Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 583
Filtrar
1.
Growth Horm IGF Res ; 77: 101607, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39033666

RESUMEN

Type 2 diabetes is characterised by the disruption of insulin and insulin-like growth factor (IGF) signalling. The key hubs of these signalling cascades - the Insulin receptor (IR) and Insulin-like growth factor 1 receptor (IGF1R) - are known to form functional IR-IGF1R hybrid receptors which are insulin resistant. However, the mechanisms underpinning IR-IGF1R hybrid formation are not fully understood, hindering the ability to modulate this for future therapies targeting this receptor. To pinpoint suitable sites for intervention, computational hotspot prediction was utilised to identify promising epitopes for targeting with point mutagenesis. Specific IGF1R point mutations F450A, R391A and D555A show reduced affinity of the hybrid receptor in a BRET based donor-saturation assay, confirming hybrid formation could be modulated at this interface. These data provide the basis for rational design of more effective hybrid receptor modulators, supporting the prospect of identifying a small molecule that specifically interacts with this target.


Asunto(s)
Mutagénesis Sitio-Dirigida , Receptor IGF Tipo 1 , Receptor de Insulina , Receptor de Insulina/genética , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Humanos , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/metabolismo , Multimerización de Proteína , Péptidos Similares a la Insulina , Antígenos CD
2.
J Chem Inf Model ; 64(14): 5657-5670, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38963805

RESUMEN

Insulin Wakayama is a clinical insulin variant where a conserved valine at the third residue on insulin's A chain (ValA3) is replaced with a leucine (LeuA3), weakening insulin receptor (IR) binding by 140-500-fold. This severe impact on binding from a subtle modification has posed an intriguing problem for decades. Although experimental investigations of natural and unnatural A3 mutations have highlighted the sensitivity of insulin-IR binding at this site, atomistic explanations of these binding trends have remained elusive. We investigate this problem computationally using λ-dynamics free energy calculations to model structural changes in response to perturbations of the ValA3 side chain and to calculate associated relative changes in binding free energy (ΔΔGbind). The Wakayama LeuA3 mutation and seven other A3 substitutions were studied in this work. The calculated ΔΔGbind results showed high agreement compared to experimental binding potencies with a Pearson correlation of 0.88 and a mean unsigned error of 0.68 kcal/mol. Extensive structural analyses of λ-dynamics trajectories revealed that critical interactions were disrupted between insulin and the insulin receptor as a result of the A3 mutations. This investigation also quantifies the effect that adding an A3 Cδ atom or losing an A3 Cγ atom has on insulin's binding affinity to the IR. Thus, λ-dynamics was able to successfully model the effects of mutations to insulin's A3 side chain on its protein-protein interactions with the IR and shed new light on a decades-old mystery: the exquisite sensitivity of hormone-receptor binding to a subtle modification of an invariant insulin residue.


Asunto(s)
Insulina , Simulación de Dinámica Molecular , Unión Proteica , Receptor de Insulina , Termodinámica , Receptor de Insulina/metabolismo , Receptor de Insulina/química , Receptor de Insulina/genética , Insulina/metabolismo , Insulina/química , Mutación , Humanos , Conformación Proteica
3.
ChemMedChem ; 19(11): e202400145, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38445366

RESUMEN

The binding process of insulin to its transmembrane receptor entails a sophisticated interplay between two proteins, each possessing two binding sites. Given the difficulties associated with the use of insulin in the treatment of diabetes, despite its remarkable efficacy, there is interest in smaller and more stable compounds than the native hormone that would effectively activate the receptor. Our study adopts a strategy focused on synthesizing extensive combinatorial libraries of bipodal compounds consisting of two distinct peptides linked to a molecular scaffold. These constructs, evaluated in a resin bead-bound format, were designed to assess their binding to the insulin receptor. Despite notable nonspecific binding, our approach successfully generated and tested millions of compounds. Rigorous evaluations via flow cytometry and specific antibodies revealed peptide sequences with specific interactions at either receptor binding Site 1 or 2. Notably, these sequences bear similarity to peptides discovered through phage display by other researchers. This convergence of chemical and biological methods underscores nature's beauty, revealing general principles in peptide binding to the insulin receptor. Overall, our study deepens the understanding of molecular interactions in ligand binding to the insulin receptor, highlighting the challenges of targeting large proteins with small synthetic peptides.


Asunto(s)
Técnicas Químicas Combinatorias , Receptor de Insulina , Receptor de Insulina/metabolismo , Receptor de Insulina/química , Humanos , Péptidos/química , Péptidos/metabolismo , Péptidos/síntesis química , Sitios de Unión , Biblioteca de Péptidos , Ligandos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/síntesis química , Estructura Molecular , Unión Proteica , Insulina/metabolismo , Insulina/química
4.
Proteins ; 92(8): 905-922, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38506327

RESUMEN

Understanding kinase-inhibitor selectivity continues to be a major objective in kinase drug discovery. We probe the molecular basis of selectivity of an allosteric inhibitor (MSC1609119A-1) of the insulin-like growth factor-I receptor kinase (IGF1RK), which has been shown to be ineffective for the homologous insulin receptor kinase (IRK). Specifically, we investigated the structural and energetic basis of the allosteric binding of this inhibitor to each kinase by combining molecular modeling, molecular dynamics (MD) simulations, and thermodynamic calculations. We predict the inhibitor conformation in the binding pocket of IRK and highlight that the charged residues in the histidine-arginine-aspartic acid (HRD) and aspartic acid-phenylalanine-glycine (DFG) motifs and the nonpolar residues in the binding pocket govern inhibitor interactions in the allosteric pocket of each kinase. We suggest that the conformational changes in the IGF1RK residues M1054 and M1079, movement of the ⍺C-helix, and the conformational stabilization of the DFG motif favor the selectivity of the inhibitor toward IGF1RK. Our thermodynamic calculations reveal that the observed selectivity can be rationalized through differences observed in the electrostatic interaction energy of the inhibitor in each inhibitor/kinase complex and the hydrogen bonding interactions of the inhibitor with the residue V1063 in IGF1RK that are not attained with the corresponding residue V1060 in IRK. Overall, our study provides a rationale for the molecular basis of recognition of this allosteric inhibitor by IGF1RK and IRK, which is potentially useful in developing novel inhibitors with improved affinity and selectivity.


Asunto(s)
Simulación de Dinámica Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas , Receptor IGF Tipo 1 , Termodinámica , Humanos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/metabolismo , Regulación Alostérica , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Sitio Alostérico , Sitios de Unión , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Receptor de Insulina/antagonistas & inhibidores , Enlace de Hidrógeno
5.
Open Biol ; 13(11): 230142, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37935358

RESUMEN

The insulin receptor (IR, with its isoforms IR-A and IR-B) and the insulin-like growth factor 1 receptor (IGF-1R) are related tyrosine kinase receptors. Recently, the portfolio of solved hormone-receptor structures has grown extensively thanks to advancements in cryo-electron microscopy. However, the dynamics of how these receptors transition between their inactive and active state are yet to be fully understood. The C-terminal part of the alpha subunit (αCT) of the receptors is indispensable for the formation of the hormone-binding site. We mutated the αCT residues Arg717 and His710 of IR-A and Arg704 and His697 of IGF-1R. We then measured the saturation binding curves of ligands on the mutated receptors and their ability to become activated. Mutations of Arg704 and His697 to Ala in IGF-1R decreased the binding of IGF-1. Moreover, the number of binding sites for IGF-1 on the His697 IGF-1R mutant was reduced to one-half, demonstrating the presence of two binding sites. Both mutations of Arg717 and His710 to Ala in IR-A inactivated the receptor. We have proved that Arg717 is important for the binding of insulin to its receptor, which suggests that Arg717 is a key residue for the transition to the active conformation.


Asunto(s)
Receptor IGF Tipo 1 , Receptor de Insulina , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/química , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ligandos , Microscopía por Crioelectrón , Insulina/metabolismo
6.
Nat Struct Mol Biol ; 30(5): 661-669, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37055497

RESUMEN

The insulin receptor (IR) family is a subfamily of receptor tyrosine kinases that controls metabolic homeostasis and cell growth. Distinct from IR and insulin-like growth factor 1 receptor, whose activation requires ligand binding, insulin receptor-related receptor (IRR)-the third member of the IR family-is activated by alkaline pH. However, the molecular mechanism underlying alkaline pH-induced IRR activation remains unclear. Here, we present cryo-EM structures of human IRR in both neutral pH inactive and alkaline pH active states. Combined with mutagenesis and cellular assays, we show that, upon pH increase, electrostatic repulsion of the pH-sensitive motifs of IRR disrupts its autoinhibited state and promotes a scissor-like rotation between two protomers, leading to a T-shaped active conformation. Together, our study reveals an unprecedented alkaline pH-dependent activation mechanism of IRR, opening up opportunities to understand the structure-function relationship of this important receptor.


Asunto(s)
Insulina , Receptor de Insulina , Humanos , Receptor de Insulina/química , Concentración de Iones de Hidrógeno , Insulina/química
7.
Annu Rev Biochem ; 92: 247-272, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37001136

RESUMEN

The insulin receptor (IR) is a type II receptor tyrosine kinase that plays essential roles in metabolism, growth, and proliferation. Dysregulation of IR signaling is linked to many human diseases, such as diabetes and cancers. The resolution revolution in cryo-electron microscopy has led to the determination of several structures of IR with different numbers of bound insulin molecules in recent years, which have tremendously improved our understanding of how IR is activated by insulin. Here, we review the insulin-induced activation mechanism of IR, including (a) the detailed binding modes and functions of insulin at site 1 and site 2 and (b) the insulin-induced structural transitions that are required for IR activation. We highlight several other key aspects of the activation and regulation of IR signaling and discuss the remaining gaps in our understanding of the IR activation mechanism and potential avenues of future research.


Asunto(s)
Insulina , Receptor de Insulina , Humanos , Receptor de Insulina/genética , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Microscopía por Crioelectrón , Insulina/química , Insulina/metabolismo , Transducción de Señal , Proteínas Tirosina Quinasas Receptoras/metabolismo , Fosforilación
8.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36835322

RESUMEN

Human InsR, IGF1R, and IRR receptor tyrosine kinases (RTK) of the insulin receptor subfamily play an important role in signaling pathways for a wide range of physiological processes and are directly associated with many pathologies, including neurodegenerative diseases. The disulfide-linked dimeric structure of these receptors is unique among RTKs. Sharing high sequence and structure homology, the receptors differ dramatically in their localization, expression, and functions. In this work, using high-resolution NMR spectroscopy supported by atomistic computer modeling, conformational variability of the transmembrane domains and their interactions with surrounding lipids were found to differ significantly between representatives of the subfamily. Therefore, we suggest that the heterogeneous and highly dynamic membrane environment should be taken into account in the observed diversity of the structural/dynamic organization and mechanisms of activation of InsR, IGF1R, and IRR receptors. This membrane-mediated control of receptor signaling offers an attractive prospect for the development of new targeted therapies for diseases associated with dysfunction of insulin subfamily receptors.


Asunto(s)
Desarrollo de Medicamentos , Receptor de Insulina , Humanos , Dominios Proteicos , Receptor de Insulina/química , Receptor de Insulina/fisiología , Transducción de Señal
9.
Proteins ; 91(1): 62-73, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35962629

RESUMEN

The insulin receptor (IR), the insulin-like growth factor-1 receptor (IGF1R), and the insulin/IGF1 hybrid receptors (hybR) are homologous transmembrane receptors. The peptide ligands, insulin and IGF1, exhibit significant structural homology and can bind to each receptor via site-1 and site-2 residues with distinct affinities. The variants of the Iridoviridae virus family show capability in expressing single-chain insulin/IGF1 like proteins, termed viral insulin-like peptides (VILPs), which can stimulate receptors from the insulin family. The sequences of VILPs lacking the central C-domain (dcVILPs) are known, but their structures in unbound and receptor-bound states have not been resolved to date. We report all-atom structural models of three dcVILPs (dcGIV, dcSGIV, and dcLCDV1) and their complexes with the receptors (µIR, µIGF1R, and µhybR), and probed the peptide/receptor interactions in each system using all-atom molecular dynamics (MD) simulations. Based on the nonbonded interaction energies computed between each residue of peptides (insulin and dcVILPs) and the receptors, we provide details on residues establishing significant interactions. The observed site-1 insulin/µIR interactions are consistent with previous experimental studies, and a residue-level comparison of interactions of peptides (insulin and dcVILPs) with the receptors revealed that, due to sequence differences, dcVILPs also establish some interactions distinct from those between insulin and IR. We also designed insulin analogs and report enhanced interactions between some analogs and the receptors.


Asunto(s)
Insulina , Virus , Insulina/metabolismo , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Receptor IGF Tipo 1/metabolismo , Modelos Estructurales , Simulación de Dinámica Molecular
10.
Biochem Biophys Res Commun ; 636(Pt 1): 121-124, 2022 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-36332473

RESUMEN

IGF1R plays an important role in regulating cellular metabolism and cell growth, and has been identified as an anti-cancer and diabetes drug target. Although research have been reported many crystal and cryo-EM structures of IGF1R, the mechanism of ligand binding remains controversial, mainly because the structure differences among its cryo-EM, crystal and homologous protein insulin receptor structures. Here, we further determined one new high-resolution symmetric cryo-EM structure of ligand-bound IGF1R and be the first to prove that the receptor could bind to two IGFI molecules by single particle cryo-electron microscopy. And the structure is very different from its homologous protein insulin receptor: the two ligands just exist at the binding site 2 with saturating ligand conditions. Then, our findings resolved the major dispute about the comformational changes of IGF1R, and proposed a new theory how IGF1R binds to its ligands. Meanwhile, these findings imply more attention may be needed to study the relationship between the special conformation and their corresponding physiological functions in future.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , Receptor IGF Tipo 1 , Humanos , Microscopía por Crioelectrón , Hormonas , Factor I del Crecimiento Similar a la Insulina/química , Ligandos , Dominios Proteicos , Receptor IGF Tipo 1/química , Receptor de Insulina/química
11.
Elife ; 112022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36413010

RESUMEN

The insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF1R) control metabolic homeostasis and cell growth and proliferation. The IR and IGF1R form similar disulfide bonds linked homodimers in the apo-state; however, their ligand binding properties and the structures in the active state differ substantially. It has been proposed that the disulfide-linked C-terminal segment of α-chain (αCTs) of the IR and IGF1R control the cooperativity of ligand binding and regulate the receptor activation. Nevertheless, the molecular basis for the roles of disulfide-linked αCTs in IR and IGF1R activation are still unclear. Here, we report the cryo-EM structures of full-length mouse IGF1R/IGF1 and IR/insulin complexes with modified αCTs that have increased flexibility. Unlike the Γ-shaped asymmetric IGF1R dimer with a single IGF1 bound, the IGF1R with the enhanced flexibility of αCTs can form a T-shaped symmetric dimer with two IGF1s bound. Meanwhile, the IR with non-covalently linked αCTs predominantly adopts an asymmetric conformation with four insulins bound, which is distinct from the T-shaped symmetric IR. Using cell-based experiments, we further showed that both IGF1R and IR with the modified αCTs cannot activate the downstream signaling potently. Collectively, our studies demonstrate that the certain structural rigidity of disulfide-linked αCTs is critical for optimal IR and IGF1R signaling activation.


Asunto(s)
Receptor IGF Tipo 1 , Receptor de Insulina , Animales , Ratones , Disulfuros/química , Ligandos , Receptor de Insulina/química , Receptor IGF Tipo 1/química , Microscopía por Crioelectrón , Multimerización de Proteína
12.
Structure ; 30(8): 1098-1108.e6, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35660159

RESUMEN

Monomers of the insulin receptor and type 1 insulin-like growth factor receptor (IGF-1R) can combine stochastically to form heterodimeric hybrid receptors. These hybrid receptors display ligand binding and signaling properties that differ from those of the homodimeric receptors. Here, we describe the cryoelectron microscopy structure of such a hybrid receptor in complex with insulin-like growth factor I (IGF-I). The structure (ca. 3.7 Å resolution) displays a single IGF-I ligand, bound in a similar fashion to that seen for IGFs in complex with IGF-1R. The IGF-I ligand engages the first leucine-rich-repeat domain and cysteine-rich region of the IGF-1R monomer (rather than those of the insulin receptor monomer), consistent with the determinants for IGF binding residing in the IGF-1R cysteine-rich region. The structure broadens our understanding of this receptor family and assists in delineating the key structural motifs involved in binding their respective ligands.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , Receptor de Insulina , Microscopía por Crioelectrón , Cisteína , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ligandos , Receptor IGF Tipo 1/química , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Receptores de Somatomedina
13.
Nat Struct Mol Biol ; 29(4): 357-368, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35361965

RESUMEN

Insulin receptor (IR) signaling controls multiple facets of animal physiology. Maximally four insulins bind to IR at two distinct sites, termed site-1 and site-2. However, the precise functional roles of each binding event during IR activation remain unresolved. Here, we showed that IR incompletely saturated with insulin predominantly forms an asymmetric conformation and exhibits partial activation. IR with one insulin bound adopts a Γ-shaped conformation. IR with two insulins bound assumes a Ƭ-shaped conformation. One insulin binds at site-1 and another simultaneously contacts both site-1 and site-2 in the Ƭ-shaped IR dimer. We further show that concurrent binding of four insulins to sites-1 and -2 prevents the formation of asymmetric IR and promotes the T-shaped symmetric, fully active state. Collectively, our results demonstrate how the synergistic binding of multiple insulins promotes optimal IR activation.


Asunto(s)
Insulinas , Receptor de Insulina , Animales , Insulina/química , Receptor de Insulina/química , Transducción de Señal
14.
J Mol Biol ; 434(5): 167458, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35074483

RESUMEN

Insulin regulates glucose homeostasis via binding and activation of the insulin receptor dimer at two distinct pairs of binding sites 1 and 2. Here, we present cryo-EM studies of full-length human insulin receptor (hIR) in an active state obtained at non-saturating, physiologically relevant insulin conditions. Insulin binds asymmetrically to the receptor under these conditions, occupying up to three of the four possible binding sites. Deletion analysis of the receptor together with site specific peptides and insulin analogs used in binding studies show that both sites 1 and 2 are required for high insulin affinity. We identify a homotypic interaction of the fibronectin type III domain (FnIII-3) of IR resulting in tight interaction of membrane proximal domains of the active, asymmetric receptor dimer. Our results show how insulin binding at two distinct types of sites disrupts the autoinhibited apo-IR dimer and stabilizes the active dimer. We propose an insulin binding and activation mechanism, which is sequential, exhibits negative cooperativity, and is based on asymmetry at physiological insulin concentrations with one to three insulin molecules activating IR.


Asunto(s)
Antígenos CD , Insulina , Receptor de Insulina , Antígenos CD/química , Antígenos CD/metabolismo , Sitios de Unión , Microscopía por Crioelectrón , Humanos , Insulina/metabolismo , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Transducción de Señal
15.
Mol Med Rep ; 25(2)2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35014690

RESUMEN

Optogenetics combined with protein engineering based on natural light­sensitive dimerizing proteins has evolved as a powerful strategy to study cellular functions. The present study focused on tropomyosin kinase receptors (Trks) that have been engineered to be light­sensitive. Trk belongs to the superfamily of receptor tyrosine kinases (RTKs), which are single­pass transmembrane receptors that are activated by natural ligands and serve crucial roles in cellular growth, differentiation, metabolism and motility. However, functional variations exist among receptors fused with light­sensitive proteins. The present study proposed a signal transduction model for light­induced receptor activation. This model is based on analysis of previous light­induced Trk receptors reported to date and comparisons to the activation mechanism of natural receptors. In this model, quantitative differences on the dimerization induced from either top­to­bottom or bottom­to­up may lead to the varying amplitude of intracellular signals. We hypothesize that the top­to­bottom propagation is more favourable for activation and yields better results compared with the bottom­to­top direction. The careful delineation of the dimerization mechanisms fine­tuning activation will guide future design for an optimum cellular output with the precision of light.


Asunto(s)
Factores de Crecimiento Nervioso/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Dimerización , Humanos , Luz , Fototransducción , Modelos Biológicos , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Receptor trkA/química , Receptor trkA/metabolismo
16.
Proteins ; 90(3): 680-690, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34661928

RESUMEN

The venomous insulin-like peptides released by certain cone snails stimulate hypoglycemic shock to immobilize fish and catch the prey. Compared to human insulin (hIns), the cone snail insulins (Con-Ins) are typically monomeric and shorter in sequence, yet they exhibit moderate hIns-like biological activity. We have modeled six variants of Con-Ins (G3, K1, K2, T1A, T1B, and T2) and carried out explicit-solvent molecular dynamics (MD) simulations of eight types of insulins, two with known structures (hIns and Con-Ins-G1) and six Con-Ins with modeled structures, to characterize key residues of each insulin that interact with the truncated human insulin receptor (µIR). We show that each insulin/µIR complex is stable during explicit-solvent MD simulations and hIns interactions indicate the highest affinity for the "site 1" of IR. The residue contact maps reveal that each insulin preferably interacts with the αCT peptide than the L1 domain of IR. Through analysis of the average nonbonded interaction energy contribution of every residue of each insulin for the µIR, we probe the residues establishing favorable interactions with the receptor. We compared the interaction energy of each residue of every Con-Ins to the µIR and observed that γ-carboxylated glutamate (Gla), His, Thr, Tyr, Tyr/His, and Asn in Con-Ins are favorable substitutions for GluA4, AsnA21, ValB12, LeuB15, GlyB20, and ArgB22 in hIns, respectively. The identified insulin analogs, although lacking the last eight residues of the B-chain of hIns, bind strongly to µIR. Our findings are potentially useful in designing potent fast-acting therapeutic insulin.


Asunto(s)
Antígenos CD/química , Hipoglucemia/etiología , Insulinas/química , Receptor de Insulina/química , Secuencia de Aminoácidos , Animales , Humanos , Simulación de Dinámica Molecular , Venenos de Moluscos/química , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
17.
Life Sci ; 285: 120020, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34624320

RESUMEN

AIMS: The bile acid (BA), tauroursodeoxycholic acid (TUDCA) regulates glucose homeostasis; however, it is not clear whether its effects on insulin signaling are due to its direct interaction with the insulin receptor (IR) or through activation of the G-coupled BA receptor, TGR5. We, herein, investigated whether the actions of TUDCA on glucose homeostasis occur via IR or TGR5 activation. MAIN METHODS: Glucose homeostasis was evaluated in high-fat diet (HFD)-obese or control (CTL) mice, after 30 days or one intraperitoneal (ip) injection of 300 mg/kg TUDCA, respectively. Molecular docking was performed to investigate the potential binding of TUDCA on the IR and TGR5. KEY FINDINGS: After 30 days of TUDCA treatment, HFD mice exhibited improvements in glucose tolerance and insulin sensitivity, which were abolished when these rodents received the IR antagonist, S961. Molecular docking experiments showed that TUDCA demonstrates high binding affinity for TGR5 and IR and strongly interacts with the insulin binding sites 1 and 2 of the IR. Consistent with this potential agonist activity of TUDCA on IR, CTL mice displayed increased hepatic phosphorylation of AKT after an ip injection of TUDCA. This effect was not associated with altered glycemia in CTL mice and was dependent on IR activation, as S961 prevented hepatic AKT activation by TUDCA. Furthermore, TUDCA activated the hepatic protein kinase A (PKA) and cAMP response element-binding protein (CREB) pathway in CTL mice, even after the administration of S961. SIGNIFICANCE: We provide novel evidence that TUDCA may be an agonist of the IR, in turn activating AKT and contributing, at least in part, to its beneficial effects upon glucose homeostasis.


Asunto(s)
Glucosa/metabolismo , Receptor de Insulina/agonistas , Ácido Tauroquenodesoxicólico/farmacología , Animales , Sitios de Unión , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis/efectos de los fármacos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Obesidad/metabolismo , Unión Proteica , Receptor de Insulina/química , Receptores Acoplados a Proteínas G/metabolismo , Ácido Tauroquenodesoxicólico/administración & dosificación
18.
Biomed Res Int ; 2021: 5561129, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34589547

RESUMEN

Diabetes mellitus termed as metabolic disorder is a collection of interlinked diseases and mainly body's inability to manage glucose level which leads to cardiovascular diseases, renal failure, neurological disorders, and many others. The drugs contemporarily used for diabetes have many inevitable side effects, and many of them have become less responsive to this multifactorial disorder. Momordica charantia commonly known as bitter gourd has many bioactive compounds with antidiabetic properties. The current study was designed to use computational methods to discover the best antidiabetic peptides devised from hypoglycemic polypeptide-P of M. charantia. The binding affinity and interaction patterns of peptides were evaluated against four receptor proteins (i.e., as agonists of insulin receptor and inhibitors of sodium-glucose cotransporter 1, dipeptidyl peptidase-IV, and glucose transporter 2) using molecular docking approach. A total of thirty-seven peptides were docked against these receptors. Out of which, top five peptides against each receptor were shortlisted based on their S-scores and binding affinities. Finally, the eight best ligands (i.e., LIVA, TSEP, EKAI, LKHA, EALF, VAEK, DFGAS, and EPGGGG) were selected as these ligands strictly followed Lipinski's rule of five and exhibited good ADMET profiling. One peptide EPGGGG showed activity towards insulin and SGLT1 receptor proteins. The top complex for both these targets was subjected to 50 ns of molecular dynamics simulations and MM-GBSA binding energy test that concluded both complexes as highly stable, and the intermolecular interactions were dominated by van der Waals and electrostatic energies. Overall, the selected ligands strongly fulfilled the drug-like evaluation criterion and proved to have good antidiabetic properties.


Asunto(s)
Hipoglucemiantes/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Momordica charantia/química , Péptidos/química , Secuencia de Aminoácidos , Dipeptidil Peptidasa 4/química , Humanos , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Péptidos/farmacocinética , Péptidos/farmacología , Receptor de Insulina/química , Termodinámica
19.
J Med Chem ; 64(19): 14848-14859, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34591477

RESUMEN

Insulin is a lifesaver for millions of diabetic patients. There is a need for new insulin analogues with more physiological profiles and analogues that will be thermally more stable than human insulin. Here, we describe the chemical engineering of 48 insulin analogues that were designed to have changed binding specificities toward isoforms A and B of the insulin receptor (IR-A and IR-B). We systematically modified insulin at the C-terminus of the B-chain, at the N-terminus of the A-chain, and at A14 and A18 positions. We discovered an insulin analogue that has Cα-carboxyamidated Glu at B31 and Ala at B29 and that has a more than 3-fold-enhanced binding specificity in favor of the "metabolic" IR-B isoform. The analogue is more resistant to the formation of insulin fibrils at 37 °C and is also more efficient in mice than human insulin. Therefore, [AlaB29,GluB31,amideB31]-insulin may be interesting for further clinical evaluation.


Asunto(s)
Antígenos CD/metabolismo , Insulina/análogos & derivados , Agregado de Proteínas , Isoformas de Proteínas/metabolismo , Receptor de Insulina/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos CD/química , Calorimetría/métodos , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Masculino , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica , Isoformas de Proteínas/química , Receptor de Insulina/química
20.
Peptides ; 145: 170640, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34450203

RESUMEN

The insulin/insulin-like growth factor signaling pathway is an evolutionary conserved pathway across metazoans and is required for development, metabolism and behavior. This pathway is associated with various human metabolic disorders and cancers. Thus, model organisms including Drosophila melanogaster and Caenorhabditis elegans provide excellent opportunities to examine the structure and function of this pathway and its influence on cellular metabolism and proliferation. In this review, we will provide an overview of human insulin and the human insulin signaling pathway and explore the recent discoveries in model organisms Drosophila melanogaster and Caenorhabditis elegans. Our review will provide information regarding the various insulin-like peptides in model organisms as well as the conserved functions of insulin signaling pathways. Further investigation of the insulin signaling pathway in model organisms could provide a promising opportunity to develop novel therapies for various metabolic disorders and insulin-mediated cancers.


Asunto(s)
Caenorhabditis elegans/metabolismo , Drosophila melanogaster/metabolismo , Insulina/metabolismo , Animales , Antígenos CD/química , Antígenos CD/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Humanos , Insulina/química , Insulina/genética , Neuropéptidos/genética , Neuropéptidos/metabolismo , Fosfohidrolasa PTEN/metabolismo , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA