Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
J Clin Endocrinol Metab ; 102(1): 1-5, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27802092

RESUMEN

Context: The most common genetic cause of permanent neonatal diabetes mellitus is activating mutations in KCNJ11, which can usually be treated using oral sulfonylureas (SUs) instead of insulin injections, although some mutations are SU unresponsive. In this work, we provide a report of the pancreatic islet endocrine cell composition and area in a patient with an SU-unresponsive KCNJ11 mutation (p.G334D), in comparison with age-matched controls. Case Description: Pancreatic autopsy tissue sections from a 2-year-old female child diagnosed with KCNJ11-related diabetes at 4 days of age and 13 age-matched controls were stained with insulin, glucagon, somatostatin, pancreatic polypeptide, and Ki67 antibodies to determine islet endocrine cell composition and area. ß-cell ultrastructure was assessed by electron microscopic (EM) analysis. The patient's pancreas (sampling from head to tail) revealed insulin-positive cells in all regions. The pancreatic ß-cell (insulin) area was significantly reduced compared with controls: 0.50% ± 0.04% versus 1.67% ± 0.20%, respectively (P < 0.00001). There were no significant differences in α-cell (glucagon) or δ-cell (somatostatin) area. EM analysis revealed secretory granules with a dense core typical of mature ß-cells as well as granules with a lighter core characteristic of immature granules. Conclusions: Our results suggest that mechanisms exist that allow preservation of ß-cells in the absence of insulin secretion. It remains to be determined to what extent this reduction in ß-cells may be reversible.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Células Secretoras de Insulina/patología , Insulina/metabolismo , Mutación/genética , Canales de Potasio de Rectificación Interna/genética , Compuestos de Sulfonilurea/farmacología , Autopsia , Biomarcadores/análisis , Glucemia/análisis , Estudios de Casos y Controles , Preescolar , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Resistencia a Medicamentos , Femenino , Humanos , Lactante , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Pronóstico
2.
Proc Natl Acad Sci U S A ; 111(33): E3395-404, 2014 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-25092300

RESUMEN

Insulin provides a classical model of a globular protein, yet how the hormone changes conformation to engage its receptor has long been enigmatic. Interest has focused on the C-terminal B-chain segment, critical for protective self-assembly in ß cells and receptor binding at target tissues. Insight may be obtained from truncated "microreceptors" that reconstitute the primary hormone-binding site (α-subunit domains L1 and αCT). We demonstrate that, on microreceptor binding, this segment undergoes concerted hinge-like rotation at its B20-B23 ß-turn, coupling reorientation of Phe(B24) to a 60° rotation of the B25-B28 ß-strand away from the hormone core to lie antiparallel to the receptor's L1-ß2 sheet. Opening of this hinge enables conserved nonpolar side chains (Ile(A2), Val(A3), Val(B12), Phe(B24), and Phe(B25)) to engage the receptor. Restraining the hinge by nonstandard mutagenesis preserves native folding but blocks receptor binding, whereas its engineered opening maintains activity at the price of protein instability and nonnative aggregation. Our findings rationalize properties of clinical mutations in the insulin family and provide a previously unidentified foundation for designing therapeutic analogs. We envisage that a switch between free and receptor-bound conformations of insulin evolved as a solution to conflicting structural determinants of biosynthesis and function.


Asunto(s)
Insulina/metabolismo , Receptor de Insulina/metabolismo , Cristalografía por Rayos X , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Unión Proteica
3.
Genetics ; 196(2): 539-55, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24281154

RESUMEN

Drosophila melanogaster has been widely used as a model of human Mendelian disease, but its value in modeling complex disease has received little attention. Fly models of complex disease would enable high-resolution mapping of disease-modifying loci and the identification of novel targets for therapeutic intervention. Here, we describe a fly model of permanent neonatal diabetes mellitus and explore the complexity of this model. The approach involves the transgenic expression of a misfolded mutant of human preproinsulin, hINS(C96Y), which is a cause of permanent neonatal diabetes. When expressed in fly imaginal discs, hINS(C96Y) causes a reduction of adult structures, including the eye, wing, and notum. Eye imaginal discs exhibit defects in both the structure and the arrangement of ommatidia. In the wing, expression of hINS(C96Y) leads to ectopic expression of veins and mechano-sensory organs, indicating disruption of wild-type signaling processes regulating cell fates. These readily measurable "disease" phenotypes are sensitive to temperature, gene dose, and sex. Mutant (but not wild-type) proinsulin expression in the eye imaginal disc induces IRE1-mediated XBP1 alternative splicing, a signal for endoplasmic reticulum stress response activation, and produces global change in gene expression. Mutant hINS transgene tester strains, when crossed to stocks from the Drosophila Genetic Reference Panel, produce F1 adults with a continuous range of disease phenotypes and large broad-sense heritability. Surprisingly, the severity of mutant hINS-induced disease in the eye is not correlated with that in the notum in these crosses, nor with eye reduction phenotypes caused by the expression of two dominant eye mutants acting in two different eye development pathways, Drop (Dr) or Lobe (L), when crossed into the same genetic backgrounds. The tissue specificity of genetic variability for mutant hINS-induced disease has, therefore, its own distinct signature. The genetic dominance of disease-specific phenotypic variability in our model of misfolded human proinsulin makes this approach amenable to genome-wide association study in a simple F1 screen of natural variation.


Asunto(s)
Diabetes Mellitus/genética , Proinsulina/genética , Animales , Animales Modificados Genéticamente , Análisis por Conglomerados , Modelos Animales de Enfermedad , Drosophila melanogaster , Ojo/metabolismo , Femenino , Dosificación de Gen , Perfilación de la Expresión Génica , Humanos , Masculino , Mutación , Fenotipo , Proinsulina/química , Pliegue de Proteína , Carácter Cuantitativo Heredable , Transcriptoma , Transgenes , Alas de Animales/metabolismo
4.
J Biol Chem ; 288(40): 29013-23, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23935095

RESUMEN

Insulin is a major autoantigen in islet autoimmunity and progression to type 1 diabetes. It has been suggested that the insulin B-chain may be critical to insulin autoimmunity in type 1 diabetes. INS-IGF2 consists of the preproinsulin signal peptide, the insulin B-chain, and eight amino acids of the C-peptide in addition to 138 amino acids from the IGF2 gene. We aimed to determine the expression of INS-IGF2 in human pancreatic islets and autoantibodies in newly diagnosed children with type 1 diabetes and controls. INS-IGF2, expressed primarily in beta cells, showed higher levels of expression in islets from normal compared with donors with either type 2 diabetes (p = 0.006) or high HbA1c levels (p < 0.001). INS-IGF2 autoantibody levels were increased in newly diagnosed patients with type 1 diabetes (n = 304) compared with healthy controls (n = 355; p < 0.001). Displacement with cold insulin and INS-IGF2 revealed that more patients than controls had doubly reactive insulin-INS-IGF2 autoantibodies. These data suggest that INS-IGF2, which contains the preproinsulin signal peptide, the B-chain, and eight amino acids of the C-peptide may be an autoantigen in type 1 diabetes. INS-IGF2 and insulin may share autoantibody-binding sites, thus complicating the notion that insulin is the primary autoantigen in type 1 diabetes.


Asunto(s)
Autoinmunidad/inmunología , Insulina/inmunología , Islotes Pancreáticos/inmunología , Proteínas Mutantes Quiméricas/inmunología , Precursores de Proteínas/inmunología , Adolescente , Autoanticuerpos/sangre , Cromosomas Humanos Par 11/genética , ADN Complementario/genética , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Electroforesis en Gel de Poliacrilamida , Femenino , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Genoma Humano/genética , Humanos , Insulina/sangre , Insulina/genética , Factor II del Crecimiento Similar a la Insulina/genética , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Persona de Mediana Edad , Proteínas Mutantes Quiméricas/sangre , Análisis de Secuencia por Matrices de Oligonucleótidos , Biosíntesis de Proteínas , Precursores de Proteínas/sangre , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo , Transcripción Genética
5.
Nature ; 493(7431): 241-5, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23302862

RESUMEN

Insulin receptor signalling has a central role in mammalian biology, regulating cellular metabolism, growth, division, differentiation and survival. Insulin resistance contributes to the pathogenesis of type 2 diabetes mellitus and the onset of Alzheimer's disease; aberrant signalling occurs in diverse cancers, exacerbated by cross-talk with the homologous type 1 insulin-like growth factor receptor (IGF1R). Despite more than three decades of investigation, the three-dimensional structure of the insulin-insulin receptor complex has proved elusive, confounded by the complexity of producing the receptor protein. Here we present the first view, to our knowledge, of the interaction of insulin with its primary binding site on the insulin receptor, on the basis of four crystal structures of insulin bound to truncated insulin receptor constructs. The direct interaction of insulin with the first leucine-rich-repeat domain (L1) of insulin receptor is seen to be sparse, the hormone instead engaging the insulin receptor carboxy-terminal α-chain (αCT) segment, which is itself remodelled on the face of L1 upon insulin binding. Contact between insulin and L1 is restricted to insulin B-chain residues. The αCT segment displaces the B-chain C-terminal ß-strand away from the hormone core, revealing the mechanism of a long-proposed conformational switch in insulin upon receptor engagement. This mode of hormone-receptor recognition is novel within the broader family of receptor tyrosine kinases. We support these findings by photo-crosslinking data that place the suggested interactions into the context of the holoreceptor and by isothermal titration calorimetry data that dissect the hormone-insulin receptor interface. Together, our findings provide an explanation for a wealth of biochemical data from the insulin receptor and IGF1R systems relevant to the design of therapeutic insulin analogues.


Asunto(s)
Insulina/química , Insulina/metabolismo , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Animales , Sitios de Unión , Calorimetría , Bovinos , Línea Celular , Cristalografía por Rayos X , Humanos , Leucina/metabolismo , Ligandos , Modelos Moleculares , Unión Proteica , Estructura Secundaria de Proteína , Reproducibilidad de los Resultados
6.
Peptides ; 35(1): 42-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22421509

RESUMEN

Prohormone convertases (PCs) 1 and 2 are the primary endoproteases involved in the post-translational processing of proThyrotropin Releasing Hormone (proTRH) to give rise to TRH and other proposed biologically active non-TRH peptides. Previous evidence suggests that PC1 is responsible for most proTRH cleavage events. Here, we used the PC1 and PC2 knockout (KO) mouse models to examine the effects of PC1 or PC2 loss on proTRH processing. The PC1KO mouse presented a decrease in five proTRH-derived peptides, whereas the PC2KO mouse showed only lesser reduction in three TRH (Gln-His-Pro), TRH-Gly (Gln-His-Pro-Gly), and the short forms preproTRH(178-184) (pFQ(7)) and preproTRH(186-199) (pSE(14)) of pFE(22) (preproTRH(178-199)). Also, PC1KO and not PC2KO showed a decrease in pEH(24) indicating that PC1 is more important in generating this peptide in the mouse, which differs from previous studies using rat proTRH. Furthermore, downstream effects on thyroid hormone levels were evident in PC1KO mice, but not PC2KO mice suggesting that PC1 plays the more critical role in producing bioactive hypophysiotropic TRH. Yet loss of PC1 did not abolish TRH entirely indicating a complementary action for both enzymes in the normal processing of proTRH. We also show that PC2 alone is responsible for catalyzing the conversion of pFE(22) to pFQ(7) and pSE(14), all peptides implicated in regulation of suckling-induced prolactin release. Collectively, results characterize the specific roles of PC1 and PC2 in proTRH processing in vivo.


Asunto(s)
Fragmentos de Péptidos/biosíntesis , Proproteína Convertasa 1/genética , Proproteína Convertasa 2/genética , Precursores de Proteínas/biosíntesis , Hormona Liberadora de Tirotropina/biosíntesis , Secuencia de Aminoácidos , Animales , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Núcleo Hipotalámico Paraventricular/metabolismo , Proproteína Convertasa 1/deficiencia , Proproteína Convertasa 2/deficiencia , Homología de Secuencia de Aminoácido , Triyodotironina/biosíntesis
7.
J Biol Chem ; 287(18): 14703-17, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22396549

RESUMEN

The proprotein convertase 1/3 is expressed in the regulated secretory pathway of neural and endocrine cells. Its major function is in the post-translational processing and activation of precursor proteins. The PC1/3 knock-out (KO) mouse model has allowed us to elucidate its physiological functions in studies focused primarily on neuroendocrine tissues. However, PC1/3 is also expressed in cells of the immune system, mainly in macrophages. The present study explores the effects of innate immune challenge in the PC1/3 KO mouse. PC1/3 KO mice have an enlarged spleen with marked disorganization of the marginal zone and red pulp. Immunohistochemical studies using various markers demonstrate a depletion of dendritic cells in PC1/3 KO spleens. When challenged with lipopolysaccharide, PC1/3 KO mice are more susceptible to septic shock than wild-type controls or other PC KO mice, such as PC2 and PC7 null mice. Plasma levels of proinflammatory cytokines (IL-6, IL-1ß, and TNF-α) were very significantly elevated in PC1/3 KO mice, consistent with a hypercytokinemia, i.e. indicative of a major systemic uncontrolled inflammatory response or cytokine storm. Peritoneal macrophages isolated from PC1/3 KO mice also demonstrate elevated cytokine secretion when treated with LPS. Electron micrographs show morphological features indicating a prolonged activation of these cells following LPS stimulation. We also present evidence that the proinflammatory T(h)1 pathway is dominant in the PC1/3 KO mouse model. We conclude that aside from its important role in neuroendocrine functions PC1/3 also has an important role in the regulation of the innate immune system, most likely through the regulation of cytokine secretion in macrophages.


Asunto(s)
Citocinas/inmunología , Regulación Enzimológica de la Expresión Génica/inmunología , Inmunidad Innata , Macrófagos Peritoneales/inmunología , Proproteína Convertasa 1/inmunología , Animales , Citocinas/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/metabolismo , Enfermedades del Sistema Inmune/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/ultraestructura , Ratones , Ratones Noqueados , Proproteína Convertasa 1/biosíntesis , Proproteína Convertasa 1/genética , Células TH1/enzimología , Células TH1/metabolismo
8.
Methods Mol Biol ; 768: 3-11, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21805235

RESUMEN

Studies of the biosynthesis of insulin in a human insulinoma beginning in 1965 provided the first evidence for a precursor of insulin, the first such prohormone to be identified. Further studies with isolated rat islets then confirmed that the precursor became labeled more rapidly than insulin and later was converted to insulin by a proteolytic processing system located mainly within the secretory granules of the beta cell and was then stored or secreted. The precursor was designated "proinsulin" in 1967 and was isolated and sequenced from beef and pork sources. These structural studies confirmed that the precursor was a single polypeptide chain which began with the B chain of insulin, continued through a connecting segment of 30-35 amino acids and terminated with the A chain. Paired basic residues were identified at the sites of excision of the C-peptide. Human proinsulin and C-peptide were then similarly obtained and sequenced. The human C-peptide assay was developed and provided a useful tool for measuring insulin levels indirectly in diabetics treated with insulin. The discovery of other precursor proteins for a variety of peptide hormones, neuropeptides, or plasma proteins then followed, with all having mainly dibasic cleavage sites for processing. The subsequent discovery of a similar biosynthetic pathway in yeast led to the identification of eukaryotic families of specialized processing subtilisin-like endopeptidases coupled with carboxypeptidase B-like exopeptidases. Most neuroendocrine peptides are processed by two specialized members of this family - PC2 and/or PC1/3 - followed by carboxypeptidase E (CPE). This brief report concentrates mainly on the role of insulin biosynthesis in providing a useful early paradigm of precursor processing in the secretory pathway.


Asunto(s)
Insulina/biosíntesis , Proproteína Convertasa 1/metabolismo , Proproteína Convertasa 2/metabolismo , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Animales , Péptido C/biosíntesis , Carboxipeptidasa H/metabolismo , Bovinos , Historia del Siglo XX , Humanos , Células Secretoras de Insulina/enzimología , Insulinoma/metabolismo , Neuropéptidos/metabolismo , Proinsulina/biosíntesis , Precursores de Proteínas/historia , Ratas , Saccharomyces cerevisiae , Porcinos
9.
J Biol Chem ; 286(20): 17399-421, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21454641

RESUMEN

Insulin is a small but beautifully organized protein with a unique two-chain structure, the first protein to be sequenced. The mechanism of its biosynthesis invited much initial speculation but was finally clarified by the discovery of proinsulin, its single-chain precursor. The rich present-day field of protein precursor processing via post-translational proteolysis within the secretory pathway arose in the early 1970s as an offshoot of studies on insulin biosynthesis, which provided a novel paradigm for the generation of many other small neuroendocrine peptides. Before long, this mechanism was also found to play a role in the production of a much wider spectrum of proteins traversing the secretory pathway (receptors, growth factors, blood-clotting components, and even many viral envelope proteins) occurring in almost all eukaryotic cells. Indeed, yeast provided a key clue in the search for the proprotein convertases, the endoproteases that work along with carboxypeptidases and other modifying enzymes, such as the amidating enzyme complex (PAM), in converting inactive or less active precursor proteins into their fully active peptide products. In this "Reflections" article, I have tried to recount the people and events in my life that led to my involvement first in basic biochemical research and then on to insulin, proinsulin, and many relevant related areas that continue to fascinate and challenge my colleagues and me, as well as many other biomedical scientists today, as diabetes mellitus increasingly threatens human health throughout our contemporary world.


Asunto(s)
Bioquímica/historia , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Bioquímica/métodos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Insulina/historia
10.
Islets ; 3(1): 21-34, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21278490

RESUMEN

Diabetes (T1DM and T2DM) is characterized by a deficit in ß-cell mass. A broader understanding of human ß-cell replication mechanism is thus important to increase ß-cell proliferation for future therapeutic interventions. Here, we show that p27 (Kip1), a CDK inhibitor, is expressed abundantly in isolated adult human islets and interacts with various positive cell cycle regulatory proteins including D-type cyclins (D1, D2 and D3) and their kinase partners, CDK4 and CDK6. Also, we see interaction of cyclin E and its kinase partner, CDK2, with p27 suggesting a critical role of p27 as a negative cell cycle regulator in human islets. Our data demonstrate interaction of p27 with GSK-3 in ß-cells and show, employing rodent ß-cells (INS-1), isolated human islets and purified ß-cells derived from human islets, that siRNA-mediated depletion of GSK-3 or p27 or 1-AKP / BIO - mediated GSK-3 inhibition results in increased ß-cell proliferation. We also see reduction of p27 levels following GSK-3 inactivation or depletion. Our data show that serum induction of quiescent INS-1 cells leads to sequential phosphorylation of p27 on its S10 and T187 residues with faster kinetics for S10 corresponding with the decreased levels of p27. Altogether our findings indicate that p27 levels in ß-cells are stabilized by GSK-3 and thus p27 down regulation following GSK-3 depletion / inactivation plays a critical role in promoting ß-cell replication.


Asunto(s)
División Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Células Secretoras de Insulina/metabolismo , Adulto , Anciano , Animales , División Celular/genética , División Celular/fisiología , Línea Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/genética , Humanos , Células Secretoras de Insulina/fisiología , Masculino , Persona de Mediana Edad , ARN Interferente Pequeño/farmacología , Ratas
11.
Islets ; 2(5): 308-17, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21099329

RESUMEN

Pancreatic ß-cell response to glucose stimulation is governed by tightly regulated signaling pathways which have not been fully characterized. A screen for novel signaling intermediates identified Pim3 as a glucose-responsive gene in the ß cell, and here, we characterize its role in the regulation of ß-cell function. Pim3 expression in the ß-cell was first observed through microarray analysis on glucose-stimulated murine insulinoma (MIN6) cells where expression was strongly and transiently induced. In the pancreas, Pim3 expression exhibited similar dynamics and was restricted to the ß cell. Perturbation of Pim3 function resulted in enhanced glucose-stimulated insulin secretion, both in MIN6 cells and in isolated islets from Pim3-/- mice, where the augmentation was specifically seen in the second phase of secretion. Consequently, Pim3-/- mice displayed an increased glucose tolerance in vivo. Interestingly, Pim3-/- mice also exhibited increased insulin sensitivity. Glucose stimulation of isolated Pim3-/- islets resulted in increased phosphorylation of ERK1/2, a kinase involved in regulating ß-cell response to glucose. Pim3 was also found to physically interact with SOCS6 and SOCS6 levels were strongly reduced in Pim3-/- islets. Overexpression of SOCS6 inhibited glucose-induced ERK1/2 activation, strongly suggesting that Pim3 regulates ERK1/2 activity through SOCS6. These data reveal that Pim3 is a novel glucose-responsive gene in the ß cell that negatively regulates insulin secretion by inhibiting the activation of ERK1/2, and through its effect on insulin sensitivity, has potentially a more global function in glucose homeostasis.


Asunto(s)
Hiperglucemia , Insulina/metabolismo , Islotes Pancreáticos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Línea Celular , Tamaño de la Célula , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Resistencia a la Insulina , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Técnicas de Cultivo de Órganos , Especificidad de Órganos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
12.
Rev Endocr Metab Disord ; 11(3): 205-15, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20938745

RESUMEN

Over the last decade our insight into the causes of neonatal diabetes has greatly expanded. Neonatal diabetes was once considered a variant of type 1 diabetes that presented early in life. Recent advances in our understanding of this disorder have established that neonatal diabetes is not an autoimmune disease, but rather is a monogenic form of diabetes resulting from mutations in a number of different genes encoding proteins that play a key role in the normal function of the pancreatic beta-cell. Moreover, a correct genetic diagnosis can affect treatment and clinical outcome. This is especially true for patients with mutations in the genes KCNJ11 or ABCC8 that encode the two protein subunits (Kir6.2 and SUR1, respectively) of the ATP-sensitive potassium channel. These patients can be treated with oral sulfonylurea drugs with better glycemic control and quality of life. Recently, mutations in the insulin gene (INS) itself have been identified as another cause of neonatal diabetes. In this article, we review the role of INS mutations in the pathophysiology of neonatal diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/congénito , Diabetes Mellitus Tipo 1/genética , Enfermedades del Recién Nacido/genética , Insulina/genética , Mutación , Secuencia de Aminoácidos , Animales , Hiperinsulinismo Congénito/etiología , Hiperinsulinismo Congénito/genética , Humanos , Recién Nacido , Insulina/biosíntesis , Modelos Biológicos , Biología Molecular , Datos de Secuencia Molecular , Mutación/fisiología , Proinsulina/genética
13.
Islets ; 2(3): 149-55, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20657753

RESUMEN

The islet of Langerhans is a highly vascularized micro-organ consisting of not only ß-cells but multiple cell types such as α-, delta-, pancreatic polypeptide- and epsilon-cells that work together to regulate glucose homeostatis. We have recently proposed a new model of the neonatal islet formation in mice by a process of fission following contiguous endocrine cell proliferation in the form of branched cord-like structures in embryos and newborns. There exist large stretches of interconnected islet structures along large blood vessels in the neonatal pancreas, which, upon further development, segregate into smaller fragments (i.e., islets) that eventually become more spherical by internal proliferation as seen in the adult pancreas. α-cells span these elongated islet-like structures in the developing pancreas, which we hypothesize represent sites of fission and facilitate the eventual formation of discrete islets. The α-cells express both prohormone convertase 2 and 1/3 (PC 2 and PC 1/3, respectively), which resulted in the processing of the proglucagon precursor into glucagon-like peptide 1, thereby leading to local production of this important ß-cell growth factor. Furthermore, while α-cells in the adult basically only express PC 2, significant activation of PC 1/3 is also observed in mouse models of insulin resistance such as pregnant, ob/ ob, db/db and prediabetic NOD mice, which may be a common mechanism in proliferating ß-cells. Our study suggests an important role of α-cells for ß-cell proliferation and further for the endocrine cell network within an islet.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/fisiología , Proproteína Convertasa 1/metabolismo , Regeneración/fisiología , Animales , Animales Recién Nacidos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Embrión de Mamíferos , Femenino , Células Secretoras de Glucagón/fisiología , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Obesos , Modelos Animales , Estado Prediabético/metabolismo , Estado Prediabético/patología , Embarazo
14.
J Neurochem ; 114(1): 215-25, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20412386

RESUMEN

Neuropeptides are produced from larger precursors by limited proteolysis, first by endopeptidases and then by carboxypeptidases. Major endopeptidases required for these cleavages include prohormone convertase (PC) 1/3 and PC2. In this study, quantitative peptidomics analysis was used to characterize the specific role PC1/3 plays in this process. Peptides isolated from hypothalamus, amygdala, and striatum of PC1/3 null mice were compared with those from heterozygous and wild-type mice. Extracts were labeled with stable isotopic tags and fractionated by HPLC, after which relative peptide levels were determined using tandem mass spectrometry. In total, 92 peptides were found, of which 35 were known neuropeptides or related peptides derived from 15 distinct secretory pathway proteins: 7B2, chromogranin A and B, cocaine- and amphetamine-regulated transcript, procholecystokinin, proenkephalin, promelanin concentrating hormone, proneurotensin, propituitary adenylate cyclase-activating peptide, proSAAS, prosomatosatin, provasoactive intestinal peptide, provasopressin, secretogranin III, and VGF. Among the peptides derived from these proteins, approximately 1/3 were decreased in the PC1/3 null mice relative to wild-type mice, approximately 1/3 showed no change, and approximately 1/3 increased in PC1/3 null. Cleavage sites were analyzed in peptides that showed no change or that decreased in PC1/3 mice, and these results were compared with peptides that showed no change or decreased in previous peptidomic studies with PC2 null mice. Analysis of these sites showed that while PC1/3 and PC2 have overlapping substrate preferences, there are particular cleavage site residues that distinguish peptides preferred by each PC.


Asunto(s)
Encéfalo/metabolismo , Péptidos/metabolismo , Proproteína Convertasa 1/genética , Secuencia de Aminoácidos , Animales , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proproteína Convertasa 2/genética , Proteómica
15.
Proc Natl Acad Sci U S A ; 107(15): 6771-6, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20348418

RESUMEN

The C-terminal segment of the human insulin receptor alpha-chain (designated alphaCT) is critical to insulin binding as has been previously demonstrated by alanine scanning mutagenesis and photo-cross-linking. To date no information regarding the structure of this segment within the receptor has been available. We employ here the technique of thermal-factor sharpening to enhance the interpretability of the electron-density maps associated with the earlier crystal structure of the human insulin receptor ectodomain. The alphaCT segment is now resolved as being engaged with the central beta-sheet of the first leucine-rich repeat (L1) domain of the receptor. The segment is alpha-helical in conformation and extends 11 residues N-terminal of the classical alphaCT segment boundary originally defined by peptide mapping. This tandem structural element (alphaCT-L1) thus defines the intact primary insulin-binding surface of the apo-receptor. The structure, together with isothermal titration calorimetry data of mutant alphaCT peptides binding to an insulin minireceptor, leads to the conclusion that putative "insulin-mimetic" peptides in the literature act at least in part as mimics of the alphaCT segment as well as of insulin. Photo-cross-linking by novel bifunctional insulin derivatives demonstrates that the interaction of insulin with the alphaCT segment and the L1 domain occurs in trans, i.e., these components of the primary binding site are contributed by alternate alpha-chains within the insulin receptor homodimer. The tandem structural element defines a new target for the design of insulin agonists for the treatment of diabetes mellitus.


Asunto(s)
Péptidos/química , Receptor de Insulina/metabolismo , Animales , Sitios de Unión , Células CHO , Calorimetría/métodos , Cricetinae , Cricetulus , Reactivos de Enlaces Cruzados/química , Cristalografía por Rayos X/métodos , Dimerización , Diseño de Fármacos , Humanos , Modelos Moleculares , Conformación Molecular , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor de Insulina/agonistas
16.
J Neurochem ; 112(5): 1168-79, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19968759

RESUMEN

Prohormone convertase 2 (PC2) functions in the generation of neuropeptides from their precursors. A quantitative peptidomics approach was used to evaluate the role of PC2 in the processing of peptides in a variety of brain regions. Altogether, 115 neuropeptides or other peptides derived from secretory pathway proteins were identified. These peptides arise from 28 distinct secretory pathway proteins, including proenkephalin, proopiomelanocortin, prodynorphin, protachykinin A and B, procholecystokinin, and many others. Forty one of the peptides found in wild-type (WT) mice were not detectable in any of the brain regions of PC2 knockout mice, and another 24 peptides were present at levels ranging from 20% to 79% of WT levels. Most of the other peptides were not substantially affected by the mutation, with levels ranging from 80% to 120% of WT levels, and only three peptides were found to increase in one or more brain regions of PC2 knockout mice. Taken together, these results are consistent with a broad role for PC2 in neuropeptide processing, but with functional redundancy for many of the cleavages. Comparison of the cleavage sites affected by the absence of PC2 confirms previous suggestions that sequences with a Trp, Tyr, and/or Pro in the P1' or P2' position are preferentially cleaved by PC2 and not by other enzymes present in the secretory pathway.


Asunto(s)
Encéfalo/metabolismo , Neuropéptidos/biosíntesis , Proproteína Convertasa 2/fisiología , Secuencia de Aminoácidos , Animales , Cromatografía Líquida de Alta Presión/métodos , Ratones , Ratones Noqueados , Neuropéptidos/genética , Neuropéptidos/aislamiento & purificación , Proproteína Convertasa 2/deficiencia , Proteómica/métodos , Espectrometría de Masa por Ionización de Electrospray/métodos
17.
Biochem Biophys Res Commun ; 391(3): 1449-54, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20034470

RESUMEN

Mutations in the preproinsulin protein that affect processing of preproinsulin to proinsulin or lead to misfolding of proinsulin are associated with diabetes. We examined the subcellular localization and secretion of 13 neonatal diabetes-associated human proinsulin proteins (A24D, G32R, G32S, L35P, C43G, G47V, F48C, G84R, R89C, G90C, C96Y, S101C and Y108C) in rat INS-1 insulinoma cells. These mutant proinsulin proteins accumulate in the endoplasmic reticulum (ER) and are poorly secreted except for G84R and in contrast to wild-type and hyperproinsulinemia-associated mutant proteins (H34D and R89H) which were sorted to secretory granules and efficiently secreted. We also examined the effect of C96Y mutant proinsulin on the synthesis and secretion of wild-type insulin and observed a dominant-negative effect of the mutant proinsulin on the synthesis and secretion of wild-type insulin due to induction of the unfolded protein response and resulting attenuation of overall translation.


Asunto(s)
Diabetes Mellitus/metabolismo , Retículo Endoplásmico/metabolismo , Enfermedades del Recién Nacido/metabolismo , Proinsulina/metabolismo , Sustitución de Aminoácidos/genética , Animales , Línea Celular Tumoral , Diabetes Mellitus/genética , Humanos , Recién Nacido , Enfermedades del Recién Nacido/genética , Mutación , Proinsulina/genética , Pliegue de Proteína , Ratas
19.
Mamm Genome ; 20(8): 486-97, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19697080

RESUMEN

While diabetic patients often present with comorbid depression, the underlying mechanisms linking diabetes and depression are unknown. The Wistar Kyoto (WKY) rat is a well-known animal model of depression and stress hyperreactivity. In addition, the WKY rat is glucose intolerant and likely harbors diabetes susceptibility alleles. We conducted a quantitative trait loci (QTL) analysis in the segregating F(2) population of a WKY x Fischer 344 (F344) intercross. We previously published QTL analyses for depressive behavior and hypothalamic-pituitary-adrenal (HPA) activity in this cross. In this study we report results from the QTL analysis for multiple metabolic phenotypes, including fasting glucose, post-restraint stress glucose, postprandial glucose and insulin, and body weight. We identified multiple QTLs for each trait and many of the QTLs overlap with those previously identified using inbred models of type 2 diabetes (T2D). Significant correlations were found between metabolic traits and HPA axis measures, as well as forced swim test behavior. Several metabolic loci overlap with loci previously identified for HPA activity and forced swim behavior in this F(2) intercross, suggesting that the genetic mechanisms underlying these traits may be similar. These results indicate that WKY rats harbor diabetes susceptibility alleles and suggest that this strain may be useful for dissecting the underlying genetic mechanisms linking diabetes, HPA activity, and depression.


Asunto(s)
Depresión/genética , Diabetes Mellitus Tipo 2/genética , Sitios de Carácter Cuantitativo , Ratas , Animales , Mapeo Cromosómico , Depresión/complicaciones , Depresión/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratas Endogámicas WKY
20.
Diabetes Res Clin Pract ; 85(3): 252-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19595468

RESUMEN

Emerging reports on human islets emphasize distinct differences from the widely accepted prototype of rodent islets, raising questions over their suitability for human studies. Here we aim at elucidating architectural differences and similarities of human versus rodent islets. The cellular composition and architecture of human and rodent islets were compared through three-dimensional (3D) reconstructions. Physiological and pathological changes were examined using islets from various mouse models such as non-obese diabetic (NOD), ob/ob, db/db mice and during pregnancy. A subpopulation of human islets is composed of clusters of alpha-cells within the central beta-cell cores, while the overall proportion of alpha-cells varies among islets. In mouse islets under normal conditions, alpha-cells are localized in the islet periphery, but they do not envelop the entire beta-cell core, so that beta-cells are exposed on the outer layer of the islet, as in most human islets. Also, an increased proportion of alpha-cells within the central core is observed in the pancreas of mouse models exhibiting increased demand for insulin. In summary, human and mouse islets share common architectural features as endocrine micro-organs. Since these may hold a key to better understanding islet plasticity, our concept of the prototypic islet should be revised.


Asunto(s)
Islotes Pancreáticos/citología , Animales , Glucemia/metabolismo , División Celular , Tamaño de la Célula , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/patología , Femenino , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/patología , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/anatomía & histología , Islotes Pancreáticos/patología , Ratones , Ratones Obesos , Estado Prediabético/patología , Embarazo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...