Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Protein Sci ; 33(6): e4999, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38723106

RESUMEN

Ticks produce chemokine-binding proteins, known as evasins, in their saliva to subvert the host's immune response. Evasins bind to chemokines and thereby inhibit the activation of their cognate chemokine receptors, thus suppressing leukocyte recruitment and inflammation. We recently described subclass A3 evasins, which, like other class A evasins, exclusively target CC chemokines but appear to use a different binding site architecture to control target selectivity among CC chemokines. We now describe the structural basis of chemokine recognition by the class A3 evasin EVA-ACA1001. EVA-ACA1001 binds to almost all human CC chemokines and inhibits receptor activation. Truncation mutants of EVA-ACA1001 showed that, unlike class A1 evasins, both the N- and C-termini of EVA-ACA1001 play minimal roles in chemokine binding. To understand the structural basis of its broad chemokine recognition, we determined the crystal structure of EVA-ACA1001 in complex with the human chemokine CCL16. EVA-ACA1001 forms backbone-backbone interactions with the CC motif of CCL16, a conserved feature of all class A evasin-chemokine complexes. A hydrophobic pocket in EVA-ACA1001, formed by several aromatic side chains and the unique disulfide bond of class A3 evasins, accommodates the residue immediately following the CC motif (the "CC + 1 residue") of CCL16. This interaction is shared with EVA-AAM1001, the only other class A3 evasins characterized to date, suggesting it may represent a common mechanism that accounts for the broad recognition of CC chemokines by class A3 evasins.


Asunto(s)
Modelos Moleculares , Humanos , Animales , Garrapatas/química , Garrapatas/metabolismo , Cristalografía por Rayos X , Sitios de Unión , Proteínas de Artrópodos/química , Proteínas de Artrópodos/metabolismo , Proteínas de Artrópodos/genética , Unión Proteica , Quimiocinas/química , Quimiocinas/metabolismo , Proteínas y Péptidos Salivales/química , Proteínas y Péptidos Salivales/metabolismo
2.
Nat Commun ; 14(1): 4204, 2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-37452046

RESUMEN

Chemokines are key regulators of leukocyte trafficking and attractive targets for anti-inflammatory therapy. Evasins are chemokine-binding proteins from tick saliva, whose application as anti-inflammatory therapeutics will require manipulation of their chemokine target selectivity. Here we describe subclass A3 evasins, which are unique to the tick genus Amblyomma and distinguished from "classical" class A1 evasins by an additional disulfide bond near the chemokine recognition interface. The A3 evasin EVA-AAM1001 (EVA-A) bound to CC chemokines and inhibited their receptor activation. Unlike A1 evasins, EVA-A was not highly dependent on N- and C-terminal regions to differentiate chemokine targets. Structures of chemokine-bound EVA-A revealed a deep hydrophobic pocket, unique to A3 evasins, that interacts with the residue immediately following the CC motif of the chemokine. Mutations to this pocket altered the chemokine selectivity of EVA-A. Thus, class A3 evasins provide a suitable platform for engineering proteins with applications in research, diagnosis or anti-inflammatory therapy.


Asunto(s)
Garrapatas , Animales , Garrapatas/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Quimiocinas/metabolismo , Quimiocinas CC/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/metabolismo
3.
mBio ; 13(4): e0206422, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35924852

RESUMEN

Cytoplasmic detection of DNA by cyclic GMP-AMP (cGAMP) synthase (cGAS) is an essential component of antiviral responses. Upon synthesis, cGAMP binds to the stimulator of interferon (IFN) genes (STING) in infected and adjacent cells through intercellular transfer by connexins forming gap-junctions, eliciting a strong IFN-ß-driven antiviral response. We demonstrate here that Genistein, a flavonoid compound naturally occurring in soy-based foods, inhibits cGAS-STING antiviral signaling at two levels. First, Genistein pretreatment of cGAMP-producing cells inhibited gap-junction intercellular communication, resulting in reduced STING responses in adjacent cells. In addition, Genistein directly blocked STING activation by the murine agonist DMXAA, by decreasing the interaction of STING with TBK1 and IKKε. As a result, Genistein attenuated STING signaling in human and mouse cells, dampening antiviral activity against Semliki Forest Virus infection. Collectively, our findings identify a previously unrecognized proviral activity of Genistein mediated via its inhibitory effects at two levels of cGAS-STING signaling. IMPORTANCE Several reports suggest that Genistein exhibits antiviral activities against DNA viruses. Our work uncovers a previously unrecognized proviral effect of Genistein, through inhibition of the cGAS-STING pathway at the level of cGAMP transfer and its sensing by STING. This suggests that the use of Genistein as an antiviral should be taken with caution as it may reduce the protective antiviral effects elicited by host STING activation.


Asunto(s)
Genisteína , Proteínas de la Membrana , Animales , Antivirales/farmacología , Genisteína/farmacología , Humanos , Inmunidad Innata/genética , Interferón beta/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Nucleotidiltransferasas/genética
4.
Biomedicines ; 10(5)2022 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-35625882

RESUMEN

The development of peptide inhibitors against intracellular targets depends upon the dual challenge of achieving a high affinity and specificity for the target and maintaining cellular permeability for biological activity. Previous efforts to develop bicyclic peptides targeted to the Grb7 signalling protein implicated in HER2+ve cancer progression have resulted in improved affinity. However, these same peptides demonstrated a lowered activity due to their decreased ability to penetrate cell membranes. Here, we report the testing of a new series of bicyclic G7 peptides designed to possess improved bioactivity. We discovered that the incorporation of two amino acids (Phe-Pro, Phe-Trp or Phe-Arg) within the bicyclic peptide framework maintains an enhanced binding affinity for the Grb7-SH2 domain compared to that of the first-generation monocyclic peptide G7-18NATE. Structure determination using X-ray crystallography revealed that the mode of binding by the expanded bicyclic G7 peptide is analogous to that of G7-18NATE. Interestingly, while the bicyclic peptide containing Phe-Trp did not display the highest affinity for Grb7-SH2 in the series, it was the most potent inhibitor of HER2+ve SKBR3 breast cancer cell migration when coupled to Penetratin. Together, this demonstrates that peptide flexibility as well as the amino acid tryptophan can play important roles in the uptake of peptides into the cell.

5.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35217625

RESUMEN

As natural chemokine inhibitors, evasin proteins produced in tick saliva are potential therapeutic agents for numerous inflammatory diseases. Engineering evasins to block the desired chemokines and avoid off-target side effects requires structural understanding of their target selectivity. Structures of the class A evasin EVA-P974 bound to human CC chemokine ligands 7 and 17 (CCL7 and CCL17) and to a CCL8-CCL7 chimera reveal that the specificity of class A evasins for chemokines of the CC subfamily is defined by conserved, rigid backbone-backbone interactions, whereas the preference for a subset of CC chemokines is controlled by side-chain interactions at four hotspots in flexible structural elements. Hotspot mutations alter target preference, enabling inhibition of selected chemokines. The structure of an engineered EVA-P974 bound to CCL2 reveals an underlying molecular mechanism of EVA-P974 target preference. These results provide a structure-based framework for engineering evasins as targeted antiinflammatory therapeutics.


Asunto(s)
Proteínas de Artrópodos/química , Quimiocinas/metabolismo , Inflamación/metabolismo , Ingeniería de Proteínas , Garrapatas/metabolismo , Animales , Proteínas de Artrópodos/metabolismo , Unión Proteica , Conformación Proteica , Receptores de Quimiocina/metabolismo
6.
Nucleic Acids Res ; 49(5): 2403-2417, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33621982

RESUMEN

TIA-1 is an RNA-binding protein that sequesters target RNA into stress granules under conditions of cellular stress. Promotion of stress granule formation by TIA-1 depends upon self-association of its prion-like domain that facilitates liquid-liquid phase separation and is thought to be enhanced via RNA binding. However, the mechanisms underlying the influence of RNA on TIA-1 self-association have not been previously demonstrated. Here we have investigated the self-associating properties of full-length TIA-1 in the presence of designed and native TIA-1 nucleic acid binding sites in vitro, monitoring phase separation, fibril formation and shape. We show that single stranded RNA and DNA induce liquid-liquid phase separation of TIA-1 in a multisite, sequence-specific manner and also efficiently promote formation of amyloid-like fibrils. Although RNA binding to a single site induces a small conformational change in TIA-1, this alone does not enhance phase separation of TIA-1. Tandem binding sites are required to enhance phase separation of TIA-1 and this is finely tuned by the protein:binding site stoichiometry rather than nucleic acid length. Native tandem TIA-1 binding sites within the 3' UTR of p53 mRNA also efficiently enhance phase separation of TIA-1 and thus may potentially act as potent nucleation sites for stress granule assembly.


Asunto(s)
ARN/metabolismo , Antígeno Intracelular 1 de las Células T/química , Regiones no Traducidas 3' , Amiloide/ultraestructura , Sitios de Unión , ADN/química , ADN/metabolismo , Humanos , Modelos Moleculares , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Conformación Proteica , ARN/química , Antígeno Intracelular 1 de las Células T/metabolismo , Antígeno Intracelular 1 de las Células T/ultraestructura , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
Nucleic Acids Res ; 48(14): 8006-8021, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32556302

RESUMEN

The poliovirus type I IRES is able to recruit ribosomal machinery only in the presence of host factor PCBP2 that binds to stem-loop IV of the IRES. When PCBP2 is cleaved in its linker region by viral proteinase 3CD, translation initiation ceases allowing the next stage of replication to commence. Here, we investigate the interaction of PCBP2 with the apical region of stem-loop IV (SLIVm) of poliovirus RNA in its full-length and truncated form. CryoEM structure reconstruction of the full-length PCBP2 in complex with SLIVm solved to 6.1 Å resolution reveals a compact globular complex of PCBP2 interacting with the cruciform RNA via KH domains and featuring a prominent GNRA tetraloop. SEC-SAXS, SHAPE and hydroxyl-radical cleavage establish that PCBP2 stabilizes the SLIVm structure, but upon cleavage in the linker domain the complex becomes more flexible and base accessible. Limited proteolysis and REMSA demonstrate the accessibility of the linker region in the PCBP2/SLIVm complex and consequent loss of affinity of PCBP2 for the SLIVm upon cleavage. Together this study sheds light on the structural features of the PCBP2/SLIV complex vital for ribosomal docking, and the way in which this key functional interaction is regulated following translation of the poliovirus genome.


Asunto(s)
Iniciación de la Cadena Peptídica Traduccional , Poliovirus/genética , ARN Viral/química , Proteínas de Unión al ARN/química , Microscopía por Crioelectrón , Modelos Moleculares , Conformación de Ácido Nucleico , Conformación Proteica , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Dispersión del Ángulo Pequeño , Difracción de Rayos X
8.
Endocrinology ; 161(8)2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32569368

RESUMEN

Ovarian-derived inhibin A and inhibin B (heterodimers of common α- and differing ß-subunits) are secreted throughout the menstrual cycle in a discordant pattern, with smaller follicles producing inhibin B, whereas the dominant follicle and corpus luteum produce inhibin A. The classical function for endocrine inhibins is to block signalling by activins (homodimers of ß-subunits) in gonadotrope cells of the anterior pituitary and, thereby, inhibit the synthesis of FSH. Whether inhibin A and inhibin B have additional physiological functions is unknown, primarily because producing sufficient quantities of purified inhibins, in the absence of contaminating activins, for preclinical studies has proven extremely difficult. Here, we describe novel methodology to enhance inhibin A and inhibin B activity and to produce these ligands free of contaminating activins. Using computational modeling and targeted mutagenesis, we identified a point mutation in the activin ß A-subunit, A347H, which completely disrupted activin dimerization and activity. Importantly, this ß A-subunit mutation had minimal effect on inhibin A bioactivity. Mutation of the corresponding residue in the inhibin ß B-subunit, G329E, similarly disrupted activin B synthesis/activity without affecting inhibin B production. Subsequently, we enhanced inhibin A potency by modifying the binding site for its co-receptor, betaglycan. Introducing a point mutation into the α-subunit (S344I) increased inhibin A potency ~12-fold. This study has identified a means to eliminate activin A/B interference during inhibin A/B production, and has facilitated the generation of potent inhibin A and inhibin B agonists for physiological exploration.


Asunto(s)
Inhibinas , Ingeniería de Proteínas/métodos , Femenino , Células HEK293 , Humanos , Inhibinas/genética , Inhibinas/aislamiento & purificación , Inhibinas/metabolismo , Inhibinas/farmacología , Proteínas de la Membrana , Modelos Moleculares , Mutagénesis/fisiología , Ovario/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/aislamiento & purificación , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , Multimerización de Proteína/genética , Estructura Cuaternaria de Proteína/genética , Estructura Terciaria de Proteína/genética , Subunidades de Proteína/genética , Subunidades de Proteína/aislamiento & purificación , Subunidades de Proteína/metabolismo , Subunidades de Proteína/farmacología , Proteínas de Saccharomyces cerevisiae , Transfección
9.
ACS Chem Biol ; 14(9): 1990-1997, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31407891

RESUMEN

Here, we report the design, synthesis, and evaluation of a series of inhibitors of Staphylococcus aureus BPL (SaBPL), where the central acyl phosphate of the natural intermediate biotinyl-5'-AMP (1) is replaced by a sulfonamide isostere. Acylsulfamide (6) and amino sulfonylurea (7) showed potent in vitro inhibitory activity (Ki = 0.007 ± 0.003 and 0.065 ± 0.03 µM, respectively) and antibacterial activity against S. aureus ATCC49775 with minimum inhibitory concentrations of 0.25 and 4 µg/mL, respectively. Additionally, the bimolecular interactions between the BPL and inhibitors 6 and 7 were defined by X-ray crystallography and molecular dynamics simulations. The high acidity of the sulfonamide linkers of 6 and 7 likely contributes to the enhanced in vitro inhibitory activities by promoting interaction with SaBPL Lys187. Analogues with alkylsulfamide (8), ß-ketosulfonamide (9), and ß-hydroxysulfonamide (10) isosteres were devoid of significant activity. Binding free energy estimation using computational methods suggests deprotonated 6 and 7 to be the best binders, which is consistent with enzyme assay results. Compound 6 was unstable in whole blood, leading to poor pharmacokinetics. Importantly, 7 has a vastly improved pharmacokinetic profile compared to that of 6 presumably due to the enhanced metabolic stability of the sulfonamide linker moiety.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Ligasas de Carbono-Nitrógeno/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Sulfonamidas/farmacología , Animales , Antibacterianos/síntesis química , Antibacterianos/farmacocinética , Proteínas Bacterianas/química , Ligasas de Carbono-Nitrógeno/química , Cristalografía por Rayos X , Diseño de Fármacos , Estabilidad de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Ratones , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Ratas , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/enzimología , Sulfonamidas/síntesis química , Sulfonamidas/farmacocinética
10.
Nat Commun ; 9(1): 2223, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29884801

RESUMEN

Macrophage migration inhibitory factor (MIF) exerts multiple effects on immune cells, as well as having functions outside the immune system. MIF can promote inflammation through the induction of other cytokines, including TNF, IL-6, and IL-1 family cytokines. Here, we show that inhibition of MIF regulates the release of IL-1α, IL-1ß, and IL-18, not by affecting transcription or translation of these cytokines, but via activation of the NLRP3 inflammasome. MIF is required for the interaction between NLRP3 and the intermediate filament protein vimentin, which is critical for NLRP3 activation. Further, we demonstrate that MIF interacts with NLRP3, indicating a role for MIF in inflammasome activation independent of its role as a cytokine. These data advance our understanding of how MIF regulates inflammation and identify it as a factor critical for NLRP3 inflammasome activation.


Asunto(s)
Inflamasomas/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Células THP-1
11.
Nucleic Acids Res ; 46(6): 3169-3186, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29346611

RESUMEN

RIG-I (retinoic acid inducible gene-I) is a cytosolic innate immune protein that senses viral dsRNA with a 5'-triphosphate overhang. Upon interaction with dsRNA a de-repression of the RIG-I CARD domains takes place that ultimately leads to the production of type I interferons and pro-inflammatory cytokines. Here we investigate the RIG-I conformational rearrangement upon interaction with an activating 5'-triphosphate-10-base pair dsRNA hairpin loop (10bp) compared with a less active 5'-triphosphate-8-base pair dsRNA hairpin loop (8bp). We use size-exclusion chromatography-coupled small-angle X-ray scattering (SAXS) and limited tryptic digest experiments to show that that upon binding to 10 bp, but not 8 bp, RIG-I becomes extended and shows greater flexibility, reflecting the release of its CARDs. We also examined the effect of different ATP analogues on the conformational changes of RIG-I/dsRNA complexes. Of the analogues tested, the addition of ATP transition state analogue ADP-AlFx further assisted in the complete activation of RIG-I in complex with 10bp and also to some extent RIG-I bound to 8bp. Together these data provide solution-based evidence for the molecular mechanism of innate immune signaling by RIG-I as stimulated by short hairpin RNA and ATP.


Asunto(s)
Adenosina Trifosfato/química , Proteína 58 DEAD Box/química , Dominios Proteicos , ARN Bicatenario/química , Adenosina Trifosfato/metabolismo , Secuencia de Bases , Cromatografía en Gel , Proteína 58 DEAD Box/metabolismo , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Receptores Inmunológicos , Dispersión del Ángulo Pequeño , Transducción de Señal , Soluciones/química , Difracción de Rayos X
12.
ACS Infect Dis ; 4(2): 175-184, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29131575

RESUMEN

We report the synthesis and evaluation of 5-halogenated-1,2,3-triazoles as inhibitors of biotin protein ligase from Staphylococcus aureus. The halogenated compounds exhibit significantly improved antibacterial activity over their nonhalogenated counterparts. Importantly, the 5-fluoro-1,2,3-triazole compound 4c displays antibacterial activity against S. aureus ATCC49775 with a minimum inhibitory concentration (MIC) of 8 µg/mL.


Asunto(s)
Antibacterianos/farmacología , Inhibidores Enzimáticos/farmacología , Ligasas/antagonistas & inhibidores , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/enzimología , Antibacterianos/química , Sitios de Unión , Biotina/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Halogenación , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Unión Proteica
13.
J Med Chem ; 60(22): 9349-9359, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29083893

RESUMEN

Grb7 is a signaling protein with critical roles in tumor cell proliferation and migration and an established cancer therapeutic target. Here we explore chemical space to develop a new bicyclic peptide inhibitor, incorporating thioether and lactam linkers that binds with affinity (KD = 1.1 µM) and specificity to the Grb7-SH2 domain. Structural analysis of the Grb7-SH2/peptide complex revealed an unexpected binding orientation underlying the binding selectivity by this new scaffold. We further incorporated carboxymethylphenylalanine and carboxyphenylalanine phosphotyrosine mimetics and arrived at an optimized inhibitor that potently binds Grb7-SH2 (KD = 0.13 µM) under physiological conditions. X-ray crystal structures of these Grb7-SH2/peptide complexes reveal the structural basis for the most potent and specific inhibitors of Grb7 developed to date. Finally, we demonstrate that cell permeable versions of these peptides successfully block Grb7 mediated interactions in a breast cancer cell line, establishing the potential of these peptides in the development of novel therapeutics targeted to Grb7.


Asunto(s)
Proteína Adaptadora GRB7/antagonistas & inhibidores , Péptidos Cíclicos/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Descubrimiento de Drogas , Quinasa 1 de Adhesión Focal/metabolismo , Proteína Adaptadora GRB7/química , Proteína Adaptadora GRB7/metabolismo , Humanos , Lactamas/síntesis química , Lactamas/química , Lactamas/farmacología , Ligandos , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/química , Fosfatos/química , Conformación Proteica , Receptor ErbB-2/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Dominios Homologos src
14.
Nucleic Acids Res ; 45(8): 4944-4957, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28184449

RESUMEN

TIA-1 (T-cell restricted intracellular antigen-1) is an RNA-binding protein involved in splicing and translational repression. It mainly interacts with RNA via its second and third RNA recognition motifs (RRMs), with specificity for U-rich sequences directed by RRM2. It has recently been shown that RRM3 also contributes to binding, with preferential binding for C-rich sequences. Here we designed UC-rich and CU-rich 10-nt sequences for engagement of both RRM2 and RRM3 and demonstrated that the TIA-1 RRM23 construct preferentially binds the UC-rich RNA ligand (5΄-UUUUUACUCC-3΄). Interestingly, this binding depends on the presence of Lys274 that is C-terminal to RRM3 and binding to equivalent DNA sequences occurs with similar affinity. Small-angle X-ray scattering was used to demonstrate that, upon complex formation with target RNA or DNA, TIA-1 RRM23 adopts a compact structure, showing that both RRMs engage with the target 10-nt sequences to form the complex. We also report the crystal structure of TIA-1 RRM2 in complex with DNA to 2.3 Šresolution providing the first atomic resolution structure of any TIA protein RRM in complex with oligonucleotide. Together our data support a specific mode of TIA-1 RRM23 interaction with target oligonucleotides consistent with the role of TIA-1 in binding RNA to regulate gene expression.


Asunto(s)
Proteínas de Unión al ADN/química , ADN/química , Proteínas de Unión a Poli(A)/química , Ribonucleósido Difosfato Reductasa/química , Cristalografía por Rayos X , ADN/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Humanos , Oligonucleótidos/química , Proteínas de Unión a Poli(A)/genética , Unión Proteica/genética , Mapas de Interacción de Proteínas/genética , Motivo de Reconocimiento de ARN/genética , Ribonucleósido Difosfato Reductasa/genética , Antígeno Intracelular 1 de las Células T
15.
Sci Rep ; 6: 27060, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27257138

RESUMEN

The design of potent and specific peptide inhibitors to therapeutic targets is of enormous utility for both proof-of-concept studies and for the development of potential new therapeutics. Grb7 is a key signaling molecule in the progression of HER2 positive and triple negative breast cancers. Here we report the crystal structure of a stapled bicyclic peptide inhibitor G7-B1 in complex with the Grb7-SH2 domain. This revealed an unexpected binding mode of the peptide, in which the staple forms an alternative contact with the surface of the target protein. Based on this structural information, we designed a new series of bicyclic G7 peptides that progressively constrain the starting peptide, to arrive at the G7-B4 peptide that binds with an approximately 2-fold enhanced affinity to the Grb7-SH2 domain (KD = 0.83 µM) compared to G7-B1 and shows low affinity binding to Grb2-, Grb10- and Grb14-SH2 domains (KD > 100 µM). Furthermore, we determined the structure of the G7-B4 bicyclic peptide in complex with the Grb7-SH2 domain, both before and after ring closing metathesis to show that the closed staple is essential to the target interaction. The G7-B4 peptide represents an advance in the development of Grb7 inhibitors and is a classical example of structure aided inhibitor development.


Asunto(s)
Proteína Adaptadora GRB7/química , Péptidos Cíclicos/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Diseño de Fármacos , Proteína Adaptadora GRB7/antagonistas & inhibidores , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína
16.
J Med Chem ; 58(19): 7707-18, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26359549

RESUMEN

The Grb7 adaptor protein is a therapeutic target for both TNBC and HER2+ breast cancer. A nonphosphorylated cyclic peptide 1 (known as G7-18NATE) inhibits Grb7 via targeting the Grb7-SH2 domain, but requires the presence of phosphate ions for both affinity and specificity. Here we report the discovery of malonate bound in the phosphotyrosine binding pocket of the apo-Grb7-SH2 structure. Based on this, we carried out the rational design and synthesis of two analogues of peptide 1 that incorporate carboxymethylphenylalanine (cmF) and carboxyphenylalanine (cF) as mimics of phosphotyrosine (pY). Binding studies using SPR confirmed that affinity for Grb7-SH2 domain is improved up to 9-fold over peptide 1 under physiological phosphate conditions (KD = 2.1-5.7 µM) and that binding is specific for Grb7-SH2 over closely related domains (low or no detectable binding to Grb2-SH2 and Grb10-SH2). X-ray crystallographic structural analysis of the analogue bearing a cmF moiety in complex with Grb7-SH2 has identified the precise contacts conferred by the pY mimic that underpin this improved molecular interaction. Together this study identifies and characterizes the tightest specific inhibitor of Grb7 to date, representing a significant development toward a new Grb7-targeted therapeutic.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Proteína Adaptadora GRB7/antagonistas & inhibidores , Péptidos Cíclicos/química , Fosfotirosina/química , Antineoplásicos/síntesis química , Sitios de Unión , Neoplasias de la Mama/tratamiento farmacológico , Cristalografía por Rayos X , Femenino , Proteína Adaptadora GRB7/metabolismo , Humanos , Malonatos/química , Terapia Molecular Dirigida , Péptidos Cíclicos/síntesis química , Peptidomiméticos , Fosfatos/química , Fosfatos/metabolismo , Conformación Proteica , Dominios Homologos src
17.
Sci Rep ; 5: 12905, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26243377

RESUMEN

The biogenesis of membranes from constituent proteins and lipids is a fundamental aspect of cell biology. In the case of proteins assembled into bacterial outer membranes, an overarching question concerns how the energy required for protein insertion and folding is accessed at this remote location of the cell. The translocation and assembly module (TAM) is a nanomachine that functions in outer membrane biogenesis and virulence in diverse bacterial pathogens. Here we demonstrate the interactions through which TamA and TamB subunits dock to bridge the periplasm, and unite the outer membrane aspects to the inner membrane of the bacterial cell. We show that specific functional features in TamA have been conserved through evolution, including residues surrounding the lateral gate and an extensive surface of the POTRA domains. Analysis by nuclear magnetic resonance spectroscopy and small angle X-ray scattering document the characteristic structural features of these POTRA domains and demonstrate rigidity in solution. Quartz crystal microbalance measurements pinpoint which POTRA domain specifically docks the TamB subunit of the nanomachine. We speculate that the POTRA domain of TamA functions as a lever arm in order to drive the activity of the TAM, assembling proteins into bacterial outer membranes.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/fisiología , Secuencia Conservada , Evolución Molecular , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína
18.
Cell Cycle ; 14(17): 2729-33, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26177105

RESUMEN

Both RNA-binding proteins (RBP) and miRNA play important roles in the regulation of mRNA expression, often acting together to regulate a target mRNA. In some cases the RBP and miRNA have been reported to act competitively, but in other instances they function cooperatively. Here, we investigated HuR function as an enhancer of let-7-mediated translational repression of c-Myc despite the separation of their binding sites. Using an in vitro system, we determined that a let-7 mimic, consisting of single-stranded (ss)DNA complementary to the let-7 binding site, enhanced the affinity of HuR for a 122-nt MYC RNA encompassing both binding sites. This finding supports the biophysical principle of cooperative binding by an RBP and miRNA purely through interactions at distal mRNA binding sites.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , MicroARNs/metabolismo , Imitación Molecular/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína 1 Similar a ELAV/química , Humanos , MicroARNs/química , Unión Proteica/fisiología , Proteínas Proto-Oncogénicas c-myc/química
19.
PLoS One ; 10(4): e0123338, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25923520

RESUMEN

Phanta is a reversibly photoswitching chromoprotein (ΦF, 0.003), useful for pcFRET, that was isolated from a mutagenesis screen of the bright green fluorescent eCGP123 (ΦF, 0.8). We have investigated the contribution of substitutions at positions His193, Thr69 and Gln62, individually and in combination, to the optical properties of Phanta. Single amino acid substitutions at position 193 resulted in proteins with very low ΦF, indicating the importance of this position in controlling the fluorescence efficiency of the variant proteins. The substitution Thr69Val in Phanta was important for supressing the formation of a protonated chromophore species observed in some His193 substituted variants, whereas the substitution Gln62Met did not significantly contribute to the useful optical properties of Phanta. X-ray crystal structures for Phanta (2.3 Å), eCGP123T69V (2.0 Å) and eCGP123H193Q (2.2 Å) in their non-photoswitched state were determined, revealing the presence of a cis-coplanar chromophore. We conclude that changes in the hydrogen-bonding network supporting the cis-chromophore, and its contacts with the surrounding protein matrix, are responsible for the low fluorescence emission of eCGP123 variants containing a His193 substitution.


Asunto(s)
Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Sustitución de Aminoácidos , Cristalografía por Rayos X , Escherichia coli/genética , Fluorescencia , Colorantes Fluorescentes/metabolismo , Enlace de Hidrógeno , Modelos Moleculares , Procesos Fotoquímicos , Conformación Proteica
20.
Tuberculosis (Edinb) ; 95(3): 259-66, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25801336

RESUMEN

Dethiobiotin synthetase (DTBS) plays a crucial role in biotin biosynthesis in microorganisms, fungi, and plants. Due to its importance in bacterial pathogenesis, and the absence of a human homologue, DTBS is a promising target for the development of new antibacterials desperately needed to combat antibiotic resistance. Here we report the first X-ray structure of DTBS from Mycobacterium tuberculosis (MtDTBS) bound to a nucleotide triphosphate (CTP). The nucleoside base is stabilized in its pocket through hydrogen-bonding interactions with the protein backbone, rather than amino acid side chains. This resulted in the unexpected finding that MtDTBS could utilise ATP, CTP, GTP, ITP, TTP, or UTP with similar Km and kcat values, although the enzyme had the highest affinity for CTP in competitive binding and surface plasmon resonance assays. This is in contrast to other DTBS homologues that preferentially bind ATP primarily through hydrogen-bonds between the purine base and the carboxamide side chain of a key asparagine. Mutational analysis performed alongside in silico experiments revealed a gate-keeper role for Asn175 in Escherichia coli DTBS that excludes binding of other nucleotide triphosphates. Here we provide evidence to show that MtDTBS has a broad nucleotide specificity due to the absence of the gate-keeper residue.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ligasas de Carbono-Nitrógeno/metabolismo , Mycobacterium tuberculosis/enzimología , Nucleótidos/metabolismo , Proteínas Bacterianas/química , Sitios de Unión , Ligasas de Carbono-Nitrógeno/química , Ligasas de Carbono-Nitrógeno/genética , Dominio Catalítico , Simulación por Computador , Cristalografía por Rayos X , Escherichia coli/enzimología , Escherichia coli/genética , Enlace de Hidrógeno , Cinética , Mutagénesis Sitio-Dirigida , Mutación , Mycobacterium tuberculosis/genética , Conformación Proteica , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA