocked out, oscillations in islet activity persist, and relatively normal blood glucose levels are maintained. Compensation must therefore occur to overcome the loss of K(ATP) channels in K(ATP) knockout mice. In a companion study, we demonstrated a substantial increase in Kir2.1 protein occurs in ß-cells lacking K(ATP) because of SUR1 deletion. In this report, we demonstrate that ß-cells of SUR1 null islets have an upregulated inward rectifying K+ current that helps to compensate for the loss of K(ATP) channels. This current is likely due to the increased expression of Kir2.1 channels. We used mathematical modeling to determine whether an ionic current having the biophysical characteristics of Kir2.1 is capable of rescuing oscillations that are similar in period to those of wild-type islets. By experimentally testing a key model prediction we suggest that Kir2.1 current upregulation is a likely mechanism for rescuing the oscillations seen in islets from mice deficient in K(ATP) channels.


Asunto(s)
Calcio/metabolismo , Islotes Pancreáticos/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Regulación hacia Arriba/fisiología , Animales , Glucemia/metabolismo , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados
11.
J Biol Chem ; 291(26): 13715-29, 2016 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-27129239

RESUMEN

Inflammatory breast cancer (IBC) is an extremely lethal cancer that rapidly metastasizes. Although the molecular attributes of IBC have been described, little is known about the underlying metabolic features of the disease. Using a variety of metabolic assays, including (13)C tracer experiments, we found that SUM149 cells, the primary in vitro model of IBC, exhibit metabolic abnormalities that distinguish them from other breast cancer cells, including elevated levels of N-acetylaspartate, a metabolite primarily associated with neuronal disorders and gliomas. Here we provide the first evidence of N-acetylaspartate in breast cancer. We also report that the oncogene RhoC, a driver of metastatic potential, modulates glutamine and N-acetylaspartate metabolism in IBC cells in vitro, revealing a novel role for RhoC as a regulator of tumor cell metabolism that extends beyond its well known role in cytoskeletal rearrangement.


Asunto(s)
Ácido Aspártico/análogos & derivados , Glutamina/metabolismo , Neoplasias Inflamatorias de la Mama/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Ácido Aspártico/biosíntesis , Ácido Aspártico/genética , Línea Celular Tumoral , Femenino , Glutamina/genética , Humanos , Neoplasias Inflamatorias de la Mama/genética , Neoplasias Inflamatorias de la Mama/patología , Proteínas de Neoplasias/genética , Proteínas de Unión al GTP rho/genética , Proteína rhoC de Unión a GTP
12.
Biophys J ; 110(3): 691-699, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26840733

RESUMEN

Metabolism in islet ß-cells displays oscillations that can trigger pulses of electrical activity and insulin secretion. There has been a decades-long debate among islet biologists about whether metabolic oscillations are intrinsic or occur in response to oscillations in intracellular Ca(2+) that result from bursting electrical activity. In this article, the dynamics of oscillatory metabolism were investigated using five different optical reporters. Reporter activity was measured simultaneously with membrane potential bursting to determine the phase relationships between the metabolic oscillations and electrical activity. Our experimental findings suggest that Ca(2+) entry into ß-cells stimulates the rate of mitochondrial metabolism, accounting for the depletion of glycolytic intermediates during each oscillatory burst. We also performed Ca(2+) clamp tests in which we clamped membrane potential with the KATP channel-opener diazoxide and KCl to fix Ca(2+) at an elevated level. These tests confirm that metabolic oscillations do not require Ca(2+) oscillations, but show that Ca(2+) plays a larger role in shaping metabolic oscillations than previously suspected. A dynamical picture of the mechanisms of oscillations emerged that requires the restructuring of contemporary mathematical ß-cell models, including our own dual oscillator model. In the companion article, we modified our model to account for these new data.


Asunto(s)
Señalización del Calcio , Células Secretoras de Insulina/metabolismo , Potenciales de la Membrana , Animales , Células Cultivadas , Células Secretoras de Insulina/fisiología , Canales KATP/metabolismo , Ratones
13.
Biophys J ; 110(3): 733-742, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26840737

RESUMEN

Pancreatic islets respond to elevated blood glucose by secreting pulses of insulin that parallel oscillations in ß-cell metabolism, intracellular Ca(2+) concentration, and bursting electrical activity. The mechanisms that maintain an oscillatory response are not fully understood, yet several models have been proposed. Only some can account for experiments supporting that metabolism is intrinsically oscillatory in ß-cells. The dual oscillator model (DOM) implicates glycolysis as the source of oscillatory metabolism. In the companion article, we use recently developed biosensors to confirm that glycolysis is oscillatory and further elucidate the coordination of metabolic and electrical signals in the insulin secretory pathway. In this report, we modify the DOM by incorporating an established link between metabolism and intracellular Ca(2+) to reconcile model predictions with experimental observations from the companion article. With modification, we maintain the distinguishing feature of the DOM, oscillatory glycolysis, but introduce the ability of Ca(2+) influx to reshape glycolytic oscillations by promoting glycolytic efflux. We use the modified model to explain measurements from the companion article and from previously published experiments with islets.


Asunto(s)
Adenosina Trifosfato/metabolismo , Señalización del Calcio , Calcio/metabolismo , Células Secretoras de Insulina/metabolismo , Potenciales de Acción , Animales , Metabolismo Energético , Glucólisis , Humanos , Células Secretoras de Insulina/fisiología , Modelos Teóricos , Periodicidad
14.
J Neurosci ; 34(34): 11405-15, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25143620

RESUMEN

The adipocyte-derived hormone leptin modulates neural systems appropriately for the status of body energy stores. Leptin inhibits lateral hypothalamic area (LHA) orexin (OX; also known as hypocretin)-producing neurons, which control feeding, activity, and energy expenditure, among other parameters. Our previous results suggest that GABAergic LHA leptin receptor (LepRb)-containing and neurotensin (Nts)-containing (LepRb(Nts)) neurons lie in close apposition with OX neurons and control Ox mRNA expression. Here, we show that, similar to leptin, activation of LHA Nts neurons by the excitatory hM3Dq DREADD (designer receptor exclusively activated by designer drugs) hyperpolarizes membrane potential and suppresses action potential firing in OX neurons in mouse hypothalamic slices. Furthermore, ablation of LepRb from Nts neurons abrogated the leptin-mediated inhibition, demonstrating that LepRb(Nts) neurons mediate the inhibition of OX neurons by leptin. Leptin did not significantly enhance GABAA-mediated inhibitory synaptic transmission, and GABA receptor antagonists did not block leptin-mediated inhibition of OX neuron activity. Rather, leptin diminished the frequency of spontaneous EPSCs onto OX neurons. Furthermore, leptin indirectly activated an ATP-sensitive potassium (K(ATP)) channel in OX neurons, which was required for the hyperpolarization of OX neurons by leptin. Although Nts did not alter OX activity, galanin, which is coexpressed in LepRb(Nts) neurons, inhibited OX neurons, whereas the galanin receptor antagonist M40 (galanin-(1-12)-Pro3-(Ala-Leu)2-Ala amide) prevented the leptin-induced hyperpolarization of OX cells. These findings demonstrate that leptin indirectly inhibits OX neurons by acting on LHA LepRb(Nts) neurons to mediate two distinct GABA-independent mechanisms of inhibition: the presynaptic inhibition of excitatory neurotransmission and the opening of K(ATP) channels.


Asunto(s)
Área Hipotalámica Lateral/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leptina/farmacología , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuropéptidos/metabolismo , Neurotensina/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Femenino , Antagonistas del GABA/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Inhibición Neural/fisiología , Neuropéptidos/genética , Neurotensina/genética , Orexinas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Leptina/deficiencia
15.
Diabetologia ; 58(10): 2298-306, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26141787

RESUMEN

AIMS/HYPOTHESIS: miR-153 is an intronic miRNA embedded in the genes that encode IA-2 (also known as PTPRN) and IA-2ß (also known as PTPRN2). Islet antigen (IA)-2 and IA-2ß are major autoantigens in type 1 diabetes and are important transmembrane proteins in dense core and synaptic vesicles. miR-153 and its host genes are co-regulated in pancreas and brain. The present experiments were initiated to decipher the regulatory network between miR-153 and its host gene Ia-2ß (also known as Ptprn2). METHODS: Insulin secretion was determined by ELISA. Identification of miRNA targets was assessed using luciferase assays and by quantitative real-time PCR and western blots in vitro and in vivo. Target protector was also employed to evaluate miRNA target function. RESULTS: Functional studies revealed that miR-153 mimic suppresses both glucose- and potassium-induced insulin secretion (GSIS and PSIS, respectively), whereas miR-153 inhibitor enhances both GSIS and PSIS. A similar effect on dopamine secretion also was observed. Using miRNA target prediction software, we found that miR-153 is predicted to target the 3'UTR region of the calcium channel gene, Cacna1c. Further studies confirmed that Cacna1c mRNA and protein are downregulated by miR-153 mimics and upregulated by miR-153 inhibitors in insulin-secreting freshly isolated mouse islets, in the insulin-secreting mouse cell line MIN6 and in the dopamine-secreting cell line PC12. CONCLUSIONS/INTERPRETATION: miR-153 is a negative regulator of both insulin and dopamine secretion through its effect on Cacna1c expression, which suggests that IA-2ß and miR-153 have opposite functional effects on the secretory pathway.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Dopamina/metabolismo , Insulina/metabolismo , MicroARNs/metabolismo , Animales , Encéfalo/metabolismo , Canales de Calcio Tipo L/genética , Línea Celular , Regulación de la Expresión Génica , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , MicroARNs/genética , Páncreas/metabolismo , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo
16.
J Biol Chem ; 289(47): 32798-810, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25271154

RESUMEN

Although the pancreatic duodenal homeobox 1 (Pdx-1) transcription factor is known to play an indispensable role in ß cell development and secretory function, recent data also implicate Pdx-1 in the maintenance of endoplasmic reticulum (ER) health. The sarco-endoplasmic reticulum Ca(2+) ATPase 2b (SERCA2b) pump maintains a steep Ca(2+) gradient between the cytosol and ER lumen. In models of diabetes, our data demonstrated loss of ß cell Pdx-1 that occurs in parallel with altered SERCA2b expression, whereas in silico analysis of the SERCA2b promoter revealed multiple putative Pdx-1 binding sites. We hypothesized that Pdx-1 loss under inflammatory and diabetic conditions leads to decreased SERCA2b levels and activity with concomitant alterations in ER health. To test this, siRNA-mediated knockdown of Pdx-1 was performed in INS-1 cells. The results revealed reduced SERCA2b expression and decreased ER Ca(2+), which was measured using fluorescence lifetime imaging microscopy. Cotransfection of human Pdx-1 with a reporter fused to the human SERCA2 promoter increased luciferase activity 3- to 4-fold relative to an empty vector control, and direct binding of Pdx-1 to the proximal SERCA2 promoter was confirmed by chromatin immunoprecipitation. To determine whether restoration of SERCA2b could rescue ER stress induced by Pdx-1 loss, Pdx1(+/-) mice were fed a high-fat diet. Isolated islets demonstrated an increased spliced-to-total Xbp1 ratio, whereas SERCA2b overexpression reduced the Xbp1 ratio to that of wild-type controls. Together, these results identify SERCA2b as a novel transcriptional target of Pdx-1 and define a role for altered ER Ca(2+) regulation in Pdx-1-deficient states.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Transactivadores/genética , Animales , Línea Celular Tumoral , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Proteínas de Homeodominio/metabolismo , Humanos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Fluorescente/métodos , Células 3T3 NIH , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transactivadores/metabolismo
17.
Nat Genet ; 38(5): 583-8, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16642022

RESUMEN

An appropriate beta cell mass is pivotal for the maintenance of glucose homeostasis. Both insulin and IGF-1 are important in regulation of beta cell growth and function (reviewed in ref. 2). To define the roles of these hormones directly, we created a mouse model lacking functional receptors for both insulin and IGF-1 only in beta cells (betaDKO), as the hormones have overlapping mechanisms of action and activate common downstream proteins. Notably, betaDKO mice were born with a normal complement of islet cells, but 3 weeks after birth, they developed diabetes, in contrast to mild phenotypes observed in single mutants. Normoglycemic 2-week-old betaDKO mice manifest reduced beta cell mass, reduced expression of phosphorylated Akt and the transcription factor MafA, increased apoptosis in islets and severely compromised beta cell function. Analyses of compound knockouts showed a dominant role for insulin signaling in regulating beta cell mass. Together, these data provide compelling genetic evidence that insulin and IGF-I-dependent pathways are not critical for development of beta cells but that a loss of action of these hormones in beta cells leads to diabetes. We propose that therapeutic improvement of insulin and IGF-I signaling in beta cells might protect against type 2 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Factor I del Crecimiento Similar a la Insulina/fisiología , Insulina/fisiología , Islotes Pancreáticos/fisiopatología , Animales , Diabetes Mellitus Experimental/etiología , Humanos , Espectrometría de Masas , Ratones , Ratones Noqueados , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/fisiología , Receptor de Insulina/genética , Receptor de Insulina/fisiología
18.
Biophys J ; 107(11): 2723-33, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25468351

RESUMEN

Insulin is released from the islets of Langerhans in discrete pulses that are linked to synchronized oscillations of intracellular free calcium ([Ca(2+)]i). Associated with each synchronized oscillation is a propagating calcium wave mediated by Connexin36 (Cx36) gap junctions. A computational islet model predicted that waves emerge due to heterogeneity in ß-cell function throughout the islet. To test this, we applied defined patterns of glucose stimulation across the islet using a microfluidic device and measured how these perturbations affect calcium wave propagation. We further investigated how gap junction coupling regulates spatiotemporal [Ca(2+)]i dynamics in the face of heterogeneous glucose stimulation. Calcium waves were found to originate in regions of the islet having elevated excitability, and this heterogeneity is an intrinsic property of islet ß-cells. The extent of [Ca(2+)]i elevation across the islet in the presence of heterogeneity is gap-junction dependent, which reveals a glucose dependence of gap junction coupling. To better describe these observations, we had to modify the computational islet model to consider the electrochemical gradient between neighboring ß-cells. These results reveal how the spatiotemporal [Ca(2+)]i dynamics of the islet depend on ß-cell heterogeneity and cell-cell coupling, and are important for understanding the regulation of coordinated insulin release across the islet.


Asunto(s)
Señalización del Calcio , Uniones Comunicantes/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Glucosa/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Factores de Tiempo
20.
J Biol Chem ; 288(46): 33312-22, 2013 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-24100037

RESUMEN

Pulses of insulin released from pancreatic ß-cells maintain blood glucose in a narrow range, although the source of these pulses is unclear. We and others have proposed that positive feedback mediated by the glycolytic enzyme phosphofructokinase-1 (PFK1) enables ß-cells to generate metabolic oscillations via autocatalytic activation by its product fructose 1,6-bisphosphate (FBP). Although much indirect evidence has accumulated in favor of this hypothesis, a direct measurement of oscillating glycolytic intermediates has been lacking. To probe glycolysis directly, we engineered a family of inter- and intramolecular FRET biosensors based on the glycolytic enzyme pyruvate kinase M2 (PKAR; pyruvate kinase activity reporter), which multimerizes and is activated upon binding FBP. When introduced into Min6 ß-cells, PKAR FRET efficiency increased rapidly in response to glucose. Importantly, however, metabolites entering downstream of PFK1 (glyceraldehyde, pyruvate, and ketoisocaproate) failed to activate PKAR, consistent with sensor activation by FBP; the dependence of PKAR on FBP was further confirmed using purified sensor in vitro. Using a novel imaging modality for monitoring mitochondrial flavin fluorescence in mouse islets, we show that slow oscillations in mitochondrial redox potential stimulated by 10 mm glucose are in phase with glycolytic efflux through PKM2, measured simultaneously from neighboring islet ß-cells expressing PKAR. These results indicate that PKM2 activity in ß-cells is oscillatory and are consistent with pulsatile PFK1 being the mediator of slow glycolytic oscillations.


Asunto(s)
Relojes Biológicos/fisiología , Transferencia Resonante de Energía de Fluorescencia/métodos , Glucólisis/fisiología , Células Secretoras de Insulina/enzimología , Fosfofructoquinasa-1/metabolismo , Piruvato Quinasa/metabolismo , Animales , Línea Celular , Fructosadifosfatos/genética , Fructosadifosfatos/metabolismo , Humanos , Células Secretoras de Insulina/citología , Masculino , Ratones , Oxidación-Reducción , Fosfofructoquinasa-1/genética , Piruvato Quinasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA