Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Diabetes Metab Syndr ; 14(6): 1651-1657, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32898742

RESUMEN

BACKGROUND AND AIMS: Several studies raise concerns about the possible association of high selenium exposure with insulin resistance and type 2 diabetes. This in silico study proposes a possible mechanism of insulin resistance in the case of overexposure to selenium. METHOD: A study was carried out using molecular modeling, where cysteines of the insulin-receptor are replaced by selenocysteines. Calculation of the interaction energy of the receptor was performed in both cases with Auto Dock Tools and Vina 4.2 software to predict whether the substitution of amino acid could lead to destabilization of the protein-ligand complex and therefore possibly insulin resistance. Finally, the docked complex was analyzed by using BIOVIA Discovery Studio Visualizer to show the type of interactions between the ligands and insulin-receptor, and to determine the distance of the ligands from the binding site on insulin-receptor. RESULTS: The results show that the substitution of cysteine by selenocysteine in the insulin receptor does not lead to stabilization of the complex receptor/insulin, but to its disruption.In addition, the types and the number of bonds between insulin and its receptor in the two cases are different, where 7 strong bonds between insulin and its receptor were found in the case of the cysteine complex compared to 6 weak bonds in the second case. CONCLUSION: Findings of this study suggest that misincorporation of selenocysteines in insulin receptor could lead to destabilization of the insulin-receptor complex and therefore may possibly cause an insulin resistance.


Asunto(s)
Antígenos CD/metabolismo , Simulación por Computador , Resistencia a la Insulina , Insulina/metabolismo , Receptor de Insulina/metabolismo , Selenio/efectos adversos , Selenocisteína/metabolismo , Antígenos CD/química , Humanos , Insulina/química , Ligandos , Modelos Moleculares , Conformación Proteica , Receptor de Insulina/química , Selenio/metabolismo , Selenocisteína/química
2.
Growth Horm IGF Res ; 55: 101343, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32877816

RESUMEN

IR and insulin-like growth factor-1 receptor (IGF-1R) share high degree of sequence and structural similarity that hinders the development of anticancer drugs targeting IGF1R, which is dysregulated in many cancers. Although IR and IGF1R mediate their activities through similar signalling pathways, yet they show different physiological effects. The exact molecular mechanism(s) how IR and IGF1R exert their distinct functions remain largely unknown. Here, we performed in silico analysis and generated GFP-fusion proteins of wild type IR and its K1079R mutant to analyze their subcellular localization, cytoplasmic and nuclear activities in comparison to IGF1R and its K1055R mutant. We showed that, like K1055R mutation in IGF1R, K1079R mutation does not impede the subcellular localization and nuclear activities of IR. Although K1079R mutation significantly decreases the kinase activity of IR but not as much as K1055R mutation, which was seen to drastically reduce the kinase activity of IGF1R. Moreover, K1079 residue in IR is seen to be sitting in a pocket which is different than the allosteric inhibitor binding pocket present in its homologue (IGF1R). This is for the first time such a study has been conducted to identify structural differences between these receptors that could be exploited for designing small molecule allosteric inhibitor(s) of IGF1R as novel anti-cancer drugs.


Asunto(s)
Antígenos CD/química , Antineoplásicos/química , Mutación , Receptor IGF Tipo 1/química , Receptor de Insulina/química , Bibliotecas de Moléculas Pequeñas/química , Regulación Alostérica , Secuencia de Aminoácidos , Antígenos CD/genética , Antineoplásicos/farmacología , Simulación por Computador , Evaluación Preclínica de Medicamentos , Humanos , Pronóstico , Conformación Proteica , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Homología de Secuencia , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/farmacología
3.
Chemistry ; 25(36): 8513-8521, 2019 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-31012517

RESUMEN

Insulin analogues, mainstays in the modern treatment of diabetes mellitus, exemplify the utility of protein engineering in molecular pharmacology. Whereas chemical syntheses of the individual A and B chains were accomplished in the early 1960s, their combination to form native insulin remains inefficient because of competing disulfide pairing and aggregation. To overcome these limitations, we envisioned an alternative approach: pairwise substitution of cysteine residues with selenocysteine (Sec, U). To this end, CysA6 and CysA11 (which form the internal intrachain A6-A11 disulfide bridge) were each replaced with Sec. The A chain[C6U, C11U] variant was prepared by solid-phase peptide synthesis; while sulfitolysis of biosynthetic human insulin provided wild-type B chain-di-S-sulfonate. The presence of selenium atoms at these sites markedly enhanced the rate and fidelity of chain combination, thus solving a long-standing challenge in chemical insulin synthesis. The affinity of the Se-insulin analogue for the lectin-purified insulin receptor was indistinguishable from that of WT-insulin. Remarkably, the thermodynamic stability of the analogue at 25 °C, as inferred from guanidine denaturation studies, was augmented (ΔΔGu ≈0.8 kcal mol-1 ). In accordance with such enhanced stability, reductive unfolding of the Se-insulin analogue and resistance to enzymatic cleavage by Glu-C protease occurred four times more slowly than that of WT-insulin. 2D-NMR and X-ray crystallographic studies demonstrated a native-like three-dimensional structure in which the diselenide bridge was accommodated in the hydrophobic core without steric clash.


Asunto(s)
Disulfuros/química , Insulina/química , Selenio/química , Cristalografía por Rayos X , Cisteína/química , Humanos , Insulina/genética , Insulina/metabolismo , Unión Proteica , Pliegue de Proteína , Estabilidad Proteica , Estructura Terciaria de Proteína , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Selenocisteína/química , Termodinámica
4.
Protein Pept Lett ; 23(9): 836-47, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27397605

RESUMEN

Type 2 diabetes mellitus (T2DM) is characterized by abnormalities in carbohydrate, lipoprotein and lipid metabolism, leading to hyperglycemia and several other complications. Insulin is the major hormone regulating these facets by eliciting various biological responses through its receptor. Insulin exerts diverse effects on cells by targeting distinct functions such as gene expression, fatty acid synthesis, glucose transport and receptor translocation. Insulin mediates these effects through signaling pathways utilizing adapter molecules like small Gproteins, lipid and tyrosine kinases. The anomalous cell response in diabetic condition is due to altered expression/function of these molecules. Thiazolidinediones (TZD's), a class of oral hypoglycemic drugs, have shown to modify these responses, leading to insulin sensitizing effect(s). The TZD's are not only PPARγ agonists, but substantial insulin sensitizing activity is observed through its direct and indirect targets of the insulin receptor pathway, which contributes to its overall performance. TZD's alter(s) cell response via downstream players, primarily IRS, Akt/PKB, PKC, GLUT4, MEK, ERK and transcription factor PGC1α. Thus, this review will focus on the alteration(s) of these molecules in various cell types in diabetic condition and their regulation by TZD's. The physiological changes that occur at the molecular level in T2DM and their modulation by TZD's will provide insights into the key players involved and the potential drug targets for future drug development. The review further highlights the key markers to be evaluated in screening of any potential anti-diabetic agent, and to standardize therapy for T2DM based upon its modulation of the various signaling pathways.


Asunto(s)
Antígenos CD/química , Antígenos CD/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Tiazolidinedionas/farmacología , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/uso terapéutico
5.
Biomed Res Int ; 2016: 3187647, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27034931

RESUMEN

Diabetes mellitus (DM) is one of the most prevalent metabolic disorders which can affect the quality of life severely. Injectable insulin is currently being used to treat DM which is mainly associated with patient inconvenience. Small molecules that can act as insulin receptor (IR) agonist would be better alternatives to insulin injection. Herein, ten bioactive small compounds derived from Gymnema sylvestre (G. sylvestre) were chosen to determine their IR binding affinity and ADMET properties using a combined approach of molecular docking study and computational pharmacokinetic elucidation. Designing structural analogues were also performed for the compounds associated with toxicity and less IR affinity. Among the ten parent compounds, six were found to have significant pharmacokinetic properties with considerable binding affinity towards IR while four compounds were associated with toxicity and less IR affinity. Among the forty structural analogues, four compounds demonstrated considerably increased binding affinity towards IR and less toxicity compared with parent compounds. Finally, molecular interaction analysis revealed that six parent compounds and four analogues interact with the active site amino acids of IR. So this study would be a way to identify new therapeutics and alternatives to insulin for diabetic patients.


Asunto(s)
Antígenos CD/química , Diabetes Mellitus Experimental/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Receptor de Insulina/química , Bibliotecas de Moléculas Pequeñas/química , Aminoácidos/química , Animales , Glucemia , Diabetes Mellitus Experimental/patología , Gymnema sylvestre/química , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/química , Insulina/química , Insulina/metabolismo , Masculino , Simulación del Acoplamiento Molecular , Extractos Vegetales/química , Receptor de Insulina/agonistas , Bibliotecas de Moléculas Pequeñas/administración & dosificación
6.
J Biol Chem ; 284(21): 14586-96, 2009 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-19321436

RESUMEN

A central tenet of molecular biology holds that the function of a protein is mediated by its structure. An inactive ground-state conformation may nonetheless be enjoined by the interplay of competing biological constraints. A model is provided by insulin, well characterized at atomic resolution by x-ray crystallography. Here, we demonstrate that the activity of the hormone is enhanced by stereospecific unfolding of a conserved structural element. A bifunctional beta-strand mediates both self-assembly (within beta-cell storage vesicles) and receptor binding (in the bloodstream). This strand is anchored by an invariant side chain (Phe(B24)); its substitution by Ala leads to an unstable but native-like analog of low activity. Substitution by d-Ala is equally destabilizing, and yet the protein diastereomer exhibits enhanced activity with segmental unfolding of the beta-strand. Corresponding photoactivable derivatives (containing l- or d-para-azido-Phe) cross-link to the insulin receptor with higher d-specific efficiency. Aberrant exposure of hydrophobic surfaces in the analogs is associated with accelerated fibrillation, a form of aggregation-coupled misfolding associated with cellular toxicity. Conservation of Phe(B24), enforced by its dual role in native self-assembly and induced fit, thus highlights the implicit role of misfolding as an evolutionary constraint. Whereas classical crystal structures of insulin depict its storage form, signaling requires engagement of a detachable arm at an extended receptor interface. Because this active conformation resembles an amyloidogenic intermediate, we envisage that induced fit and self-assembly represent complementary molecular adaptations to potential proteotoxicity. The cryptic threat of misfolding poses a universal constraint in the evolution of polypeptide sequences.


Asunto(s)
Evolución Molecular , Insulina/química , Insulina/metabolismo , Pliegue de Proteína , Secuencia de Aminoácidos , Amiloide/efectos de los fármacos , Amiloide/efectos de la radiación , Amiloide/ultraestructura , Reactivos de Enlaces Cruzados/farmacología , Humanos , Insulina/análogos & derivados , Luz , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Fenilalanina/metabolismo , Pliegue de Proteína/efectos de los fármacos , Pliegue de Proteína/efectos de la radiación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Soluciones , Estereoisomerismo
7.
J Biol Chem ; 280(22): 20932-6, 2005 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-15799978

RESUMEN

Mutational analyses of the secreted recombinant insulin receptor extracellular domain have identified a ligand binding site composed of residues located in the L1 domain (amino acids 1-470) and at the C terminus of the alpha subunit (amino acids 705-715). To evaluate the physiological significance of this ligand binding site, we have transiently expressed cDNAs encoding full-length receptors with alanine mutations of the residues forming the functional epitopes of this binding site and determined their insulin binding properties. Insulin bound to wild-type receptors with complex kinetics, which were fitted to a two-component sequential model; the Kd of the high affinity component was 0.03 nM and that of the low affinity component was 0.4 nM. Mutations of Arg14, Phe64, Phe705, Glu706, Tyr708, Asn711, and Val715 inactivated the receptor. Alanine mutation of Asn15 resulted in a 20-fold decrease in affinity, whereas mutations of Asp12, Gln34, Leu36, Leu37, Leu87, Phe89, Tyr91, Lys121, Leu709, and Phe714 all resulted in 4-10-fold decreases. When the effects of the mutations were compared with those of the same mutations of the secreted recombinant receptor, significant differences were observed for Asn15, Leu37, Asp707, Leu709, Tyr708, Asn711, Phe714, and Val715, suggesting that the molecular basis for the interaction of each form of the receptor with insulin differs. We also examined the effects of alanine mutations of Asn15, Gln34, and Phe89 on insulin-induced receptor autophosphorylation. They had no effect on the maximal response to insulin but produced an increase in the EC50 commensurate with their effect on the affinity of the receptor for insulin.


Asunto(s)
Insulina/metabolismo , Receptor de Insulina/genética , Alanina/química , Sitios de Unión , Línea Celular , Clonación Molecular , Análisis Mutacional de ADN , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Epítopos/química , Humanos , Concentración 50 Inhibidora , Insulina/química , Cinética , Mutación , Fosforilación , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptor IGF Tipo 1/química , Receptor de Insulina/química , Transfección , Tirosina/química
8.
J Biol Chem ; 280(9): 8180-7, 2005 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-15611135

RESUMEN

Protein-tyrosine phosphatase-L1 (PTPL1, also known as FAP-1, PTP1E, PTP-BAS, and PTPN13) is mutated in a significant number of colorectal tumors and may play a role in down-regulating signaling responses mediated by phosphatidylinositol 3-kinase, although the precise substrates are as yet unknown. In this study, we describe a 1.8 A resolution crystal structure of a fully active fragment of PTPL1 encompassing the catalytic domain. PTPL1 adopts the standard PTP fold, albeit with an unusually positioned additional N-terminal helix, and shows an ordered phosphate in the active site. Interestingly, a positively charged pocket is located near the PTPL1 catalytic site, reminiscent of the second phosphotyrosine binding site in PTP1B, which is required to dephosphorylate peptides containing two adjacent phosphotyrosine residues (as occurs for example in the activated insulin receptor). We demonstrate that PTPL1, like PTP1B, interacts with and dephosphorylates a bis-phosphorylated insulin receptor peptide more efficiently than monophosphorylated peptides, indicating that PTPL1 may down-regulate the phosphatidylinositol 3-kinase pathway, by dephosphorylating insulin or growth factor receptors that contain tandem phosphotyrosines. The structure also reveals that four out of five PTPL1 mutations found in colorectal cancers are located on solvent-exposed regions remote from the active site, consistent with these mutants being normally active. In contrast, the fifth mutation, which changes Met-2307 to Thr, is close to the active site cysteine and decreases activity significantly. Our studies provide the first molecular description of the PTPL1 catalytic domain and give new insight into the function of PTPL1.


Asunto(s)
Neoplasias Colorrectales/genética , Mutación , Proteínas Tirosina Fosfatasas/química , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Clonación Molecular , Cristalografía por Rayos X , ADN/química , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Péptidos/química , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Fosfotirosina/química , Unión Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 13 , Receptor de Insulina/química , Transducción de Señal
9.
Biosci Biotechnol Biochem ; 68(5): 1067-72, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15170111

RESUMEN

Regulators for pancreatic amylase were examined. Rats were fed ad libitum a 20% amino acid (AA) mixture diet (Con), a 60% AA diet (HA), a branched-chain amino acid (BCAA)-rich diet (BC), or a diet supplemented with AA other than BCAA (OA) for 7 d, or fed the Con, HA, BC diets or diets supplemented with individual BCAA. Activity and mRNA levels of pancreatic amylase in the BC and HA groups were lower than those in the Con and OA groups. Leucine and isoleucine contributed to these effects of the BC diet. The mRNA levels correlated with individual pancreatic BCAA concentrations but not with plasma insulin level. In conclusion, dietary BCAA, especially leucine and isoleucine, may reduce amylase mRNA and activity in rats.


Asunto(s)
Aminoácidos de Cadena Ramificada/farmacología , Amilasas/metabolismo , Páncreas/enzimología , Aminoácidos de Cadena Ramificada/administración & dosificación , Aminoácidos de Cadena Ramificada/metabolismo , Amilasas/genética , Animales , Peso Corporal , Dieta , Suplementos Dietéticos , Ingestión de Alimentos , Regulación de la Expresión Génica , Insulina/sangre , Proteínas Sustrato del Receptor de Insulina , Fosfoproteínas/sangre , Fosfoproteínas/química , Fosfotirosina/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor de Insulina/sangre , Receptor de Insulina/química
10.
J Biol Chem ; 279(13): 12868-75, 2004 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-14722096

RESUMEN

Protein tyrosine phosphatase-1B (PTP-1B) is a negative regulator of insulin signaling. It is thought to carry out this role by interacting with and dephosphorylating the activated insulin receptor (IR). However, little is known regarding the nature of the cellular interaction between these proteins, especially because the IR is localized to the plasma membrane and PTP-1B to the endoplasmic reticulum. Using confocal microscopy and fluorescence resonance energy transfer (FRET), the interaction between PTP-1B and the IR was examined in co-transfected human embryonic kidney 293 cells. Biological activities were not significantly affected for either PTP-1B or the IR with the fusion of W1B-green fluorescent protein (GFP) to the N terminus of PTP-1B (W1B-PTP-1B) or the fusion of Topaz-GFP to the C terminus of the IR (Topaz-IR). FRET between W1B and Topaz was monitored in cells transfected with either wild type PTP-1B (W1B-PTP-1B) or the substrate-trapping form PTP-1B(D181A) (W1B-PTP-1B(D181A)) and Topaz-IR. Co-expression of W1B-PTP-1B with Topaz-IR resulted in distribution of Topaz-IR to the plasma membrane, but no FRET was obtained upon insulin treatment. In contrast, co-expression of W1B-PTP-1B(D181A) with Topaz-IR caused an increase in cytosolic Topaz-IR fluorescence and, in some cells, a significant basal FRET signal, suggesting that PTP-1B is interacting with the IR during its synthesis. Stimulation of these cells with insulin resulted in a rapid induction of FRET that increased over time and was localized to a perinuclear spot. Co-expression of Topaz-IR with a GFP-labeled RhoB endosomal marker and treatment of the cells with insulin identified a perinuclear endosome compartment as the site of localization. Furthermore, the insulin-induced FRET could be prevented by the treatment of the cells with a specific PTP-1B inhibitor. These results suggest that PTP-1B appears not only to interact with and dephosphorylate the insulin-stimulated IR in a perinuclear endosome compartment but is also involved in maintaining the IR in a dephosphorylated state during its biosynthesis.


Asunto(s)
Endosomas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Receptor de Insulina/metabolismo , Western Blotting , Línea Celular , Membrana Celular/metabolismo , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Transferencia Resonante de Energía de Fluorescencia , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Procesamiento de Imagen Asistido por Computador , Insulina/metabolismo , Cinética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Modelos Químicos , Fosforilación , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/química , Receptor de Insulina/química , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección
11.
EMBO Rep ; 4(3): 313-9, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12634852

RESUMEN

The dynamics of the interaction of the insulin receptor with a substrate-trapping mutant of protein-tyrosine phosphatase 1B (PTP1B) were monitored in living human embryonic kidney cells using bioluminescence resonance energy transfer (BRET). Insulin dose-dependently stimulates this interaction, which could be followed in real time for more than 30 minutes. The effect of insulin on the BRET signal could be detected at early time-points (30 seconds), suggesting that in intact cells the tyrosine-kinase activity of the insulin receptor is tightly controlled by PTP1B. Interestingly, the basal (insulin-independent) interaction of the insulin receptor with PTP1B was much weaker with a soluble form of the tyrosine-phosphatase than with the endoplasmic reticulum (ER)-targeted form. Inhibition of insulin-receptor processing using tunicamycin suggests that the basal interaction occurs during insulin-receptor biosynthesis in the ER. Therefore, localization of PTP1B in this compartment might be important for the regulation of insulin receptors during their biosynthesis.


Asunto(s)
Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/metabolismo , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Células Cultivadas , ADN Complementario/genética , Humanos , Riñón , Cinética , Mediciones Luminiscentes , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/genética , Receptor de Insulina/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Dominios Homologos src
12.
J Biol Chem ; 277(49): 47380-4, 2002 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-12270939

RESUMEN

The human insulin receptor is expressed as two isoforms that are generated by alternate splicing of its mRNA; the B isoform has 12 additional amino acids (718-729) encoded by exon 11 of the gene. The isoforms have been reported to have different ligand binding properties. To further characterize their insulin binding properties, we have performed structure-directed alanine-scanning mutagenesis of a major insulin binding site of the receptor, formed from the receptor L1 domain (amino acids 1-470) and amino acids 705-715 at the C terminus of the alpha subunit. Alanine mutants of each isoform were transiently expressed as recombinant secreted extracellular domain in 293 cells, and their insulin binding properties were evaluated by competitive binding assays. Mutation of Arg(86) and Phe(96) of each isoform resulted in receptors that were not secreted. The Kds of unmutated receptors were almost identical for both isoforms. Several new mutations compromising insulin binding were identified. In L1, mutation of Leu(37) decreased affinity 20- to 40-fold and mutations of Val(94), Glu(97), Glu(120), and Lys(121) 3 to 10-fold for each isoform. A number of mutations produced differential effects on the two isoforms. Mutation of Asn(15) in the L1 domain and Phe(714) at the C terminus of the alpha subunit inactivated the A isoform but only reduced the affinity of the B isoform 40- to 60-fold. At the C terminus of the alpha subunit, mutations of Asp(707), Val(713), and Val(715) produced 7- to 16-fold reductions in affinity of the A isoform but were without effect on the B isoform. In contrast, alanine mutations of Tyr(708) and Asn(711) inactivated the B isoform but only reduced the affinities of the A isoform 11- and 6-fold, respectively. In conclusion, alanine-scanning mutagenesis of the insulin receptor A and B isoforms has identified several new side chains contributing to insulin binding and indicates that the energetic contributions of certain side chains differ in each isoform, suggesting that different molecular mechanisms are used to obtain the same affinity.


Asunto(s)
Insulina/metabolismo , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Arginina/química , Línea Celular , ADN Complementario/metabolismo , Ensayo de Inmunoadsorción Enzimática , Epítopos , Exones , Humanos , Cinética , Ligandos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Oligonucleótidos/farmacología , Fenilalanina/química , Unión Proteica , Isoformas de Proteínas , Estructura Terciaria de Proteína , Transfección , Valina/química
13.
J Biol Chem ; 277(19): 16718-25, 2002 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-11875066

RESUMEN

To define the structures within the insulin receptor (IR) that are required for high affinity ligand binding, we have used IR fragments consisting of four amino-terminal domains (L1, cysteine-rich, L2, first fibronectin type III domain) fused to sequences encoded by exon 10 (including the carboxyl terminus of the alpha-subunit). The fragments contained one or both cysteine residues (amino acids 524 and 682) that form disulfides between alpha-subunits in native IR. A dimeric fragment designated IR593.CT (amino acids 1-593 and 704-719) bound (125)I-insulin with high affinity comparable to detergent-solubilized wild type IR and mIR.Fn0/Ex10 (amino acids 1-601 and 650-719) and greater than that of dimeric mIR.Fn0 (amino acids 1-601 and 704-719) and monomeric IR473.CT (amino acids 1-473 and 704-719). However, neither IR593.CT nor mIR.Fn0 exhibited negative cooperativity (a feature characteristic of the native insulin receptor and mIR.Fn0/Ex10), as shown by failure of unlabeled insulin to accelerate dissociation of bound (125)I-insulin. Anti-receptor monoclonal antibodies that recognize epitopes in the first fibronectin type III domain (amino acids 471-593) and inhibit insulin binding to wild type IR inhibited insulin binding to mIR.Fn0/Ex10 but not IR593.CT or mIR.Fn0. We conclude the following: 1) precise positioning of the carboxyl-terminal sequence can be a critical determinant of binding affinity; 2) dimerization via the first fibronectin domain alone can contribute to high affinity ligand binding; and 3) the second dimerization domain encoded by exon 10 is required for ligand cooperativity and modulation by antibodies.


Asunto(s)
Receptor de Insulina/química , Receptor de Insulina/fisiología , Aminoácidos/química , Animales , Anticuerpos/química , Sitios de Unión , Unión Competitiva , Western Blotting , Bovinos , ADN Complementario/metabolismo , Dimerización , Relación Dosis-Respuesta a Droga , Exones , Fibronectinas/química , Humanos , Immunoblotting , Concentración 50 Inhibidora , Factor I del Crecimiento Similar a la Insulina/farmacología , Ligandos , Pruebas de Precipitina , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Receptor de Insulina/inmunología , Proteínas Recombinantes/metabolismo , Factores de Tiempo
14.
J Biol Chem ; 276(50): 46933-40, 2001 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-11598120

RESUMEN

Low catalytic efficiency of protein kinases often results from intrasteric inhibition caused by the activation loop blocking the active site. In the insulin receptor's kinase domain, Asp-1161 and Tyr-1162 in the peptide substrate-like sequence of the unphosphorylated activation loop can interact with four invariant residues in the active site: Lys-1085, Asp-1132, Arg-1136, and Gln-1208. Contributions of these six residues to intrasteric inhibition were tested by mutagenesis, and the unphosphorylated kinase domains were characterized. The mutations Q1208S, K1085N, and Y1162F each relieved intrasteric inhibition, increasing catalytic efficiency but without changing the rate-limiting step of the reaction. The mutants R1136Q and D1132N were virtually inactive. Steric accessibility of the active site was ranked by relative changes in iodide quenching of intrinsic fluorescence, and A-loop conformation was ranked by limited tryptic cleavage. Together these ranked the openness of the active site cleft as R1136Q approximately D1132N > or = D1161A > Y1162F approximately K1085N > Q1208S > or = wild-type. These findings demonstrate the importance of specific invariant residues for intrasteric inhibition and show that diverse activation loop conformations can produce similar steady-state kinetic properties. This suggests a broader range of regulatory properties for the activation loop than expected from a simple off-versus-on switch for kinase activation.


Asunto(s)
Receptor de Insulina/química , Adenosina Difosfato/metabolismo , Animales , Arginina/química , Ácido Aspártico/química , Sitios de Unión , Catálisis , Clonación Molecular , ADN Complementario/metabolismo , Electroforesis en Gel de Poliacrilamida , Enlace de Hidrógeno , Cinética , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Mutación , Péptidos/química , Fosforilación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Ratas , Receptor de Insulina/metabolismo , Espectrometría de Fluorescencia , Especificidad por Sustrato , Factores de Tiempo , Tirosina/química
15.
J Biol Chem ; 276(36): 33419-27, 2001 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-11445579

RESUMEN

In this study, we report on the isolation of a PDZ domain protein, here designated as IIP-1, insulin-like growth factor-1 (IGF-1) receptor-interacting protein-1, which binds to the IGF-1 receptor, but not to the related insulin receptor, and which is involved in the regulation of cell motility. The interaction between the IGF-1 receptor and IIP-1 as well as a splice variant IIP-1/p26 was demonstrated in the yeast two-hybrid system. Using co-precipitation experiments, we confirmed the interaction in transfected cells as well as in vitro. Analysis of deletion mutants indicates that the PDZ domain of IIP-1 mediates interaction with the C-terminal tail of the IGF-1 receptor (serine-threonine-cysteine). This finding demonstrates that the C terminus of the IGF-1 receptor acts as novel PDZ domain binding site. Immunofluorescence analysis revealed an overlapping localization of IIP-1 and the IGF-1 receptor in the breast cancer cell line MCF-7. A functional connection between IIP-1 and the IGF-1 receptor is further supported by the finding that the level of expression of IIP-1 and the IGF-1 receptor strongly correlates in different normal and cancer cells. Furthermore, overexpression of IIP-1 resulted in an attenuation of migration of MCF-7 cells, which is one of the biological activities mediated by the IGF-1 signaling system.


Asunto(s)
Proteínas Portadoras/química , Receptor IGF Tipo 1/química , Receptor de Insulina/química , Células 3T3 , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas Portadoras/metabolismo , División Celular , Movimiento Celular , Clonación Molecular , Cisteína/química , ADN Complementario/metabolismo , Eliminación de Gen , Biblioteca de Genes , Glutatión Transferasa/metabolismo , Humanos , Immunoblotting , Células Jurkat , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutagénesis , Fosforilación , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Serina/química , Transducción de Señal , Treonina/química , Transfección , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos
16.
Science ; 284(5416): 974-7, 1999 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-10320380

RESUMEN

Insulin elicits a spectrum of biological responses by binding to its cell surface receptor. In a screen for small molecules that activate the human insulin receptor tyrosine kinase, a nonpeptidyl fungal metabolite (L-783,281) was identified that acted as an insulin mimetic in several biochemical and cellular assays. The compound was selective for insulin receptor versus insulin-like growth factor I (IGFI) receptor and other receptor tyrosine kinases. Oral administration of L-783,281 to two mouse models of diabetes resulted in significant lowering in blood glucose levels. These results demonstrate the feasibility of discovering novel insulin receptor activators that may lead to new therapies for diabetes.


Asunto(s)
Ascomicetos/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Indoles/farmacología , Insulina/farmacología , Receptor de Insulina/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Sitios de Unión , Glucemia/metabolismo , Células CHO , Cricetinae , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Activación Enzimática , Receptores ErbB/metabolismo , Prueba de Tolerancia a la Glucosa , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes/química , Hipoglucemiantes/metabolismo , Hipoglucemiantes/uso terapéutico , Indoles/química , Indoles/metabolismo , Indoles/uso terapéutico , Insulina/sangre , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina , Ratones , Ratones Mutantes , Ratones Obesos , Imitación Molecular , Fosfoproteínas/metabolismo , Fosforilación , Conformación Proteica/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/química , Transducción de Señal
17.
Antioxid Redox Signal ; 1(1): 45-53, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-11225731

RESUMEN

Previous studies on cultured skeletal muscle cells have indicated that the insulin-induced expression of GLUT4 transporter protein is inhibited by nitric oxide (NO). Therefore, we determined the effect of NO on the insulin-induced autophosphorylation of the insulin receptor kinase (IRK), i.e., the first step in the insulin-mediated signal transduction pathway. The experiments showed that the insulin-induced autophosphorylation of the insulin receptor beta-chain is strongly inhibited by the NO donors 1,1-diethyl-2-hydroxy-2-nitrosohydrazine (DEA-NO) or S-nitroso-N-acetylpenicillamine (SNAP). The inhibitory effect was ameliorated in cells depleted of glutathione (GSH), suggesting the possibility that S-nitroso-glutathione may operate as an intermediate NO donor. Complementary experiments with different Cys --> Ala mutant proteins showed, surprisingly, that all mutant proteins were inhibited by DEA-NO. Three-dimensional models of the nonphosphorylated IR beta-chain nitrosylated at the accessible cysteine residues 1056, 1138, 1234, or 1245 revealed that derivatization of any of these four cysteine residues leads essentially to the same structural changes of the IRK domain. These changes involve a movement of the amino-terminal lobe against the carboxy-terminal lobe in a direction opposite to the direction of the "lobe closure" that was previously proposed to facilitate the accessibility for ATP and the expression of catalytic activity. Our findings suggest that the occurrence of several functionally relevant cysteine residues in distinct regions of the IRK protein increases the probability of regulatory redox interactions and thus the redox sensitivity of the IRK.


Asunto(s)
Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Receptor de Insulina/metabolismo , Animales , Butionina Sulfoximina/farmacología , Células CHO , Cricetinae , Medio de Cultivo Libre de Suero , Glutatión/metabolismo , Humanos , Hidrazinas/farmacología , Insulina/farmacología , Modelos Moleculares , Óxidos de Nitrógeno , Penicilamina/análogos & derivados , Penicilamina/farmacología , Fosforilación/efectos de los fármacos , Pruebas de Precipitina , Conformación Proteica , Receptor de Insulina/química , Receptor de Insulina/genética , S-Nitroso-N-Acetilpenicilamina , Transfección
18.
FEBS Lett ; 395(2-3): 286-92, 1996 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-8898113

RESUMEN

Two new kinases of Dictyostelium discoideum were identified by screening of a (lambda)gt11 expression library with a phosphotyrosine specific antibody. Amino-acid sequences derived from cDNA and genomic clones indicate that DPYK3 is a protein of 150 kDa and DPYK4, a protein of 75 kDa. The C-terminal fragments of each protein were produced in Escherichia coli and shown to be autocatalytically phosphorylated at tyrosine residues. A common feature of these kinases is the presence of two different sequence stretches in tandem that are related to kinase catalytic domains. The sequence relationships of DPYK3 and 4 to other protein kinases, and the positions of their catalytic domain sequences within the phylogenetic tree of protein kinases were analysed. Domains I of both kinases and domain II of DPYK3 constitute, together with the catalytic domains of two previously described tyrosine kinases of D. discoideum, a branch of their own, separate from the tyrosine kinase domains in sensu strictu. Domain II in DPYK4 is found on a different branch close to serine/threonine kinases.


Asunto(s)
Dictyostelium/enzimología , Filogenia , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/clasificación , Secuencia de Aminoácidos , Animales , Sitios de Unión , Clonación Molecular , Secuencia de Consenso , ADN Complementario , Dictyostelium/genética , Escherichia coli , Humanos , Ratones , Datos de Secuencia Molecular , Plantas , Estructura Secundaria de Proteína , Proteínas Tirosina Quinasas/biosíntesis , Receptor de Insulina/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/clasificación , Homología de Secuencia de Aminoácido
19.
Artículo en Inglés | MEDLINE | ID: mdl-8896329

RESUMEN

To explore the initial stages of olfactory transduction, we have used biochemical techniques to characterize proteins associated with the dendritic plasma membrane from the olfactory receptor neurons of the spiny lobster Panulirus argus. In particular, we have studied proteins that interact with taurine, an amino acid that is an important odorant for this species. The cross-linker bis(sulfosuccinimidyl)suberate (BS3) was used to covalently link [3H]-taurine to cell surface proteins on membrane from the aesthetasc (olfactory) sensilla of the lateral filament of the antennule. A radioligand-receptor binding assay was used to show that this cross-linkage was highly specific for taurine at 0.2 mM BS3. In inhibition studies, of all the unlabeled odorants tested at excess concentrations (taurine, L-glutamate, adenosine-5'-monophosphate), only taurine significantly inhibited the cross-linkage of [3H]-taurine to the membrane. Membranes containing cross-linked proteins were solubilized, and proteins were separated on SDS-PAGE and examined with autoradiography. Bands with molecular weights of 100, 82, 62, 51, and 34kD were evident on the gels. However, only the 100 and 62 kD bands were consistently labeled with [3H]-taurine, and this labeling was completely inhibited in the presence of excess unlabeled taurine but not adenosine-5'monophosphate. The taurine-evoked behavioral search response of spiny lobsters was significantly reduced following treatment of their antennules with BS3 + taurine as compared with animals treated with BS3 alone, suggesting that the taurine-labeled binding proteins include taurine receptor proteins involved in the first stage of olfactory transduction.


Asunto(s)
Bulbo Olfatorio/química , Receptor de Insulina/química , Receptores de Neurotransmisores/química , Adenosina Monofosfato/metabolismo , Animales , Autorradiografía , Sitios de Unión , Reactivos de Enlaces Cruzados/metabolismo , Electroforesis en Gel de Poliacrilamida , Conducta Exploratoria/efectos de los fármacos , Ácido Glutámico/metabolismo , Peso Molecular , Nephropidae , Succinimidas/metabolismo , Taurina/metabolismo
20.
J Biol Chem ; 270(14): 8122-30, 1995 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-7713916

RESUMEN

The deduced primary sequence of the cytoplasmic protein-tyrosine kinase domain of the insulin receptor contains a conserved kinase homology region (receptor residues 1002-1257) flanked by a juxtamembrane region and a C-terminal tail. A soluble 48-kDa derivative (residues 959-1355) containing these regions but lacking the first six residues of the juxtamembrane region had earlier been synthesized in Sf9 cells using a baculovirus expression system. The catalytic core of the kinase domain was studied first by proteolytic analysis of the 48-kDa kinase and then by expressing a series of truncated kinase domains in transiently transfected COS cells. Based on these studies, two core kinases of 34 (residues 985-1283) and 35 (residues 978-1283) kDa, respectively, were overexpressed in Sf9 cells. Biochemical characterization of the 35-kDa kinase revealed that the core kinase conserved the major functional properties of the native receptor kinase domain. Activity of the 35-kDa kinase toward a synthetic peptide increased more than 200-fold upon autophosphorylation, which occurred exclusively at Tyr-1158, Tyr-1162, and Tyr-1163; the largest increase was observed between bis- and trisphosphorylation of the kinase. The activated 35- and 48-kDa kinases were similar with respect to specific activity and ATP and Mg2+ requirements for peptide phosphorylation. Moreover, autophosphorylation appeared to initiate predominantly at Tyr-1162, immediately followed by phosphorylation at Tyr-1158 and then at Tyr-1163. The rate of autophosphorylation was dependent on enzyme concentration, consistent with a trans-phosphorylation mechanism. Finally, the 35-kDa kinase was crystallized, making possible elucidation of its three-dimensional structure by x-ray crystallography.


Asunto(s)
Receptor de Insulina/química , Receptor de Insulina/genética , Secuencia de Aminoácidos , Animales , Baculoviridae/genética , Secuencia de Bases , Catálisis , Línea Celular , Clonación Molecular , Cristalización , ADN Complementario , Humanos , Datos de Secuencia Molecular , Fosforilación , Receptor de Insulina/metabolismo , Spodoptera , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA