Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 295(10): 3080-3098, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32005662

RESUMEN

Globular protein sequences encode not only functional structures (the native state) but also protein foldability, i.e. a conformational search that is both efficient and robustly minimizes misfolding. Studies of mutations associated with toxic misfolding have yielded insights into molecular determinants of protein foldability. Of particular interest are residues that are conserved yet dispensable in the native state. Here, we exploited the mutant proinsulin syndrome (a major cause of permanent neonatal-onset diabetes mellitus) to investigate whether toxic misfolding poses an evolutionary constraint. Our experiments focused on an invariant aromatic motif (PheB24-PheB25-TyrB26) with complementary roles in native self-assembly and receptor binding. A novel class of mutations provided evidence that insulin can bind to the insulin receptor (IR) in two different modes, distinguished by a "register shift" in this motif, as visualized by molecular dynamics (MD) simulations. Register-shift variants are active but defective in cellular foldability and exquisitely susceptible to fibrillation in vitro Indeed, expression of the corresponding proinsulin variant induced endoplasmic reticulum stress, a general feature of the mutant proinsulin syndrome. Although not present among vertebrate insulin and insulin-like sequences, a prototypical variant ([GlyB24]insulin) was as potent as WT insulin in a rat model of diabetes. Although in MD simulations the shifted register of receptor engagement is compatible with the structure and allosteric reorganization of the IR-signaling complex, our results suggest that this binding mode is associated with toxic misfolding and so is disallowed in evolution. The implicit threat of proteotoxicity limits sequence variation among vertebrate insulins and insulin-like growth factors.


Asunto(s)
Evolución Molecular , Insulina/análogos & derivados , Secuencias de Aminoácidos , Animales , Sitios de Unión , Glucemia/análisis , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Células HEK293 , Humanos , Insulina/metabolismo , Insulina/uso terapéutico , Simulación de Dinámica Molecular , Unión Proteica , Pliegue de Proteína , Estabilidad Proteica , Ratas , Receptor de Insulina/metabolismo , Relación Estructura-Actividad , Termodinámica
2.
J Biol Chem ; 293(1): 47-68, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29114035

RESUMEN

Thermal degradation of insulin complicates its delivery and use. Previous efforts to engineer ultra-stable analogs were confounded by prolonged cellular signaling in vivo, of unclear safety and complicating mealtime therapy. We therefore sought an ultra-stable analog whose potency and duration of action on intravenous bolus injection in diabetic rats are indistinguishable from wild-type (WT) insulin. Here, we describe the structure, function, and stability of such an analog, a 57-residue single-chain insulin (SCI) with multiple acidic substitutions. Cell-based studies revealed native-like signaling properties with negligible mitogenic activity. Its crystal structure, determined as a novel zinc-free hexamer at 2.8 Å, revealed a native insulin fold with incomplete or absent electron density in the C domain; complementary NMR studies are described in the accompanying article. The stability of the analog (ΔGU 5.0(±0.1) kcal/mol at 25 °C) was greater than that of WT insulin (3.3(±0.1) kcal/mol). On gentle agitation, the SCI retained full activity for >140 days at 45 °C and >48 h at 75 °C. These findings indicate that marked resistance to thermal inactivation in vitro is compatible with native duration of activity in vivo Further, whereas WT insulin forms large and heterogeneous aggregates above the standard 0.6 mm pharmaceutical strength, perturbing the pharmacokinetic properties of concentrated formulations, dynamic light scattering, and size-exclusion chromatography revealed only limited SCI self-assembly and aggregation in the concentration range 1-7 mm Such a combination of favorable biophysical and biological properties suggests that SCIs could provide a global therapeutic platform without a cold chain.


Asunto(s)
Hipoglucemiantes/química , Insulina/análogos & derivados , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Humanos , Hipoglucemiantes/metabolismo , Insulina/genética , Insulina/metabolismo , Modelos Moleculares , Agregado de Proteínas , Conformación Proteica , Ingeniería de Proteínas , Multimerización de Proteína , Estabilidad Proteica , Solubilidad , Porcinos , Temperatura
3.
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
4.
J Biol Chem ; 291(25): 12978-90, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27129279

RESUMEN

Crystallographic studies of insulin bound to receptor domains have defined the primary hormone-receptor interface. We investigated the role of Tyr(B26), a conserved aromatic residue at this interface. To probe the evolutionary basis for such conservation, we constructed 18 variants at B26. Surprisingly, non-aromatic polar or charged side chains (such as Glu, Ser, or ornithine (Orn)) conferred high activity, whereas the weakest-binding analogs contained Val, Ile, and Leu substitutions. Modeling of variant complexes suggested that the B26 side chains pack within a shallow depression at the solvent-exposed periphery of the interface. This interface would disfavor large aliphatic side chains. The analogs with highest activity exhibited reduced thermodynamic stability and heightened susceptibility to fibrillation. Perturbed self-assembly was also demonstrated in studies of the charged variants (Orn and Glu); indeed, the Glu(B26) analog exhibited aberrant aggregation in either the presence or absence of zinc ions. Thus, although Tyr(B26) is part of insulin's receptor-binding surface, our results suggest that its conservation has been enjoined by the aromatic ring's contributions to native stability and self-assembly. We envisage that such classical structural relationships reflect the implicit threat of toxic misfolding (rather than hormonal function at the receptor level) as a general evolutionary determinant of extant protein sequences.


Asunto(s)
Insulina/química , Secuencia de Aminoácidos , Secuencia Conservada , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estabilidad Proteica , Estructura Secundaria de Proteína , Subunidades de Proteína/química , Receptor de Insulina/química , Relación Estructura-Actividad , Tirosina/química
5.
J Biol Chem ; 291(53): 27023-27041, 2016 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-27875310

RESUMEN

Insulin, a protein critical for metabolic homeostasis, provides a classical model for protein design with application to human health. Recent efforts to improve its pharmaceutical formulation demonstrated that iodination of a conserved tyrosine (TyrB26) enhances key properties of a rapid-acting clinical analog. Moreover, the broad utility of halogens in medicinal chemistry has motivated the use of hybrid quantum- and molecular-mechanical methods to study proteins. Here, we (i) undertook quantitative atomistic simulations of 3-[iodo-TyrB26]insulin to predict its structural features, and (ii) tested these predictions by X-ray crystallography. Using an electrostatic model of the modified aromatic ring based on quantum chemistry, the calculations suggested that the analog, as a dimer and hexamer, exhibits subtle differences in aromatic-aromatic interactions at the dimer interface. Aromatic rings (TyrB16, PheB24, PheB25, 3-I-TyrB26, and their symmetry-related mates) at this interface adjust to enable packing of the hydrophobic iodine atoms within the core of each monomer. Strikingly, these features were observed in the crystal structure of a 3-[iodo-TyrB26]insulin analog (determined as an R6 zinc hexamer). Given that residues B24-B30 detach from the core on receptor binding, the environment of 3-I-TyrB26 in a receptor complex must differ from that in the free hormone. Based on the recent structure of a "micro-receptor" complex, we predict that 3-I-TyrB26 engages the receptor via directional halogen bonding and halogen-directed hydrogen bonding as follows: favorable electrostatic interactions exploiting, respectively, the halogen's electron-deficient σ-hole and electronegative equatorial band. Inspired by quantum chemistry and molecular dynamics, such "halogen engineering" promises to extend principles of medicinal chemistry to proteins.


Asunto(s)
Química Farmacéutica , Insulina/análogos & derivados , Receptor de Insulina/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Halógenos , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Insulina/química , Insulina/genética , Insulina/metabolismo , Modelos Moleculares , Fenilalanina/química , Fenilalanina/genética , Fenilalanina/metabolismo , Unión Proteica , Receptor de Insulina/química , Relación Estructura-Actividad , Tirosina/química , Tirosina/genética , Tirosina/metabolismo
6.
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
7.
Chembiochem ; 17(5): 415-20, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26715336

RESUMEN

Isotope-edited two-dimensional Fourier transform infrared spectroscopy (2 D FTIR) can potentially provide a unique probe of protein structure and dynamics. However, general methods for the site-specific incorporation of stable (13) C=(18) O labels into the polypeptide backbone of the protein molecule have not yet been established. Here we describe, as a prototype for the incorporation of specific arrays of isotope labels, the total chemical synthesis-via a key ester insulin intermediate-of 97 % enriched [(1-(13) C=(18) O)Phe(B24) ] human insulin: stable-isotope labeled at a single backbone amide carbonyl. The amino acid sequence as well as the positions of the disulfide bonds and the correctly folded structure were unambiguously confirmed by the X-ray crystal structure of the synthetic protein molecule. In vitro assays of the isotope labeled [(1-(13) C=(18) O)Phe(B24) ] human insulin showed that it had full insulin receptor binding activity. Linear and 2 D IR spectra revealed a distinct red-shifted amide I carbonyl band peak at 1595 cm(-1) resulting from the (1-(13) C=(18) O)Phe(B24) backbone label. This work illustrates the utility of chemical synthesis to enable the application of advanced physical methods for the elucidation of the molecular basis of protein function.


Asunto(s)
Isótopos de Carbono/química , Insulina/química , Isótopos de Oxígeno/química , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular
8.
J Biol Chem ; 289(34): 23683-92, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002580

RESUMEN

Misfolding of proinsulin variants in the pancreatic ß-cell, a monogenic cause of permanent neonatal-onset diabetes mellitus, provides a model for a disease of protein toxicity. A hot spot for such clinical mutations is found at position B8, conserved as glycine within the vertebrate insulin superfamily. We set out to investigate the molecular basis of the aberrant properties of a proinsulin clinical mutant in which residue Gly(B8) is replaced by Ser(B8). Modular total chemical synthesis was used to prepare the wild-type [Gly(B8)]proinsulin molecule and three analogs: [D-Ala(B8)]proinsulin, [L-Ala(B8)]proinsulin, and the clinical mutant [L-Ser(B8)]proinsulin. The protein diastereomer [D-Ala(B8)]proinsulin produced higher folding yields at all pH values compared with the wild-type proinsulin and the other two analogs, but showed only very weak binding to the insulin receptor. The clinical mutant [L-Ser(B8)]proinsulin impaired folding at pH 7.5 even in the presence of protein-disulfide isomerase. Surprisingly, although [L-Ser(B8)]proinsulin did not fold well under the physiological conditions investigated, once folded the [L-Ser(B8)]proinsulin protein molecule bound to the insulin receptor more effectively than wild-type proinsulin. Such paradoxical gain of function (not pertinent in vivo due to impaired secretion of the mutant insulin) presumably reflects induced fit in the native mechanism of hormone-receptor engagement. This work provides insight into the molecular mechanism of a clinical mutation in the insulin gene associated with diabetes mellitus. These results dramatically illustrate the power of total protein synthesis, as enabled by modern chemical ligation methods, for the investigation of protein folding and misfolding.


Asunto(s)
Alanina/química , Diabetes Mellitus/metabolismo , Enfermedades del Recién Nacido/metabolismo , Proinsulina/síntesis química , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Humanos , Recién Nacido , Proinsulina/química , Espectrometría de Masa por Ionización de Electrospray , Estereoisomerismo
9.
J Biol Chem ; 289(34): 23367-81, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-24993826

RESUMEN

Insulin provides a model for the therapeutic application of protein engineering. A paradigm in molecular pharmacology was defined by design of rapid-acting insulin analogs for the prandial control of glycemia. Such analogs, a cornerstone of current diabetes regimens, exhibit accelerated subcutaneous absorption due to more rapid disassembly of oligomeric species relative to wild-type insulin. This strategy is limited by a molecular trade-off between accelerated disassembly and enhanced susceptibility to degradation. Here, we demonstrate that this trade-off may be circumvented by nonstandard mutagenesis. Our studies employed Lys(B28), Pro(B29)-insulin ("lispro") as a model prandial analog that is less thermodynamically stable and more susceptible to fibrillation than is wild-type insulin. We have discovered that substitution of an invariant tyrosine adjoining the engineered sites in lispro (Tyr(B26)) by 3-iodo-Tyr (i) augments its thermodynamic stability (ΔΔGu 0.5 ± 0.2 kcal/mol), (ii) delays onset of fibrillation (lag time on gentle agitation at 37 °C was prolonged by 4-fold), (iii) enhances affinity for the insulin receptor (1.5 ± 0.1-fold), and (iv) preserves biological activity in a rat model of diabetes mellitus. (1)H NMR studies suggest that the bulky iodo-substituent packs within a nonpolar interchain crevice. Remarkably, the 3-iodo-Tyr(B26) modification stabilizes an oligomeric form of insulin pertinent to pharmaceutical formulation (the R6 zinc hexamer) but preserves rapid disassembly of the oligomeric form pertinent to subcutaneous absorption (T6 hexamer). By exploiting this allosteric switch, 3-iodo-Tyr(B26)-lispro thus illustrates how a nonstandard amino acid substitution can mitigate the unfavorable biophysical properties of an engineered protein while retaining its advantages.


Asunto(s)
Insulina/análogos & derivados , Mutagénesis , Animales , Fenómenos Biofísicos , Dicroismo Circular , Insulina/química , Insulina/genética , Insulina/farmacocinética , Masculino , Resonancia Magnética Nuclear Biomolecular , Ingeniería de Proteínas , Ratas , Ratas Endogámicas Lew , Receptor de Insulina/metabolismo , Espectrofotometría Ultravioleta
10.
J Biol Chem ; 289(50): 34709-27, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25305014

RESUMEN

Crystallographic studies of insulin bound to fragments of the insulin receptor have recently defined the topography of the primary hormone-receptor interface. Here, we have investigated the role of Phe(B24), an invariant aromatic anchor at this interface and site of a human mutation causing diabetes mellitus. An extensive set of B24 substitutions has been constructed and tested for effects on receptor binding. Although aromaticity has long been considered a key requirement at this position, Met(B24) was found to confer essentially native affinity and bioactivity. Molecular modeling suggests that this linear side chain can serve as an alternative hydrophobic anchor at the hormone-receptor interface. These findings motivated further substitution of Phe(B24) by cyclohexanylalanine (Cha), which contains a nonplanar aliphatic ring. Contrary to expectations, [Cha(B24)]insulin likewise exhibited high activity. Furthermore, its resistance to fibrillation and the rapid rate of hexamer disassembly, properties of potential therapeutic advantage, were enhanced. The crystal structure of the Cha(B24) analog, determined as an R6 zinc-stabilized hexamer at a resolution of 1.5 Å, closely resembles that of wild-type insulin. The nonplanar aliphatic ring exhibits two chair conformations with partial occupancies, each recapitulating the role of Phe(B24) at the dimer interface. Together, these studies have defined structural requirements of an anchor residue within the B24-binding pocket of the insulin receptor; similar molecular principles are likely to pertain to insulin-related growth factors. Our results highlight in particular the utility of nonaromatic side chains as probes of the B24 pocket and suggest that the nonstandard Cha side chain may have therapeutic utility.


Asunto(s)
Diseño de Fármacos , Insulina/química , Insulina/metabolismo , Fenilalanina , Receptor de Insulina/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Insulina/genética , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Pliegue de Proteína , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/química
11.
Proc Natl Acad Sci U S A ; 109(28): 11166-71, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22736795

RESUMEN

The primary hormone-binding surface of the insulin receptor spans one face of the N-terminal ß-helix of the α-subunit (the L1 domain) and an α-helix in its C-terminal segment (αCT). Crystallographic analysis of the free ectodomain has defined a contiguous dimer-related motif in which the αCT α-helix packs against L1 ß-strands 2 and 3. To relate structure to function, we exploited expanded genetic-code technology to insert photo-activatable probes at key sites in L1 and αCT. The pattern of αCT-mediated photo-cross-linking within the free and bound receptor is in accord with the crystal structure and prior mutagenesis. Surprisingly, L1 photo-probes in ß-strands 2 and 3, predicted to be shielded by αCT, efficiently cross-link to insulin. Furthermore, anomalous mutations were identified on neighboring surfaces of αCT and insulin that impair hormone-dependent activation of the intracellular receptor tyrosine kinase (contained within the transmembrane ß-subunit) disproportionately to their effects on insulin binding. Taken together, these results suggest that αCT, in addition to its hormone-recognition role, provides a signaling element in the mechanism of receptor activation.


Asunto(s)
Proteínas Tirosina Quinasas/química , Receptor de Insulina/metabolismo , Aminoacil-ARNt Sintetasas/metabolismo , Bacillus/metabolismo , Sitios de Unión , Membrana Celular/metabolismo , Reactivos de Enlaces Cruzados/farmacología , Cristalografía por Rayos X/métodos , Escherichia coli/metabolismo , Hormonas/metabolismo , Modelos Biológicos , Conformación Molecular , Mutagénesis , Mutación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transducción de Señal , Tirosina/química
12.
Eye (Lond) ; 38(5): 1005-1011, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37980397

RESUMEN

INTRODUCTION: Glauc-Strat-Fast is a clinical tool recommended by The Royal College of Ophthalmologists to classify glaucoma patients into strata of risk for significant future sight loss and an estimate of resource requirement. The aim of this study was to map the movement of glaucoma patients across stratification boundaries on Glauc-Strat-Fast during the COVID-19 pandemic. SUBJECTS AND METHODS: Glauc-Strat-Fast was applied to a consecutive sample of 100 primary open angle glaucoma patients in a backlog at Worcestershire Acute Hospitals NHS Trust. Stratification outcomes were compared between clinic visits prior to the COVID-19 pandemic versus the follow-up visit. Patients were stratified twice separately based on their worse eye (i.e., most affected) and better eye (i.e., least affected) according to Glauc-Strat-Fast. RESULTS: Amount of slippage (difference between target follow-up and actual follow-up) ranged from 2 to 32 months. There was a statistically significant average reduction in visual field mean deviation for better and worse eyes between visits (p = <0.001). At follow-up, no worse eyes were classified as being low risk (green), while 96 were classified as high risk (red). For better eyes, elevation of risk into the highest strata of Glauc-Strat-Fast observed a three-fold increase in patients (19 versus 56) between visits. DISCUSSION: This retrospective real-world analysis highlights patients' movement into the highest strata on the Glauc-Strat-Fast tool and demonstrates a significant deterioration in visual outcomes during a period of extensive appointment slippage. The findings demonstrate the utility of Glauc-Strat-Fast as a tool for improved patient management.


Asunto(s)
COVID-19 , Glaucoma de Ángulo Abierto , Glaucoma , Humanos , Estudios Retrospectivos , Pandemias , Presión Intraocular , Trastornos de la Visión , Ceguera , Medición de Riesgo
13.
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
15.
J Biol Chem ; 285(40): 30989-1001, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20663888

RESUMEN

Protein sequences encode both structure and foldability. Whereas the interrelationship of sequence and structure has been extensively investigated, the origins of folding efficiency are enigmatic. We demonstrate that the folding of proinsulin requires a flexible N-terminal hydrophobic residue that is dispensable for the structure, activity, and stability of the mature hormone. This residue (Phe(B1) in placental mammals) is variably positioned within crystal structures and exhibits (1)H NMR motional narrowing in solution. Despite such flexibility, its deletion impaired insulin chain combination and led in cell culture to formation of non-native disulfide isomers with impaired secretion of the variant proinsulin. Cellular folding and secretion were maintained by hydrophobic substitutions at B1 but markedly perturbed by polar or charged side chains. We propose that, during folding, a hydrophobic side chain at B1 anchors transient long-range interactions by a flexible N-terminal arm (residues B1-B8) to mediate kinetic or thermodynamic partitioning among disulfide intermediates. Evidence for the overall contribution of the arm to folding was obtained by alanine scanning mutagenesis. Together, our findings demonstrate that efficient folding of proinsulin requires N-terminal sequences that are dispensable in the native state. Such arm-dependent folding can be abrogated by mutations associated with ß-cell dysfunction and neonatal diabetes mellitus.


Asunto(s)
Modelos Químicos , Proinsulina/química , Pliegue de Proteína , Sustitución de Aminoácidos , Cristalografía por Rayos X , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Mutación Missense , Resonancia Magnética Nuclear Biomolecular , Proinsulina/genética , Proinsulina/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína
16.
J Biol Chem ; 285(7): 5040-55, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19959476

RESUMEN

Proinsulin exhibits a single structure, whereas insulin-like growth factors refold as two disulfide isomers in equilibrium. Native insulin-related growth factor (IGF)-I has canonical cystines (A6-A11, A7-B7, and A20-B19) maintained by IGF-binding proteins; IGF-swap has alternative pairing (A7-A11, A6-B7, and A20-B19) and impaired activity. Studies of mini-domain models suggest that residue B5 (His in insulin and Thr in IGFs) governs the ambiguity or uniqueness of disulfide pairing. Residue B5, a site of mutation in proinsulin causing neonatal diabetes, is thus of broad biophysical interest. Here, we characterize reciprocal B5 substitutions in the two proteins. In insulin, His(B5) --> Thr markedly destabilizes the hormone (DeltaDeltaG(u) 2.0 +/- 0.2 kcal/mol), impairs chain combination, and blocks cellular secretion of proinsulin. The reciprocal IGF-I substitution Thr(B5) --> His (residue 4) specifies a unique structure with native (1)H NMR signature. Chemical shifts and nuclear Overhauser effects are similar to those of native IGF-I. Whereas wild-type IGF-I undergoes thiol-catalyzed disulfide exchange to yield IGF-swap, His(B5)-IGF-I retains canonical pairing. Chemical denaturation studies indicate that His(B5) does not significantly enhance thermodynamic stability (DeltaDeltaG(u) 0.2 +/- 0.2 kcal/mol), implying that the substitution favors canonical pairing by destabilizing competing folds. Whereas the activity of Thr(B5)-insulin is decreased 5-fold, His(B5)-IGF-I exhibits 2-fold increased affinity for the IGF receptor and augmented post-receptor signaling. We propose that conservation of Thr(B5) in IGF-I, rescued from structural ambiguity by IGF-binding proteins, reflects fine-tuning of signal transduction. In contrast, the conservation of His(B5) in insulin highlights its critical role in insulin biosynthesis.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/química , Factor I del Crecimiento Similar a la Insulina/farmacología , Insulina/química , Insulina/farmacología , Animales , Línea Celular , Dicroismo Circular , Disulfuros , Glicosilación , Humanos , Insulina/síntesis química , Factor I del Crecimiento Similar a la Insulina/síntesis química , Espectroscopía de Resonancia Magnética , Ratones , Fosforilación/efectos de los fármacos , Proinsulina/biosíntesis , Proinsulina/genética , Proinsulina/metabolismo , Pliegue de Proteína , Estabilidad Proteica , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
17.
J Biol Chem ; 285(16): 11755-9, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20181952

RESUMEN

Bottom-up control of supramolecular protein assembly can provide a therapeutic nanobiotechnology. We demonstrate that the pharmacological properties of insulin can be enhanced by design of "zinc staples" between hexamers. Paired (i, i+4) His substitutions were introduced at an alpha-helical surface. The crystal structure contains both classical axial zinc ions and novel zinc ions at hexamer-hexamer interfaces. Although soluble at pH 4, the combined electrostatic effects of the substitutions and bridging zinc ions cause isoelectric precipitation at neutral pH. Following subcutaneous injection in a diabetic rat, the analog effected glycemic control with a time course similar to that of long acting formulation Lantus. Relative to Lantus, however, the analog discriminates at least 30-fold more stringently between the insulin receptor and mitogenic insulin-like growth factor receptor. Because aberrant mitogenic signaling may be associated with elevated cancer risk, such enhanced specificity may improve safety. Zinc stapling provides a general strategy to modify the pharmacokinetic and biological properties of a subcutaneous protein depot.


Asunto(s)
Insulina/análogos & derivados , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , Preparaciones de Acción Retardada , Diseño de Fármacos , Humanos , Insulina/administración & dosificación , Insulina/síntesis química , Insulina/química , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Ingeniería de Proteínas/métodos , Estructura Secundaria de Proteína , Ratas , Electricidad Estática , Zinc/química
18.
J Biol Chem ; 285(14): 10806-21, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20106984

RESUMEN

Insulin fibrillation provides a model for a broad class of amyloidogenic diseases. Conformational distortion of the native monomer leads to aggregation-coupled misfolding. Whereas beta-cells are protected from proteotoxicity by hexamer assembly, fibrillation limits the storage and use of insulin at elevated temperatures. Here, we have investigated conformational distortions of an engineered insulin monomer in relation to the structure of an insulin fibril. Anomalous (13)C NMR chemical shifts and rapid (15)N-detected (1)H-(2)H amide-proton exchange were observed in one of the three classical alpha-helices (residues A1-A8) of the hormone, suggesting a conformational equilibrium between locally folded and unfolded A-chain segments. Whereas hexamer assembly resolves these anomalies in accordance with its protective role, solid-state (13)C NMR studies suggest that the A-chain segment participates in a fibril-specific beta-sheet. Accordingly, we investigated whether helicogenic substitutions in the A1-A8 segment might delay fibrillation. Simultaneous substitution of three beta-branched residues (Ile(A2) --> Leu, Val(A3) --> Leu, and Thr(A8) --> His) yielded an analog with reduced thermodynamic stability but marked resistance to fibrillation. Whereas amide-proton exchange in the A1-A8 segment remained rapid, (13)Calpha chemical shifts exhibited a more helical pattern. This analog is essentially without activity, however, as Ile(A2) and Val(A3) define conserved receptor contacts. To obtain active analogs, substitutions were restricted to A8. These analogs exhibit high receptor-binding affinity; representative potency in a rodent model of diabetes mellitus was similar to wild-type insulin. Although (13)Calpha chemical shifts remain anomalous, significant protection from fibrillation is retained. Together, our studies define an "Achilles' heel" in a globular protein whose repair may enhance the stability of pharmaceutical formulations and broaden their therapeutic deployment in the developing world.


Asunto(s)
Amiloide/química , Diabetes Mellitus Experimental/metabolismo , Diseño de Fármacos , Insulina/química , Insulina/farmacología , Amiloide/metabolismo , Animales , Antibióticos Antineoplásicos/toxicidad , Cristalografía por Rayos X , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/tratamiento farmacológico , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Estructura Secundaria de Proteína , Ratas , Ratas Endogámicas Lew , Receptor IGF Tipo 1/metabolismo , Estreptozocina/toxicidad
19.
J Biol Chem ; 284(46): 32178-87, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19773552

RESUMEN

Insulin binds with high affinity to the insulin receptor (IR) and with low affinity to the type 1 insulin-like growth factor (IGF) receptor (IGFR). Such cross-binding, which reflects homologies within the insulin-IGF signaling system, is of clinical interest in relation to the association between hyperinsulinemia and colorectal cancer. Here, we employ nonstandard mutagenesis to design an insulin analog with enhanced affinity for the IR but reduced affinity for the IGFR. Unnatural amino acids were introduced by chemical synthesis at the N- and C-capping positions of a recognition alpha-helix (residues A1 and A8). These sites adjoin the hormone-receptor interface as indicated by photocross-linking studies. Specificity is enhanced more than 3-fold on the following: (i) substitution of Gly(A1) by D-Ala or D-Leu, and (ii) substitution of Thr(A8) by diaminobutyric acid (Dab). The crystal structure of [D-Ala(A1),Dab(A8)]insulin, as determined within a T(6) zinc hexamer to a resolution of 1.35 A, is essentially identical to that of human insulin. The nonstandard side chains project into solvent at the edge of a conserved receptor-binding surface shared by insulin and IGF-I. Our results demonstrate that modifications at this edge discriminate between IR and IGFR. Because hyperinsulinemia is typically characterized by a 3-fold increase in integrated postprandial insulin concentrations, we envisage that such insulin analogs may facilitate studies of the initiation and progression of cancer in animal models. Future development of clinical analogs lacking significant IGFR cross-binding may enhance the safety of insulin replacement therapy in patients with type 2 diabetes mellitus at increased risk of colorectal cancer.


Asunto(s)
Diseño de Fármacos , Insulina/análogos & derivados , Insulina/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Sitios de Unión , Dicroismo Circular , Reactivos de Enlaces Cruzados/farmacología , Cristalografía por Rayos X , Humanos , Insulina/química , Factor I del Crecimiento Similar a la Insulina/farmacología , Modelos Químicos , Modelos Moleculares , Mutación/genética , Pliegue de Proteína , Estructura Terciaria de Proteína , Receptor IGF Tipo 1/genética , Receptor de Insulina/química , Receptor de Insulina/genética , Relación Estructura-Actividad
20.
Biochemistry ; 48(47): 11283-95, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19863112

RESUMEN

The insulin from the Atlantic hagfish (Myxine glutinosa) has been one of the most studied insulins from both a structural and a biological viewpoint; however, some aspects of its biology remain controversial, and there has been no satisfying structural explanation for its low biological potency. We have re-examined the receptor binding kinetics, as well as the metabolic and mitogenic properties, of this phylogenetically ancient insulin, as well as that from another extant representative of the ancient chordates, the river lamprey (Lampetra fluviatilis). Both insulins share unusual binding kinetics and biological properties with insulin analogues that have single mutations at residues that contribute to the hexamerization surface. We propose and demonstrate by reciprocal amino acid substitutions between hagfish and human insulins that the reduced biological activity of hagfish insulin results from unfavorable substitutions, namely, A10 (Ile to Arg), B4 (Glu to Gly), B13 (Glu to Asn), and B21 (Glu to Val). We likewise suggest that the altered biological activity of lamprey insulin may reflect substitutions at A10 (Ile to Lys), B4 (Glu to Thr), and B17 (Leu to Val). The substitution of Asp at residue B10 in hagfish insulin and of His at residue A8 in both hagfish and lamprey insulins may help compensate for unfavorable changes in other regions of the molecules. The data support the concept that the set of unusual properties of insulins bearing certain mutations in the hexamerization surface may reflect those of the insulins evolutionarily closer to the ancestral insulin gene product.


Asunto(s)
Anguila Babosa , Insulina/química , Insulina/metabolismo , Lampreas , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión , Anguila Babosa/genética , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/metabolismo , Insulina/genética , Cinética , Lampreas/genética , Mitógenos , Modelos Moleculares , Mutación , Filogenia , Receptor de Insulina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA