Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Basic Clin Pharmacol Toxicol ; 127(3): 163-177, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32363722

RESUMEN

The prevalence of obesity and associated comorbidities such as type 2 diabetes and cardiovascular disease is increasing globally. Body-weight loss reduces the risk of morbidity and mortality in obese individuals, and thus, pharmacotherapies that induce weight loss can be of great value in improving the health and well-being of people living with obesity. Treatment with amylin and calcitonin receptor agonists reduces food intake and induces weight loss in several animal models, and a number of companies have started clinical testing for peptide analogues in the treatment of obesity and/or type 2 diabetes. Studies predominantly performed in rodent models show that amylin and the dual amylin/calcitonin receptor agonist salmon calcitonin achieve their metabolic effects by engaging areas in the brain associated with regulating homeostatic energy balance. In particular, signalling via neuronal circuits in the caudal hindbrain and the hypothalamus is implicated in mediating effects on food intake and energy expenditure. We review the current literature investigating the interaction of amylin/calcitonin receptor agonists with neurocircuits that induce the observed metabolic effects. Moreover, the status of drug development of amylin and calcitonin receptor agonists for the treatment of metabolic diseases is summarized.


Asunto(s)
Agonistas de los Receptores de Amilina/farmacología , Agonistas de los Receptores de Amilina/uso terapéutico , Enfermedades Metabólicas/tratamiento farmacológico , Receptores de Calcitonina/agonistas , Receptores de Calcitonina/uso terapéutico , Animales , Metabolismo Energético , Humanos , Hipotálamo , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Leptina , Ratones , Ratas , Rombencéfalo
2.
Diabetologia ; 63(3): 561-576, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31984442

RESUMEN

AIMS/HYPOTHESIS: Peptide hormones are first synthesised as larger, inactive precursors that are converted to their active forms by endopeptidase cleavage and post-translational modifications, such as amidation. Recent, large-scale genome-wide studies have suggested that two coding variants of the amidating enzyme, peptidylglycine α-amidating monooxygenase (PAM), are associated with impaired insulin secretion and increased type 2 diabetes risk. We aimed to elucidate the role of PAM in modulating beta cell peptide amidation, beta cell function and the development of diabetes. METHODS: PAM transcript and protein levels were analysed in mouse islets following induction of endoplasmic reticulum (ER) or cytokine stress, and PAM expression patterns were examined in human islets. To study whether haploinsufficiency of PAM accelerates the development of diabetes, Pam+/- and Pam+/+ mice were fed a low-fat diet (LFD) or high-fat diet (HFD) and glucose homeostasis was assessed. Since aggregates of the PAM substrate human islet amyloid polypeptide (hIAPP) lead to islet inflammation and beta cell failure, we also investigated whether PAM haploinsufficiency accelerated hIAPP-induced diabetes and islet amyloid formation in Pam+/- and Pam+/+ mice with beta cell expression of hIAPP. RESULTS: Immunostaining revealed high expression of PAM in alpha, beta and delta cells in human pancreatic islets. Pam mRNA and PAM protein expression were reduced in mouse islets following administration of an HFD, and in isolated islets following induction of ER stress with thapsigargin, or cytokine stress with IL-1ß, IFN-γ and TFN-α. Despite Pam+/- only having 50% PAM expression and enzyme activity as compared with Pam+/+ mice, glucose tolerance and body mass composition were comparable in the two models. After 24 weeks of HFD, both Pam+/- and Pam+/+ mice had insulin resistance and impaired glucose tolerance, but no differences in glucose tolerance, insulin sensitivity or plasma insulin levels were observed in PAM haploinsufficient mice. Islet amyloid formation and beta cell function were also similar in Pam+/- and Pam+/+ mice with beta cell expression of hIAPP. CONCLUSIONS/INTERPRETATION: Haploinsufficiency of PAM in mice does not accelerate the development of diet-induced obesity or hIAPP transgene-induced diabetes.


Asunto(s)
Amidina-Liasas/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Oxigenasas de Función Mixta/genética , Amidina-Liasas/fisiología , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Progresión de la Enfermedad , Epistasis Genética/fisiología , Femenino , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxigenasas de Función Mixta/fisiología , Ratas , Ratas Endogámicas Lew , Factores de Riesgo
3.
Handb Exp Pharmacol ; 245: 271-312, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29043504

RESUMEN

Human islet amyloid polypeptide or amylin (hA) is a 37-amino acid peptide hormone produced and co-secreted with insulin by pancreatic ß-cells. Under physiological conditions, hA regulates a broad range of biological processes including insulin release and slowing of gastric emptying, thereby maintaining glucose homeostasis. However, under the pathological conditions associated with type 2 diabetes mellitus (T2DM), hA undergoes a conformational transition from soluble random coil monomers to alpha-helical oligomers and insoluble ß-sheet amyloid fibrils or amyloid plaques. There is a positive correlation between hA oligomerization/aggregation, hA toxicity, and diabetes progression. Because the homeostatic balance between hA synthesis, release, and uptake is lost in diabetics and hA aggregation is a hallmark of T2DM, this chapter focuses on the biophysical and cell biology studies investigating molecular mechanisms of hA uptake, trafficking, and degradation in pancreatic cells and its relevance to h's toxicity. We will also discuss the regulatory role of endocytosis and proteolytic pathways in clearance of toxic hA species. Finally, we will discuss potential pharmacological approaches for specific targeting of hA trafficking pathways and toxicity in islet ß-cells as potential new avenues toward treatments of T2DM patients.


Asunto(s)
Amiloidosis/etiología , Diabetes Mellitus Tipo 2/etiología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Islotes Pancreáticos/metabolismo , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Endocitosis , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Complejo de la Endopetidasa Proteasomal/fisiología , Agregado de Proteínas
4.
Mol Endocrinol ; 30(4): 446-54, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26900721

RESUMEN

The islet in type 2 diabetes is characterized by ß-cell loss, increased ß-cell apoptosis, and islet amyloid derived from islet amyloid polypeptide (IAPP). When protein misfolding protective mechanisms are overcome, human IAPP (h-IAPP) forms membrane permeant toxic oligomers that induce ß-cell dysfunction and apoptosis. In humans with type 2 diabetes (T2D) and mice transgenic for h-IAPP, endoplasmic reticulum (ER) stress has been inferred from nuclear translocation of CCAAT/enhancer-binding protein homologous protein (CHOP), an established mediator of ER stress. To establish whether h-IAPP toxicity is mediated by ER stress, we evaluated diabetes onset and ß-cell mass in h-IAPP transgenic (h-TG) mice with and without deletion of CHOP in comparison with wild-type controls. Diabetes was delayed in h-TG CHOP(-/-) mice, with relatively preserved ß-cell mass and decreased ß-cell apoptosis. Deletion of CHOP attenuates dysfunction of the autophagy/lysosomal pathway in ß-cells of h-TG mice, uncovering a role for CHOP in mediating h-IAPP-induced dysfunction of autophagy. As deletion of CHOP delayed but did not prevent h-IAPP-induced ß-cell loss and diabetes, we examined CHOP-independent stress pathways. JNK, a target of the IRE-1pTRAF2 complex, and the Bcl-2 family proapoptotic mediator BIM, a target of ATF4, were comparably activated by h-IAPP expression in the presence and absence of CHOP. Therefore, although these studies affirm that CHOP is a mediator of h-IAPP-induced ER stress, it is not the only one. Therefore, suppression of CHOP alone is unlikely to be a durable therapeutic strategy to protect against h-IAPP toxicity because multiple stress pathways are activated.


Asunto(s)
Apoptosis , Células Secretoras de Insulina/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Factor de Transcripción CHOP/genética , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Estrés del Retículo Endoplásmico , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Factor de Transcripción CHOP/metabolismo
5.
J Biol Chem ; 290(24): 15121-32, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-25922077

RESUMEN

Deposition of ß cell toxic islet amyloid is a cardinal finding in type 2 diabetes. In addition to the main amyloid component islet amyloid polypeptide (IAPP), heparan sulfate proteoglycan is constantly present in the amyloid deposit. Heparan sulfate (HS) side chains bind to IAPP, inducing conformational changes of the IAPP structure and an acceleration of fibril formation. We generated a double-transgenic mouse strain (hpa-hIAPP) that overexpresses human heparanase and human IAPP but is deficient of endogenous mouse IAPP. Culture of hpa-hIAPP islets in 20 mm glucose resulted in less amyloid formation compared with the amyloid load developed in cultured islets isolated from littermates expressing human IAPP only. A similar reduction of amyloid was achieved when human islets were cultured in the presence of heparin fragments. Furthermore, we used CHO cells and the mutant CHO pgsD-677 cell line (deficient in HS synthesis) to explore the effect of cellular HS on IAPP-induced cytotoxicity. Seeding of IAPP aggregation on CHO cells resulted in caspase-3 activation and apoptosis that could be prevented by inhibition of caspase-8. No IAPP-induced apoptosis was seen in HS-deficient CHO pgsD-677 cells. These results suggest that ß cell death caused by extracellular IAPP requires membrane-bound HS. The interaction between HS and IAPP or the subsequent effects represent a possible therapeutic target whose blockage can lead to a prolonged survival of ß cells.


Asunto(s)
Amiloide/biosíntesis , Apoptosis/fisiología , Proteoglicanos de Heparán Sulfato/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Islotes Pancreáticos/metabolismo , Animales , Secuencia de Bases , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Cartilla de ADN , Glucuronidasa/metabolismo , Humanos , Islotes Pancreáticos/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
J Inorg Biochem ; 140: 143-52, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25108186

RESUMEN

Human islet amyloid polypeptide (hIAPP), the major component of the amyloid deposits found in the pancreatic islets of patients with type 2 diabetes mellitus (T2DM), plays a central role in the loss of insulin-secreting pancreatic beta cells. Misfolded hIAPP fibrillating in islet beta cells may be one of the causations for T2DM. Studies have showed that fibrosis of hIAPP was inhibited by copper compounds while hIAPP-induced cytotoxicity was greatly stimulated. In this study, the suppression effects of three different forms of copper compounds CuCl2, CuSO4 and Cu(Gly)2 on amyloid fibril formation were examined in vitro. The results demonstrated that Cu(II) could interact with hIAPP to suppress the fibrosis without involvement of the anions. The fibrosis of hIAPP was inhibited by CuCl2, CuSO4 and Cu(Gly)2 with a similar degree. The particle size of hIAPP aggregates was decreased, which was further confirmed in atomic force microscopy (AFM) and transmission electron microscopy (TEM) images. Moreover, approximative cytotoxicity-enhancing levels between CuCl2, CuSO4 and Cu(Gly)2 on hIAPP were also observed in INS-1 cells. Studies on the action mechanisms displayed that copper compounds increased hIAPP-induced cytotoxicity by facilitating apoptosis-promoting effect of hIAPP, which was dominated mainly by cation. Furthermore, Cu(II)-promoted ROS overproduction and mitochondrial disruption might be the main reason for the enhanced apoptosis. Taken together, our studies demonstrate clear interaction mechanisms of Cu(II) and hIAPP in pancreatic beta cells, and provide useful information for our understanding and treatment of T2DM.


Asunto(s)
Apoptosis/efectos de los fármacos , Cobre/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Islotes Pancreáticos/citología , Mitocondrias/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular Tumoral , Dicroismo Circular , Fluorescencia , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Islotes Pancreáticos/metabolismo , Microscopía de Fuerza Atómica , Mitocondrias/fisiología , Ratas
8.
Eur J Neurosci ; 40(7): 3055-66, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25040689

RESUMEN

Amylin reduces meal size by activating noradrenergic neurons in the area postrema (AP). Neurons in the AP also mediate the eating-inhibitory effects of salmon calcitonin (sCT), a potent amylin agonist, but the phenotypes of the neurons mediating its effect are unknown. Here we investigated whether sCT activates similar neuronal populations to amylin, and if its anorectic properties also depend on AP function. Male rats underwent AP lesion (APX) or sham surgery. Meal patterns were analysed under ad libitum and post-deprivation conditions. The importance of the AP in mediating the anorectic action of sCT was examined in feeding experiments of dose-response effects of sCT in APX vs. sham rats. The effect of sCT to induce Fos expression was compared between surgery groups, and relative to amylin. The phenotype of Fos-expressing neurons in the brainstem was examined by testing for the co-expression of dopamine beta hydroxylase (DBH) or tryptophan hydroxylase (TPH). By measuring the apposition of vesicular glutamate transporter-2 (VGLUT2)-positive boutons, potential glutamatergic input to amylin- and sCT-activated AP neurons was compared. Similar to amylin, an intact AP was necessary for sCT to reduce eating. Further, co-expression between Fos activation and DBH after amylin or sCT did not differ markedly, while co-localization of Fos and TPH was minor. Approximately 95% of neurons expressing Fos and DBH after amylin or sCT treatment were closely apposed to VGLUT2-positive boutons. Our study suggests that the hindbrain pathways engaged by amylin and sCT share many similarities, including the mediation by AP neurons.


Asunto(s)
Área Postrema/fisiología , Calcitonina/fisiología , Ingestión de Alimentos/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Neuronas/metabolismo , Animales , Área Postrema/efectos de los fármacos , Área Postrema/metabolismo , Calcitonina/farmacología , Dopamina beta-Hidroxilasa/análisis , Ingestión de Alimentos/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Masculino , Neuronas/efectos de los fármacos , Fenotipo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Triptófano Hidroxilasa/análisis , Proteína 2 de Transporte Vesicular de Glutamato/análisis
9.
J Clin Invest ; 124(8): 3292-4, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25036704

RESUMEN

A hallmark of type 2 diabetes is the reduction of pancreatic islet ß cell mass through induction of apoptosis and lack of regeneration. In most patients, ß cell dysfunction is associated with the presence of extracellular amyloid plaques adjacent to ß cells and intracellular toxic oligomers that are comprised of islet amyloid polypeptide (IAPP). In this issue of the JCI, three independent research groups reveal that a functional autophagy system normally prevents the accumulation of toxic IAPP oligomers in human IAPP-expressing murine models. Furthermore, mice expressing human IAPP but deficient for ß cell autophagy through genetic deletion of the autophagy initiator ATG7 developed ß cell apoptosis and overt diabetes. Together, these studies indicate that autophagy protects ß cells from the accumulation of toxic IAPP oligomers and suggest that enhancing autophagy may be a novel target for prevention of type 2 diabetes.


Asunto(s)
Autofagia/fisiología , Diabetes Mellitus Tipo 2/etiología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Animales , Femenino , Humanos , Masculino
10.
J Clin Invest ; 124(8): 3634-44, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25036706

RESUMEN

Pancreatic islets in patients with type 2 diabetes mellitus (T2DM) are characterized by loss of ß cells and formation of amyloid deposits derived from islet amyloid polypeptide (IAPP). Here we demonstrated that treatment of INS-1 cells with human IAPP (hIAPP) enhances cell death, inhibits cytoproliferation, and increases autophagosome formation. Furthermore, inhibition of autophagy increased the vulnerability of ß cells to the cytotoxic effects of hIAPP. Based on these in vitro findings, we examined the pathogenic role of hIAPP and its relation to autophagy in hIAPP-knockin mice. In animals fed a standard diet, hIAPP had no toxic effects on ß cell function; however, hIAPP-knockin mice did not exhibit a high-fat-diet-induced compensatory increase in ß cell mass, which was due to limited ß cell proliferation and enhanced ß cell apoptosis. Importantly, expression of hIAPP in mice with a ß cell-specific autophagy defect resulted in substantial deterioration of glucose tolerance and dispersed cytoplasmic expression of p62-associated toxic oligomers, which were otherwise sequestrated within p62-positive inclusions. Together, our results indicate that increased insulin resistance in combination with reduced autophagy may enhance the toxic potential of hIAPP and enhance ß cell dysfunction and progression of T2DM.


Asunto(s)
Autofagia/fisiología , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Animales , Proteína 7 Relacionada con la Autofagia , Ciclo Celular , Línea Celular , Supervivencia Celular , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Resistencia a la Insulina/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Polipéptido Amiloide de los Islotes Pancreáticos/toxicidad , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/toxicidad
11.
J Clin Invest ; 124(8): 3489-500, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25036708

RESUMEN

Type 2 diabetes (T2D) is characterized by a deficiency in ß cell mass, increased ß cell apoptosis, and extracellular accumulation of islet amyloid derived from islet amyloid polypeptide (IAPP), which ß cells coexpress with insulin. IAPP expression is increased in the context of insulin resistance, the major risk factor for developing T2D. Human IAPP is potentially toxic, especially as membrane-permeant oligomers, which have been observed to accumulate within ß cells of patients with T2D and rodents expressing human IAPP. Here, we determined that ß cell IAPP content is regulated by autophagy through p62-dependent lysosomal degradation. Induction of high levels of human IAPP in mouse ß cells resulted in accumulation of this amyloidogenic protein as relatively inert fibrils within cytosolic p62-positive inclusions, which temporarily averts formation of toxic oligomers. Mice hemizygous for transgenic expression of human IAPP did not develop diabetes; however, loss of ß cell-specific autophagy in these animals induced diabetes, which was attributable to accumulation of toxic human IAPP oligomers and loss of ß cell mass. In human IAPP-expressing mice that lack ß cell autophagy, increased oxidative damage and loss of an antioxidant-protective pathway appeared to contribute to increased ß cell apoptosis. These findings indicate that autophagy/lysosomal degradation defends ß cells against proteotoxicity induced by oligomerization-prone human IAPP.


Asunto(s)
Autofagia/fisiología , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Proteínas de Choque Térmico/deficiencia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Lisosomas/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Estructura Cuaternaria de Proteína , Proteolisis , Ratas , Ratas Transgénicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Sequestosoma-1
12.
PLoS Comput Biol ; 9(8): e1003211, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24009497

RESUMEN

IAPP, a 37 amino-acid peptide hormone belonging to the calcitonin family, is an intrinsically disordered protein that is coexpressed and cosecreted along with insulin by pancreatic islet ß-cells in response to meals. IAPP plays a physiological role in glucose regulation; however, in certain species, IAPP can aggregate and this process is linked to ß-cell death and Type II Diabetes. Using replica exchange molecular dynamics with extensive sampling (16 replicas per sequence and 600 ns per replica), we investigate the structure of the monomeric state of two species of aggregating peptides (human and cat IAPP) and two species of non-aggregating peptides (pig and rat IAPP). Our simulations reveal that the pig and rat conformations are very similar, and consist of helix-coil and helix-hairpin conformations. The aggregating sequences, on the other hand, populate the same helix-coil and helix-hairpin conformations as the non-aggregating sequence, but, in addition, populate a hairpin structure. Our exhaustive simulations, coupled with available peptide-activity data, leads us to a structure-activity relationship (SAR) in which we propose that the functional role of IAPP is carried out by the helix-coil conformation, a structure common to both aggregating and non-aggregating species. The pathological role of this peptide may have multiple origins, including the interaction of the helical elements with membranes. Nonetheless, our simulations suggest that the hairpin structure, only observed in the aggregating species, might be linked to the pathological role of this peptide, either as a direct precursor to amyloid fibrils, or as part of a cylindrin type of toxic oligomer. We further propose that the helix-hairpin fold is also a possible aggregation prone conformation that would lead normally non-aggregating variants of IAPP to form fibrils under conditions where an external perturbation is applied. The SAR relationship is used to suggest the rational design of therapeutics for treating diabetes.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Secuencia de Aminoácidos , Animales , Gatos , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Conformación Proteica , Pliegue de Proteína , Ratas , Alineación de Secuencia , Porcinos
13.
Sheng Li Xue Bao ; 65(3): 347-54, 2013 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-23788193

RESUMEN

The calcitonin gene-related peptide (CGRP) family mainly includes CGRPα, CGRPß, adrenomedullin, calcitonin and amylin. The members of CGRP family and their receptors are widely distributed in the central and peripheral nervous systems. Studies show that members of CGRP family such as CGRP and adrenomedullin play important roles in the transmission of nociceptive information. At spinal level, CGRP promotes the transmission of nociceptive information, spinal morphine tolerance, migraine, inflammatory pain and neuropathic pain. At superspinal level, CGRP suppresses the transmission of nociceptive information. Adrenomedullin is a pain-related neuropeptide which has recently been demonstrated. It facilitates the transmission of nociceptive information and is involved in the development and maintenance of opioid tolerance. The involvement of amylin and calcitonin in pain is not clear yet.


Asunto(s)
Analgésicos Opioides/farmacología , Péptido Relacionado con Gen de Calcitonina/fisiología , Tolerancia a Medicamentos , Nocicepción , Dolor/fisiopatología , Adrenomedulina/fisiología , Animales , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología
14.
Chem Biol Drug Des ; 82(4): 446-52, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23758815

RESUMEN

Human islet amyloid polypeptide (hIAPP) is highly amyloidogenic, whereas its homologs in rodents are non-amyloidogenic. This observed non-amyloidogenecity of rodent IAPP has been attributed to substitutions by proline in a region of IAPP that forms the core of the fibril. By employing molecular dynamics simulation, we have analyzed effects of position-specific proline substitution on amyloidogenesis of the core region of the hIAPP fibril (22-28). We depict that substitution to proline at the 25th position is primarily responsible for the loss of amyloidogenecity of the peptide. In addition, 25th and 26th double mutation to proline and valine has been observed to show significant fibril destabilizing ability. On the contrary, substitution at 28th position to proline has the least ability to destabilize the amyloid fibril. Results obtained from this study are particularly important to design variants of the existing antihyperglycemic drug with minimalistic mutation approach for use in patients with diabetes.


Asunto(s)
Amiloide/biosíntesis , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Prolina/química , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína
15.
Am J Physiol Endocrinol Metab ; 304(9): E944-50, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23482449

RESUMEN

Peptide YY(3-36) [PYY(3-36)] is postulated to act as a hormonal signal from gut to brain to inhibit food intake. PYY(3-36) potently reduces food intake when administered systemically or into the brain. If action of endogenous PYY(3-36) is necessary for normal satiation to occur, then pharmacological blockade of its receptors should increase food intake. Here, we determined the effects of iv infusion of Y1, Y2, and Y5 receptor antagonists (BIBP 3226, BIIE 0246, CGP 71683) during the first 3 h of the dark period on food intake in non-food-deprived rats. Our results showed that 1) Y2 receptor blockade reversed the anorexic response to iv infusion of PYY(3-36) but did not increase food intake when administered alone; 2) Y1 and Y5 receptor antagonists neither attenuated PYY(3-36)-induced anorexia nor altered food intake when given alone; and 3) Y2 receptor blockade attenuated anorexic responses to gastric infusions of casein hydrolysate and long-chain triglycerides, but not maltodextrin. Previous work showed that Y2 antagonist BIIE 0246 does not penetrate the blood-brain barrier. Together, these results support the hypothesis that gut PYY(3-36) action at Y2 receptors peripheral to the blood brain barrier plays an essential role in mediating satiety responses to gastric delivery of protein and long-chain triglycerides, but not polysaccharide.


Asunto(s)
Fragmentos de Péptidos/fisiología , Péptido YY/fisiología , Respuesta de Saciedad/fisiología , Animales , Anorexia/psicología , Colecistoquinina/fisiología , Ingestión de Alimentos/efectos de los fármacos , Hambre/fisiología , Intubación Gastrointestinal , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Masculino , Polisacáridos/farmacología , Hidrolisados de Proteína/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de la Hormona Gastrointestinal/antagonistas & inhibidores , Triglicéridos/farmacología
16.
Curr Pharm Des ; 19(38): 6739-48, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23530509

RESUMEN

The more common sporadic form of Alzheimer disease (SAD) and the metabolic syndrome are two highly prevalent pathological conditions of Western society due to incorrect diet, lifestyle, and vascular risk factors. Due to the increasing aging of populations, prevalence of AD in western industrialized countries will rise in the near future and, thus, new knowledge in the area of molecular biology and epigenetics will probably help to reverse the neurodegenerative process. Recent data have suggested metabolic syndrome as an independent risk factor for SAD. Furthermore, biological plausibility for this relationship has been framed within the metabolic cognitive syndrome concept, and some authors designed SAD as a brain diabetes or diabetes 3. Then, impaired signaling of insulin and from some adipokines involved in the so called adipoinsular axis, like leptin, ghrelin or amylin could give a metabolic basis to explain the origin and progression of SAD. Thus, dipokines like leptin, ghrelin and amylin, or their mimetic compounds, could contribuite to inhibit apoptosis and inflammation processes and, thus, generate protective responses in the nervous system. Moreover, these adipokines might promote the activation of a cognitive process which may retard or even partially reverse selected aspects of Alzheimer's disease or ageing memory loss.


Asunto(s)
Adipoquinas/fisiología , Enfermedad de Alzheimer/etiología , Metabolismo Energético , Síndrome Metabólico/complicaciones , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Complicaciones de la Diabetes/etiología , Ghrelina/fisiología , Glucosa/metabolismo , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Leptina/fisiología
18.
Scand J Gastroenterol ; 47(2): 191-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22229533

RESUMEN

OBJECTIVE: The current study used islet amyloid polypeptide (IAPP) knockout mice (KO mice) to investigate the physiological role of IAPP in the regulation of food intake (FI). MATERIAL AND METHODS: FI and body weight were measured in KO and wild-type (WT) mice for 27 weeks. In an additional short-term experiment, IAPP (25 pmol·kg(-1)min(-1)) was infused subcutaneously for 3 days in KO and WT mice, and FI, meal pattern, and body weight were analyzed. RESULTS: In the long-term experiment, no significant differences in body weight were seen between WT and KO mice at any point. FI, meal number, and meal size did not differ significantly between the groups in any of the five selected weeks that were studied. In the short-term experiment, FI decreased significantly during IAPP infusion in both WT and KO groups. FI was significantly lower in the KO mice compared with WT on days 1 and 2 (p < 0.05 and p < 0.01, respectively). CONCLUSIONS: The data showing no differences in FI and body weight were seen between KO and WT mice, indicating that FI can be controlled in the absence of IAPP. The more marked anorectic effect seen in the KO mice during IAPP infusion suggests that IAPP receptors and/or IAPP post-receptor signaling pathways are up-regulated in mice lacking endogenous IAPP.


Asunto(s)
Depresores del Apetito/farmacología , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Animales , Peso Corporal , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Factores de Tiempo
19.
Cell Mol Life Sci ; 69(12): 1947-65, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22193913

RESUMEN

Amylin is an important control of nutrient fluxes because it reduces energy intake, modulates nutrient utilization by inhibiting postprandial glucagon secretion, and increases energy disposal by preventing compensatory decreases of energy expenditure in weight-reduced individuals. The best investigated function of amylin which is cosecreted with insulin is to reduce eating by promoting meal-ending satiation. This effect is thought to be mediated by a stimulation of specific amylin receptors in the area postrema. Secondary brain sites to mediate amylin action include the nucleus of the solitary tract and the lateral parabrachial nucleus, which convey the neural signal to the lateral hypothalamic area and other hypothalamic nuclei. Amylin may also signal adiposity because plasma levels of amylin are increased in adiposity and because higher amylin concentrations in the brain result in reduced body weight gain and adiposity, while amylin receptor antagonists increase body adiposity. The central mechanisms involved in amylin's effect on energy expenditure are much less known. A series of recent experiments in animals and humans indicate that amylin is a promising option for anti-obesity therapy especially in combination with other hormones. The most extensive dataset is available for the combination therapy of amylin and leptin. Ongoing research focuses on the mechanisms of these interactions.


Asunto(s)
Metabolismo Energético/fisiología , Homeostasis , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Adiposidad/efectos de los fármacos , Adiposidad/fisiología , Animales , Fármacos Antiobesidad/farmacología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/fisiología , Metabolismo Energético/efectos de los fármacos , Femenino , Vaciamiento Gástrico/efectos de los fármacos , Vaciamiento Gástrico/fisiología , Humanos , Insulina/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/efectos de los fármacos , Leptina/fisiología , Masculino , Ratones , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Ratas , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/efectos de los fármacos , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/fisiología , Saciedad/efectos de los fármacos , Saciedad/fisiología
20.
J Biol Rhythms ; 26(5): 423-33, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21921296

RESUMEN

Type 2 diabetes mellitus (T2DM) is complex metabolic disease that arises as a consequence of interactions between genetic predisposition and environmental triggers. One recently described environmental trigger associated with development of T2DM is disturbance of circadian rhythms due to shift work, sleep loss, or nocturnal lifestyle. However, the underlying mechanisms behind this association are largely unknown. To address this, the authors examined the metabolic and physiological consequences of experimentally controlled circadian rhythm disruption in wild-type (WT) Sprague Dawley and diabetes-prone human islet amyloid polypeptide transgenic (HIP) rats: a validated model of T2DM. WT and HIP rats at 3 months of age were exposed to 10 weeks of either a normal light regimen (LD: 12:12-h light/dark) or experimental disruption in the light-dark cycle produced by either (1) 6-h advance of the light cycle every 3 days or (2) constant light protocol. Subsequently, blood glucose control, beta-cell function, beta-cell mass, turnover, and insulin sensitivity were examined. In WT rats, 10 weeks of experimental disruption of circadian rhythms failed to significantly alter fasting blood glucose levels, glucose-stimulated insulin secretion, beta-cell mass/turnover, or insulin sensitivity. In contrast, experimental disruption of circadian rhythms in diabetes-prone HIP rats led to accelerated development of diabetes. The mechanism subserving early-onset diabetes was due to accelerated loss of beta-cell function and loss of beta-cell mass attributed to increases in beta-cell apoptosis. Disruption of circadian rhythms may increase the risk of T2DM by accelerating the loss of beta-cell function and mass characteristic in T2DM.


Asunto(s)
Ritmo Circadiano/fisiología , Diabetes Mellitus Tipo 2/fisiopatología , Células Secretoras de Insulina/fisiología , Animales , Glucemia/metabolismo , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Células Secretoras de Insulina/citología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Masculino , Actividad Motora , Fotoperiodo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA