Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Struct Mol Biol ; 30(10): 1495-1504, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37679563

RESUMEN

Anion exchanger 1 (AE1), a member of the solute carrier (SLC) family, is the primary bicarbonate transporter in erythrocytes, regulating pH levels and CO2 transport between lungs and tissues. Previous studies characterized its role in erythrocyte structure and provided insight into transport regulation. However, key questions remain regarding substrate binding and transport, mechanisms of drug inhibition and modulation by membrane components. Here we present seven cryo-EM structures in apo, bicarbonate-bound and inhibitor-bound states. These, combined with uptake and computational studies, reveal important molecular features of substrate recognition and transport, and illuminate sterol binding sites, to elucidate distinct inhibitory mechanisms of research chemicals and prescription drugs. We further probe the substrate binding site via structure-based ligand screening, identifying an AE1 inhibitor. Together, our findings provide insight into mechanisms of solute carrier transport and inhibition.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito , Bicarbonatos , Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Bicarbonatos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Sitios de Unión , Dominios Proteicos
2.
Thyroid ; 33(4): 492-500, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36762945

RESUMEN

Background: Autoimmune thyroid diseases (AITD) represent the most common autoimmune diseases. However, current therapies focus on relieving the symptoms instead of curing AITD, and new therapies to reverse the autoimmune attack on the thyroid are needed. HLA-DRß1-Arg74 is the key HLA class II allele that triggers AITD by presenting pathogenic thyroglobulin (Tg) peptides that activate thyroid self-reactive T cells. We hypothesized that blocking the presentation of Tg peptides to T cells within the HLA-DRß1-Arg74 peptide binding cleft could reverse the autoimmune response to the thyroid in AITD. Methods: The approach we used to block Tg peptide presentation within HLA-DRß1-Arg74 is to design retro-inverso D-amino acid (RID) peptides that have high affinity to the HLA-DRß1-Arg74 peptide binding pocket. Results: By using computational approaches and molecular dynamics simulations, we designed two RID peptides, RT-15 and VT-15, that blocked peptide binding to recombinant HLA-DRß1-Arg74 molecule, as well as T cell activation in vitro. Furthermore, RT-15 and VT-15 blocked in vivo T cell activation by thyroglobulin in humanized NOD-DR3 mice induced with experimental autoimmune thyroiditis. Conclusions: In summary, we discovered two RID peptides that block thyroglobulin peptide binding to HLA-DRß1-Arg74 and their presentation to T cells in AITD. These findings set the stage for a personalized medicine therapeutic approach for AITD patients who carry the DRß1-Arg74 allele. This antigen-specific therapeutic strategy can potentially be extended to other autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Enfermedad de Hashimoto , Tiroiditis Autoinmune , Ratones , Animales , Tiroiditis Autoinmune/tratamiento farmacológico , Tiroglobulina , Autoinmunidad , Presentación de Antígeno , Ratones Endogámicos NOD , Péptidos/química
3.
Front Immunol ; 12: 796552, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34987519

RESUMEN

Autoimmune polyglandular syndrome type 3 variant (APS3v) refers to an autoimmune condition in which both type 1 diabetes (T1D) and autoimmune thyroiditis (AITD) develop in the same individual. HLA-DR3 confers the strongest susceptibility to APS3v. Previously we reported a unique amino acid signature pocket that predisposes to APS3v. We found that this pocket is flexible and can trigger APS3v by presenting both thyroid (Tg.1571, TPO.758) and islet (GAD.492) peptides to induce autoimmune response. We hypothesized that blocking the specific APS3v-HLA-DR3 pocket from presenting thyroid/islet antigens can block the autoimmune response in APS3v. To test this hypothesis we performed a virtual screen of small molecules blocking APS3v-HLA-DR3, and identified 11 small molecules hits that were predicted to block APS3v-HLA-DR3. Using the baculovirus-produced recombinant APS3v-HLA-DR3 protein we tested the 11 small molecules in an in vitro binding assay. We validated 4 small molecule hits, S9, S5, S53 and S15, that could block the APS3v-HLA-DR3 pocket in vitro. We then developed a novel humanized APS3v mouse model induced by co-immunizing a peptide mix of Tg.1571, TPO.758 and GAD.492. The immunized mice developed strong T-cell and antibody responses to the thyroid/islet peptides, as well as mouse thyroglobulin. In addition, the mice showed significantly lower free T4 levels compared to controls. Using the APS3v mouse model, we showed that one of the 4 small molecules, Cepharanthine (S53), blocked T-cell activation by thyroid/islet peptides ex vivo and in vivo. These findings suggested Cepharanthine may have a therapeutic potential in APS3v patients carrying the specific APS3v-HLA-DR3 pocket.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Bencilisoquinolinas/uso terapéutico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Antígeno HLA-DR3/metabolismo , Islotes Pancreáticos/inmunología , Poliendocrinopatías Autoinmunes/tratamiento farmacológico , Linfocitos T/inmunología , Tiroiditis Autoinmune/tratamiento farmacológico , Animales , Presentación de Antígeno , Autoantígenos/inmunología , Sitios de Unión/genética , Células Cultivadas , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Glutamato Descarboxilasa/inmunología , Antígeno HLA-DR3/genética , Humanos , Inmunidad Humoral , Inmunización , Yoduro Peroxidasa/inmunología , Proteínas de Unión a Hierro/inmunología , Activación de Linfocitos , Ratones , Ratones SCID , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Poliendocrinopatías Autoinmunes/inmunología , Tiroglobulina/genética , Tiroglobulina/inmunología , Tiroiditis Autoinmune/inmunología
4.
J Autoimmun ; 115: 102543, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32951964

RESUMEN

Over the past four decades, the number of people with Type 1 Diabetes (T1D) has increased by 4% per year, making it an important public health challenge. Currently, no curative therapy exists for T1D and the only available treatment is insulin replacement. HLA-DQ8 has been shown to present antigenic islet peptides driving the activation of CD4+ T-cells in T1D patients. Specifically, the insulin peptide InsB:9-23 activates self-reactive CD4+ T-cells, causing pancreatic beta cell destruction. The aim of the current study was to identify retro-inverso-d-amino acid based peptides (RI-D-peptides) that can suppress T-cell activation by blocking the presentation of InsB:9-23 peptide within HLA-DQ8 pocket. We identified a RI-D-peptide (RI-EXT) that inhibited InsB:9-23 binding to recombinant HLA-DQ8 molecule, as well as its binding to DQ8 expressed on human B-cells. RI-EXT prevented T-cell activation in a cellular antigen presentation assay containing human DQ8 cells loaded with InsB:9-23 peptide and murine T-cells expressing a human T-cell receptor specific for the InsB:9-23-DQ8 complex. Moreover, RI-EXT blocked T-cell activation by InsB:9-23 in a humanized DQ8 mice both ex vivo and in vivo, as shown by decreased production of IL-2 and IFN-γ and reduced lymphocyte proliferation. Interestingly, RI-EXT also blocked lymphocyte activation and proliferation by InsB:9-23 in PBMCs isolated from recent onset DQ8-T1D patients. In summary, we discovered a RI-D-peptide that blocks InsB:9-23 binding to HLA-DQ8 and its presentation to T-cells in T1D. These findings set the stage for using our approach as a novel therapy for patients with T1D and potentially other autoimmune diseases.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Antígenos HLA-DQ/metabolismo , Células Secretoras de Insulina/inmunología , Péptidos/antagonistas & inhibidores , Animales , Presentación de Antígeno/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Femenino , Antígenos HLA-DQ/inmunología , Antígenos HLA-DQ/aislamiento & purificación , Humanos , Células Secretoras de Insulina/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Simulación de Dinámica Molecular , Péptidos/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
5.
J Autoimmun ; 108: 102402, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31980336

RESUMEN

We have previously identified a signature HLA-DR3 pocket variant, designated HLA-DRß1-Arg74 that confers a high risk for Graves' Disease (GD). In view of the key role of HLA-DRß1-Arg74 in triggering GD we hypothesized that thyroid-stimulating hormone receptor (TSHR) peptides that bind to the HLA-DRß1-Arg74 pocket with high affinity represent key pathogenic TSHR peptides triggering GD, and that blocking their presentation to CD4+ T-cells can be used as a novel therapeutic approach in GD. There were several previous attempts to identify the major pathogenic TSHR peptide utilizing different methodologies, however the results were inconsistent and inconclusive. Therefore, the aim of our study was to use TSHR peptide binding affinity to HLA-DRß1-Arg74 as a method to identify the key pathogenic TSHR peptides that trigger GD. Using virtual screening and ELISA and cellular binding assays we identified 2 TSHR peptides that bound with high affinity to HLA-DRß1-Arg74 - TSHR.132 and TSHR.197. Peptide immunization studies in humanized DR3 mice showed that only TSHR.132, but not TSHR.197, induced autoreactive T-cell proliferation and cytokine responses. Next, we induced experimental autoimmune Graves' disease (EAGD) in a novel BALB/c-DR3 humanized mouse model we created and confirmed TSHR.132 as a major DRß1-Arg74 binding peptide triggering GD in our mouse model. Furthermore, we demonstrated that Cepharanthine, a compound we have previously identified as DRß1-Arg74 blocker, could block the presentation and T-cell responses to TSHR.132 in the EAGD model.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Bencilisoquinolinas/farmacología , Antígeno HLA-DR3/inmunología , Péptidos/antagonistas & inhibidores , Péptidos/inmunología , Receptores de Tirotropina/inmunología , Secuencia de Aminoácidos , Animales , Bencilisoquinolinas/química , Mapeo Epitopo/métodos , Epítopos de Linfocito T/inmunología , Citometría de Flujo , Enfermedad de Graves/diagnóstico , Enfermedad de Graves/tratamiento farmacológico , Enfermedad de Graves/inmunología , Antígeno HLA-DR3/genética , Humanos , Inmunohistoquímica , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Moleculares , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Péptidos/química , Unión Proteica , Receptores de Tirotropina/química , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
6.
Proteins ; 88(3): 414-430, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31587361

RESUMEN

Bromodomains (BrDs), a conserved structural module in chromatin-associated proteins, are well known for recognizing ε-N-acetyl lysine residues on histones. One of the most relevant BrDs is BRD4, a tandem BrD containing protein (BrD1 and BrD2) that plays a critical role in numerous diseases including cancer. Growing evidence shows that the two BrDs of BRD4 have different biological functions; hence selective ligands that can be used to study their functions are of great interest. Here, as a follow-up of our previous work, we first provide a detailed characterization study of the in silico rational design of Olinone as part of a series of five tetrahydropyrido indole-based compounds as BRD4 BrD1 inhibitors. Additionally, we investigated the molecular basis for Olinone's selective recognition by BrD1 over BrD2. Molecular dynamics simulations, free energy calculations, and conformational analyses of the apo-BRD4-BrD1|2 and BRD4-BrD1|2/Olinone complexes showed that Olinone's selectivity is facilitated by five key residues: Leu92 in BrD1|385 in BrD2 of ZA loop, Asn140|433, Asp144|His437 and Asp145|Glu438 of BC loop, and Ile146|Val49 of helix C. Furthermore, the difference in hydrogen bonds number and in mobility of the ZA and BC loops of the acetyl-lysine binding site between BRD4 BrD1/Olinone and BrD2/Olinone complexes also contribute to the difference in Olinone's binding affinity and selectivity toward BrD1 over BrD2. Altogether, our computer-aided molecular design techniques can effectively guide the development of small-molecule BRD4 BrD1 inhibitors, explain their selectivity origin, and further open doors to the design of new therapeutically improved derivatives.


Asunto(s)
Apoproteínas/antagonistas & inhibidores , Proteínas de Ciclo Celular/antagonistas & inhibidores , Histona Acetiltransferasas/antagonistas & inhibidores , Indoles/química , Factores de Transcripción/antagonistas & inhibidores , Apoproteínas/química , Apoproteínas/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Cristalografía por Rayos X , Diseño de Fármacos , Histona Acetiltransferasas/química , Histona Acetiltransferasas/metabolismo , Humanos , Enlace de Hidrógeno , Indoles/metabolismo , Cinética , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad , Termodinámica , Factores de Transcripción/química , Factores de Transcripción/metabolismo
7.
Biophys J ; 114(7): 1579-1589, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29642028

RESUMEN

Intramembrane-cleaving proteases (I-CLiPs) activate pools of single-pass helical membrane protein signaling precursors that are key in the physiology of prokaryotic and eukaryotic cells. Proteases typically cleave peptide bonds within extended or flexible regions of their substrates, and thus the mechanism underlying the ability of I-CLiPs to hydrolyze the presumably α-helical transmembrane domain (TMD) of these membrane proteins is unclear. Using deep-ultraviolet resonance Raman spectroscopy in combination with isotopic labeling, we show that although predominantly in canonical α-helical conformation, the TMD of the established I-CLiP substrate Gurken displays 310-helical geometry. As measured by microscale thermophoresis, this substrate binds with high affinity to the I-CLiPs GlpG rhomboid and MCMJR1 presenilin homolog in detergent micelles. Binding results in deep-ultraviolet resonance Raman spectra, indicating conformational changes consistent with unwinding of the 310-helical region of the substrate's TMD. This 310-helical conformation is key for intramembrane proteolysis, as the substitution of a single proline residue in the TMD of Gurken by alanine suppresses 310-helical content in favor of α-helical geometry and abolishes cleavage without affecting binding to the I-CLiP. Complemented by molecular dynamics simulations of the TMD of Gurken, our vibrational spectroscopy data provide biophysical evidence in support of a model in which the transmembrane region of cleavable I-CLiP substrates displays local deviations in canonical α-helical conformation characterized by chain flexibility, and binding to the enzyme results in conformational changes that facilitate local unwinding of the transmembrane helix for cleavage.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteolisis , Secuencia de Aminoácidos , Simulación de Dinámica Molecular , Péptido Hidrolasas/metabolismo , Conformación Proteica en Hélice alfa
8.
J Autoimmun ; 76: 1-9, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27670087

RESUMEN

Autoimmune polyglandular syndrome 3 variant (APS3v) refers to the co-occurrence of autoimmune thyroiditis (AITD) and type 1 diabetes (T1D) within the same individual. HLA class II confers the strongest susceptibility to APS3v. We previously identified a unique amino acid signature of the HLA-DR pocket (designated APS3v HLA-DR pocket) that predisposes to APS3v. We hypothesized that both thyroid and islet peptides can be presented by the unique APS3v HLA-DR pocket, triggering AITD + T1D together. To test this hypothesis we screened islet and thyroid peptides for their ability to bind to the APS3v HLA-DR pocket. Virtual screen of all possible thyroglobulin (Tg), thyroid-stimulating hormone receptor (TSHR), thyroid peroxidase (TPO), insulin (Ins), and glutamic acid decarboxylase 65 (GAD65) peptides identified 36 peptides that bound to this unique pocket. In vitro binding assays using baculovirus-produced recombinant APS3v HLA-DR identified 11 thyroid/islet peptides (of the 36 predicted binders) that bound with high affinity. By immunizing humanized HLA-DR3 mice carrying the APS3v HLA-DR pocket we identified 4 peptides (Tg.1571, GAD.492, TPO.758, TPO.338) that were presented by antigen presenting cells and elicited T-cell response. We conclude that both thyroid and islet peptides can bind to this flexible APS3v HLA-DR pocket and induce thyroid and islet specific T-cell responses. These findings set the stage to developing specific inhibitors of the APS3v HLA-DR pocket as a precision medicine approach to treating or preventing APS3v in patients that carry this genetic HLA-DR pocket variant.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/inmunología , Antígenos HLA-DR/inmunología , Péptidos/inmunología , Linfocitos T/inmunología , Tiroiditis Autoinmune/inmunología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Antígenos HLA-DR/química , Antígenos HLA-DR/metabolismo , Humanos , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Modelos Moleculares , Péptidos/química , Péptidos/metabolismo , Unión Proteica/inmunología , Conformación Proteica , Linfocitos T/metabolismo , Glándula Tiroides/inmunología , Tiroiditis Autoinmune/metabolismo
9.
PLoS One ; 11(2): e0148762, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26910052

RESUMEN

The MAGE (melanoma associated antigen) protein family are tumour-associated proteins normally present only in reproductive tissues such as germ cells of the testis. The human genome encodes over 60 MAGE genes of which one class (containing MAGE-A3 and MAGE-A4) are exclusively expressed in tumours, making them an attractive target for the development of targeted and immunotherapeutic cancer treatments. Some MAGE proteins are thought to play an active role in driving cancer, modulating the activity of E3 ubiquitin ligases on targets related to apoptosis. Here we determined the crystal structures of MAGE-A3 and MAGE-A4. Both proteins crystallized with a terminal peptide bound in a deep cleft between two tandem-arranged winged helix domains. MAGE-A3 (but not MAGE-A4), is predominantly dimeric in solution. Comparison of MAGE-A3 and MAGE-A3 with a structure of an effector-bound MAGE-G1 suggests that a major conformational rearrangement is required for binding, and that this conformational plasticity may be targeted by allosteric binders.


Asunto(s)
Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
10.
J Comput Chem ; 37(10): 913-9, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26691434

RESUMEN

Dynamic characteristics of protein surfaces are among the factors determining their functional properties, including their potential participation in protein-protein interactions. The presence of clusters of static residues-"stability patches" (SPs)-is a characteristic of protein surfaces involved in intermolecular recognition. The mechanism, by with SPs facilitate molecular recognition, however, remains unclear. Analyzing the surface dynamic properties of the growth hormone and of its high-affinity variant we demonstrated that reshaping of the SPs landscape may be among the factors accountable for the improved affinity of this variant to the receptor. We hypothesized that SPs facilitate molecular recognition by moderating the conformational entropy of the unbound state, diminishing enthalpy-entropy compensation upon binding, and by augmenting the favorable entropy of desolvation. SPs mapping emerges as a valuable tool for investigating the structural basis of the stability of protein complexes and for rationalizing experimental approaches, such as affinity maturation, aimed at improving it.


Asunto(s)
Hormona de Crecimiento Humana/química , Receptores de Somatotropina/química , Humanos , Simulación de Dinámica Molecular , Estabilidad Proteica , Termodinámica
11.
J Biol Chem ; 291(8): 4079-90, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26703475

RESUMEN

We previously showed that an HLA-DR variant containing arginine at position 74 of the DRß1 chain (DRß1-Arg74) is the specific HLA class II variant conferring risk for autoimmune thyroid diseases (AITD). We also identified 5 thyroglobulin (Tg) peptides that bound to DRß1-Arg74. We hypothesized that blocking the binding of these peptides to DRß1-Arg74 could block the continuous T-cell activation in thyroiditis needed to maintain the autoimmune response to the thyroid. The aim of the current study was to identify small molecules that can block T-cell activation by Tg peptides presented within DRß1-Arg74 pockets. We screened a large and diverse library of compounds and identified one compound, cepharanthine that was able to block peptide binding to DRß1-Arg74. We then showed that Tg.2098 is the dominant peptide when inducing experimental autoimmune thyroiditis (EAT) in NOD mice expressing human DRß1-Arg74. Furthermore, cepharanthine blocked T-cell activation by thyroglobulin peptides, in particular Tg.2098 in mice that were induced with EAT. For the first time we identified a small molecule that can block Tg peptide binding and presentation to T-cells in autoimmune thyroiditis. If confirmed cepharanthine could potentially have a role in treating human AITD.


Asunto(s)
Alcaloides/farmacología , Presentación de Antígeno/efectos de los fármacos , Cadenas HLA-DRB1/inmunología , Tiroiditis Autoinmune/inmunología , Alcaloides/química , Animales , Cadenas HLA-DRB1/genética , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Transgénicos , Péptidos/genética , Péptidos/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Tiroglobulina/genética , Tiroglobulina/inmunología , Tiroiditis Autoinmune/genética , Tiroiditis Autoinmune/patología
12.
Chem Senses ; 40(8): 577-86, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26377607

RESUMEN

The sweet taste receptor, a heterodimeric G protein-coupled receptor comprised of T1R2 and T1R3, binds sugars, small molecule sweeteners, and sweet proteins to multiple binding sites. The dipeptide sweetener, aspartame binds in the Venus Flytrap Module (VFTM) of T1R2. We developed homology models of the open and closed forms of human T1R2 and human T1R3 VFTMs and their dimers and then docked aspartame into the closed form of T1R2's VFTM. To test and refine the predictions of our model, we mutated various T1R2 VFTM residues, assayed activity of the mutants and identified 11 critical residues (S40, Y103, D142, S144, S165, S168, Y215, D278, E302, D307, and R383) in and proximal to the binding pocket of the sweet taste receptor that are important for ligand recognition and activity of aspartame. Furthermore, we propose that binding is dependent on 2 water molecules situated in the ligand pocket that bridge 2 carbonyl groups of aspartame to residues D142 and L279. These results shed light on the activation mechanism and how signal transmission arising from the extracellular domain of the T1R2 monomer of the sweet receptor leads to the perception of sweet taste.


Asunto(s)
Aspartame/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Edulcorantes/metabolismo , Animales , Aspartame/química , Sitios de Unión , Dimerización , Células HEK293 , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Cuaternaria de Proteína , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Edulcorantes/química , Gusto/fisiología
13.
Nucleic Acids Res ; 42(19): 12272-83, 2014 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-25260586

RESUMEN

We present the results of microsecond molecular dynamics simulations carried out by the ABC group of laboratories on a set of B-DNA oligomers containing the 136 distinct tetranucleotide base sequences. We demonstrate that the resulting trajectories have extensively sampled the conformational space accessible to B-DNA at room temperature. We confirm that base sequence effects depend strongly not only on the specific base pair step, but also on the specific base pairs that flank each step. Beyond sequence effects on average helical parameters and conformational fluctuations, we also identify tetranucleotide sequences that oscillate between several distinct conformational substates. By analyzing the conformation of the phosphodiester backbones, it is possible to understand for which sequences these substates will arise, and what impact they will have on specific helical parameters.


Asunto(s)
ADN Forma B/química , Emparejamiento Base , Secuencia de Bases , Simulación de Dinámica Molecular , Conformación de Ácido Nucleico
14.
PLoS Comput Biol ; 8(9): e1002693, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028290

RESUMEN

In RNA interference, a guide strand derived from a short dsRNA such as a microRNA (miRNA) is loaded into Argonaute, the central protein in the RNA Induced Silencing Complex (RISC) that silences messenger RNAs on a sequence-specific basis. The positions of any mismatched base pairs in an miRNA determine which Argonaute subtype is used. Subsequently, the Argonaute-guide complex binds and silences complementary target mRNAs; certain Argonautes cleave the target. Mismatches between guide strand and the target mRNA decrease cleavage efficiency. Thus, loading and silencing both require that signals about the presence of a mismatched base pair are communicated from the mismatch site to effector sites. These effector sites include the active site, to prevent target cleavage; the binding groove, to modify nucleic acid binding affinity; and surface allosteric sites, to control recruitment of additional proteins to form the RISC. To examine how such signals may be propagated, we analyzed the network of internal allosteric pathways in Argonaute exhibited through correlations of residue-residue interactions. The emerging network can be described as a set of pathways emanating from the core of the protein near the active site, distributed into the bulk of the protein, and converging upon a distributed cluster of surface residues. Nucleotides in the guide strand "seed region" have a stronger relationship with the protein than other nucleotides, concordant with their importance in sequence selectivity. Finally, any of several seed region guide-target mismatches cause certain Argonaute residues to have modified correlations with the rest of the protein. This arises from the aggregation of relatively small interaction correlation changes distributed across a large subset of residues. These residues are in effector sites: the active site, binding groove, and surface, implying that direct functional consequences of guide-target mismatches are mediated through the cumulative effects of a large number of internal allosteric pathways.


Asunto(s)
Proteínas Argonautas/fisiología , MicroARNs/fisiología , Modelos Genéticos , Mapeo de Interacción de Proteínas/métodos , Interferencia de ARN , Transducción de Señal/genética , Sitios de Unión , Simulación por Computador , Unión Proteica
15.
Proteins ; 80(5): 1283-98, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22275138

RESUMEN

Silencing in RNAi is strongly affected by guide-strand/target-mRNA mismatches. Target nucleation is thought to occur at positions 2-8 of the guide ("seed region"); successful hybridization in this region is the primary determinant of target-binding affinity and hence target cleavage. To define a molecular basis for the target sequence selectivity in RNAi, we studied all possible distinct single mismatches in seven positions of the seed region-a total of 21 substitutions. We report results from soft-core thermodynamic integration simulations to determine changes in targeting binding-free energies to Argonaute due to single mismatches in the guide strand, which arise during binding of an imperfectly matched target mRNA. In agreement with experiment, most mismatches impair target binding, consistent with a prominent role for binding affinity changes in RNAi sequence selectivity. Individual Argonaute residues located near the mismatched base pair are found to contribute significantly to binding affinity changes. We also use this methodology to analyze the mismatch-dependent free energy changes for dissociation of a DNA•RNA hybrid from Argonaute, as a model for the escape of miRNAs from the silencing pathway. Several mismatched sequences of the miRNA have increased affinity to Argonaute, implying that some mismatches may reduce the probability for escape. Furthermore, calculations of base-substitution-dependent free energy changes for binding ssDNA reveal mild sequence sensitivity as expected for guide strand binding to Argonaute. Our findings give a thermodynamic basis for RNAi target sequence selectivity and suggest that miRNA mismatches may increase silencing effectiveness and thus could be evolutionarily advantageous.


Asunto(s)
Proteínas Argonautas/química , Proteínas Argonautas/fisiología , MicroARNs/química , MicroARNs/fisiología , Simulación de Dinámica Molecular , Interferencia de ARN/fisiología , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Emparejamiento Base , ADN/genética , ADN/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Termodinámica
16.
J Am Chem Soc ; 133(7): 2040-3, 2011 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-21271695

RESUMEN

The CREB binding protein (CBP) is a human transcriptional coactivator consisting of several conserved functional modules, which interacts with distinct transcription factors including nuclear receptors, CREB, and STAT proteins. Despite the importance of CBP in transcriptional regulation, many questions regarding the role of its particular domains in CBP functions remain unanswered. Therefore, developing small molecules capable of selectively modulating a single domain of CBP is of invaluable aid at unraveling its prominent activities. Here we report the design, synthesis, and biological evaluation of conformationally restricted peptides as novel modulators for the acetyl-lysine binding bromodomain (BRD) of CBP. Utilizing a target structure-guided and computer-aided rational design approach, we developed a series of cyclic peptides with affinity for CBP BRD significantly greater than those of its biological ligands, including lysine-acetylated histones and tumor suppressor p53. The best cyclopeptide of the series exhibited a K(d) of 8.0 µM, representing a 24-fold improvement in affinity over that of the linear lysine 382-acetylated p53 peptide. This lead peptide is highly selective for CBP BRD over BRDs from other transcriptional proteins. Cell-based functional assays carried out in colorectal carcinoma HCT116 cells further demonstrated the efficacy of this compound to modulate p53 stability and function in response to DNA damage. Our results strongly argue that these CBP modulators can effectively inhibit p53 transcriptional activity by blocking p53K382ac binding to CBP BRD and promoting p53 instability by changes of its post-translational modification states, a different mechanism than that of the p53 inhibitors reported to date.


Asunto(s)
Proteína de Unión a CREB/efectos de los fármacos , Diseño de Fármacos , Péptidos Cíclicos/síntesis química , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Estructura Molecular , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Relación Estructura-Actividad
17.
Proc Natl Acad Sci U S A ; 107(39): 16899-903, 2010 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-20837527

RESUMEN

There is strong genetic association between type 1A diabetes (T1D) and autoimmune thyroid disease (AITD). T1D and AITD frequently occur together in the same individual, a condition classified as a variant of the autoimmune polyglandular syndrome type 3 (APS3). Because T1D and AITD are individually strongly associated with different HLA class II sequences, we asked which HLA class II pocket sequence and structure confer joint susceptibility to both T1D and AITD in the same individual (APS3v). We sequenced the HLA-DR gene in 105 APS3v patients and 153 controls, and identified a pocket amino acid signature, DRß-Tyr-26, DRß-Leu-67, DRß-Lys-71, and DRß-Arg-74, that was strongly associated with APS3v (P = 5.4 × 10(-14), odds ratio = 8.38). Logistic regression analysis demonstrated that DRß-Leu-67 (P = 9.4 × 10(-13)) and DRß-Arg-74 (P = 1.21 × 10(-13)) gave strong independent effects on disease susceptibility. Structural modeling studies demonstrated that pocket 4 was critical for the development of T1D+AITD; all disease-associated amino acids were linked to areas of the pocket that interact directly with the peptide and, therefore, influence peptide binding. The disease-susceptible HLA-DR pocket was more positively charged (Lys-71, Arg-74) compared with the protective pocket (Ala-71, Gln-74). We conclude that a specific pocket amino acid signature confers joint susceptibility to T1D+AITD in the same individual by causing significant structural changes in the MHC II peptide binding pocket and influencing peptide binding and presentation. Moreover, Arg-74 is a major amino acid position for the development of several autoimmune diseases. These findings suggest that blocking the critical Arg-74 pocket might offer a method for treating certain autoimmune conditions.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Antígenos HLA-DR/genética , Tiroiditis Autoinmune/genética , Secuencias de Aminoácidos , Arginina/química , Arginina/genética , Sitios de Unión , Femenino , Antígenos HLA-DR/química , Humanos , Masculino , Unión Proteica , Conformación Proteica , Análisis de Secuencia de Proteína
18.
Biophys J ; 98(2): 240-7, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20338845

RESUMEN

Phosphoinositides like phosphatidylinositol 4,5-bisphosphate (PIP(2)) are negatively charged lipids that play a pivotal role in membrane trafficking, signal transduction, and protein anchoring. We have designed a force field for the PIP(2) headgroup using quantum mechanical methods and characterized its properties inside a lipid bilayer using molecular dynamics simulations. Macroscopic properties such as area/headgroup, density profiles, and lipid order parameters calculated from these simulations agree well with the experimental values. However, microscopically, the PIP(2) introduces a local perturbation of the lipid bilayer. The average PIP(2) headgroup orientation of 45 degrees relative to the bilayer normal induces a unique, distance-dependent organization of the lipids that surround PIP(2). The headgroups of these lipids preferentially orient closer to the bilayer normal. This perturbation creates a PIP(2) lipid microdomain with the neighboring lipids. We propose that the PIP(2) lipid microdomain enables the PIP(2) to function as a membrane-bound anchoring molecule.


Asunto(s)
Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Fosfatidilinositol 4,5-Difosfato/química , 1,2-Dipalmitoilfosfatidilcolina/química , Algoritmos
19.
J Autoimmun ; 35(1): 45-51, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20303712

RESUMEN

An HLA-DR variant containing Arginine at position 74 of the DRbeta1 chain confers a strong genetic susceptibility to autoimmune thyroid diseases (AITD), Graves' disease (GD) and Hashimoto's thyroiditis (HT), while Glutamine at position DRbeta1-74 is protective. We hypothesized that the DRbeta1-Arg74 variant is able to present pathogenic thyroglobulin (Tg) peptides to T-cells more efficiently, thereby triggering thyroid autoimmunity. Indeed, we have previously identified 4 human Tg (hTg) peptides that bind specifically to DRbeta1-Arg74 with much weaker binding to the protective variant DRbeta1-Gln74. The aim of our study was to examine in vivo whether an hTg peptide that binds strongly and specifically to DRbeta1-Arg74 is capable of stimulating T-cells during the induction of thyroiditis in a "humanized" mouse expressing human DR3, and in patients positive for Tg antibodies. Sequencing of exon 2 of the DR transgene in the DR3 mice, null for endogenous MHC II molecules, confirmed that they expressed the disease-associated DRbeta1-Arg74 variant, thus making them an ideal in vivo model to test the presentation of hTg peptides by DRbeta1-Arg74 HLA-DR. Induction of EAT in the DR3 mice lead to T-cell stimulation and proliferation to Tg.2098, a strong and specific DRbeta1-Arg74 binder, while a non-binding control peptide, Tg.2766 did not induce this response. Moreover, Tg.2098 stimulated T-cells from 4 individuals who were positive for thyroglobulin antibodies, demonstrating that Tg.2098 is an immunogenic peptide capable of being presented in vivo and activating T-cells in EAT and AITD. Energetic analysis of the complex formed by Tg.2098 and DRbeta-Arg74 has shown that the origin of the affinity was determined by residues 1, 7 and 9 in the peptide, while the selectivity of the peptide for the MHC was determined by the Asp in position 4. The disease-protective substitution R74Q, leads to reduction in affinity due to changes in local interaction with D4 as well as non-local interaction with other residues. The electrostatic potential on the surface of the DRbeta-Arg74-Tg.2098 complex has a unique signature which may be recognized by T-cell receptors leading to autoimmune thyroiditis. Taken together these findings suggest that Tg.2098, a strong and specific binder to the disease-associated HLA-DRbeta-Arg74, is a major human T-cell epitope and participant in the pathoetiology of AITD.


Asunto(s)
Epítopos de Linfocito T/inmunología , Fragmentos de Péptidos/inmunología , Linfocitos T/metabolismo , Tiroglobulina/inmunología , Tiroiditis Autoinmune/inmunología , Animales , Presentación de Antígeno/genética , Proliferación Celular , Mapeo Epitopo , Predisposición Genética a la Enfermedad , Antígeno HLA-DR3/genética , Antígenos de Histocompatibilidad Clase II/genética , Interleucina-2/metabolismo , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Polimorfismo Genético , Unión Proteica/genética , Unión Proteica/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Tiroiditis Autoinmune/genética
20.
Proteins ; 78(2): 447-56, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19731376

RESUMEN

Targeted therapeutic intervention in receptor-ligand interactions of p53-mediated tumor suppression can impact progression of disease, aging, and variation in genetic expression. Here, we conducted a number of molecular simulations, based on structures of p53 in complex with its transcriptional coactivating CBP bromodomain, determined by NMR spectroscopy, to investigate the energetics of the binding complex. Building on the observation that acetylation of K382 in p53 serves as the essential triggering switch for a specific interaction with CBP, we assessed the differential effect of acetylation on binding from simulations of an octapeptide derived from p53 with acetylated and nonacetylated K382 (residues 379-386). Cluster analysis of the simulations shows that acetylation of the free peptide does not significantly change the population of the preferred conformation of the peptide in solution for binding to CBP. Conversion of the acetylated K382 to nonacetylated form with free energy perturbation (FEP) simulations of the p53 CBP complex and the free peptide showed that the relative contribution of the acetyl group to binding is 4.8 kcal/mol. An analysis of residue contributions to the binding energy using an MM-GBSA approach agrees with the FEP results and sheds additional light on the origin of selectivity in p53 binding to the CBP bromodomain.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Proteína de Unión a CREB/química , Humanos , Simulación de Dinámica Molecular , Mutación , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Termodinámica , Activación Transcripcional , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...