Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
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
2.
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
3.
Arch Biochem Biophys ; 687: 108386, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32360748

RESUMEN

Growth factor receptor bound protein 7 (Grb7) is a mammalian adaptor protein participating in signaling pathways implicated in cell migration, metastatic invasion, cell proliferation and tumor-associated angiogenesis. We expressed tagged versions of wild type Grb7 and the mutant Grb7Δ, lacking its calmodulin-binding domain (CaM-BD), in human embryonic kidney (HEK) 293 cells and rat glioma C6 cells to identify novel binding partners using shot-gun proteomics. Among the new identified proteins, we validated the ubiquitin-ligase Nedd4 (neural precursor cell expressed developmentally down-regulated protein 4), the heat-shock protein Hsc70/HSPA8 (heat shock cognate protein 70) and the cell cycle regulatory protein caprin-1 (cytoplasmic activation/proliferation-associated protein 1) in rat glioma C6 cells. Our results suggest a role of Grb7 in pathways where these proteins are implicated. These include protein trafficking and degradation, stress-response, chaperone-mediated autophagy, apoptosis and cell proliferation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteína Adaptadora GRB7/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Animales , Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Línea Celular Tumoral , Proteína Adaptadora GRB7/genética , Células HEK293 , Humanos , Mutación , Unión Proteica , Dominios Proteicos/genética , Estructura Secundaria de Proteína , Proteómica , Ratas
4.
Protein Expr Purif ; 176: 105722, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32768454

RESUMEN

Heat Shock Factor 1 (HSF1) is the master regulator of the heat shock response, a universal survival mechanism throughout eukaryotic species used to buffer potentially lethal proteotoxic conditions. HSF1's function in vivo is regulated by several factors, including post translational modifications and elevated temperatures, whereupon it forms trimers to bind with heat shock elements in DNA. Unsurprisingly, HSF1 is also extremely sensitive to elevated temperatures in vitro, which poses specific technical challenges when producing HSF1 using a recombinant expression system. Although there are several useful publications which outline steps taken for HSF1 expression and purification, studies that describe specific strategies and detailed protocols to overcome HSF1 trimerisation and degradation are currently lacking. Herein, we have reported our detailed experimental protocol for the expression and purification of monomeric human HSF1 (HsHSF1) as a major species. We also propose a refined method of inducing HsHSF1 activation in vitro, that we consider more accurately mimics HsHSF1 activation in vivo and is therefore more physiologically relevant.


Asunto(s)
Expresión Génica , Factores de Transcripción del Choque Térmico , Factores de Transcripción del Choque Térmico/biosíntesis , Factores de Transcripción del Choque Térmico/química , Factores de Transcripción del Choque Térmico/genética , Factores de Transcripción del Choque Térmico/aislamiento & purificación , Humanos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
5.
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
6.
Int J Mol Sci ; 21(4)2020 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-32079204

RESUMEN

Grb7 is a signalling adapter protein that engages activated receptor tyrosine kinases at cellular membranes to effect downstream pathways of cell migration, proliferation and survival. Grb7's cellular location was shown to be regulated by the small calcium binding protein calmodulin (CaM). While evidence for a Grb7/CaM interaction is compelling, a direct interaction between CaM and purified Grb7 has not been demonstrated and quantitated. In this study we sought to determine this, and prepared pure full-length Grb7, as well as its RA-PH and SH2 subdomains, and tested for CaM binding using surface plasmon resonance. We report a direct interaction between full-length Grb7 and CaM that occurs in a calcium dependent manner. While no binding was observed to the SH2 domain alone, we observed a high micromolar affinity interaction between the Grb7 RA-PH domain and CaM, suggesting that the Grb7/CaM interaction is mediated through this region of Grb7. Together, our data support the model of a CaM interaction with Grb7 via its RA-PH domain.


Asunto(s)
Calmodulina/genética , Proteína Adaptadora GRB7/genética , Dominios Homólogos a Pleckstrina/genética , Calmodulina/metabolismo , Movimiento Celular , Proliferación Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteína Adaptadora GRB7/metabolismo , Unión Proteica , Conformación Proteica , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Resonancia por Plasmón de Superficie , Dominios Homologos src/genética
7.
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
8.
Molecules ; 24(20)2019 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-31627265

RESUMEN

Grb7 is an adapter protein, overexpressed in HER2+ve breast and other cancers, and identified as a therapeutic target. Grb7 promotes both proliferative and migratory cellular pathways through interaction of its SH2 domain with upstream binding partners including HER2, SHC, and FAK. Here we present the evaluation of a series of monocyclic and bicyclic peptide inhibitors that have been developed to specifically and potently target the Grb7 SH2-domain. All peptides tested were found to inhibit signaling in both ERK and AKT pathways in SKBR-3 and MDA-MB-231 cell lines. Proliferation, migration, and invasion assays revealed, however, that the second-generation bicyclic peptides were not more bioactive than the first generation G7-18NATE peptide, despite their higher in vitro affinity for the target. This was found not to be due to steric hindrance by the cell-permeability tag, as ascertained by ITC, but to differences in the ability of the bicyclic peptides to interact with and penetrate cellular membranes, as determined using SPR and mass spectrometry. These studies reveal that just small differences to amino acid composition can greatly impact the effectiveness of peptide inhibitors to their intracellular target and demonstrate that G7-18NATE remains the most effective peptide inhibitor of Grb7 developed to date.


Asunto(s)
Antineoplásicos/farmacología , Células Epiteliales/efectos de los fármacos , Proteína Adaptadora GRB7/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Péptidos Cíclicos/farmacología , Transducción de Señal/efectos de los fármacos , Secuencia de Aminoácidos , Antineoplásicos/síntesis química , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteína Adaptadora GRB7/genética , Proteína Adaptadora GRB7/metabolismo , Humanos , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Péptidos Cíclicos/síntesis química , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Relación Estructura-Actividad , Dominios Homologos src/efectos de los fármacos
9.
Biochim Biophys Acta ; 1858(8): 1841-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27163492

RESUMEN

We have studied the effect of penetratin and a truncated analogue on the bilayer structure using dual polarisation interferometry, to simultaneously measure changes in mass per unit area and birefringence (an optical parameter representing bilayer order) with high sensitivity during the binding and dissociation from the membrane. Specifically, we studied penetratin (RQIKIWFQNRRMKWKK), along with a shortened and biotinylated version known as R8K-biotin (RRMKWKKK(Biotin)-NH2). Overall both peptides bound only weakly to the neutral DMPC and POPC bilayers, while much higher binding was observed for the anionic DMPC/DMPG and POPC/POPG. The binding of penetratin to gel-phase DMPC/DMPG was adequately represented by a two-state model, whereas on the fluid-phase POPC/POPG it exhibited a distinctly different binding pattern, best represented by a three-state kinetic model. However, R8K-biotin did not bind well to DMPC/DMPG and showed a more transitory and superficial binding to POPC/POPG. Comparing the modelling results for both peptides binding to POPC/POPG suggests an important role for a securely bound intermediate prior to penetratin insertion and translocation. Overall these results further elucidate the mechanism of penetratin, and provide another example of the significance of the ability of DPI to measure structural changes and the use of kinetic analysis to investigate the stages of peptide-membrane interactions.


Asunto(s)
Péptidos de Penetración Celular/farmacología , Membrana Dobles de Lípidos/química , Secuencia de Aminoácidos , Biotinilación , Birrefringencia , Proteínas Portadoras/farmacología , Dimiristoilfosfatidilcolina/química , Geles , Interferometría , Cinética , Liposomas/química , Lípidos de la Membrana/química , Modelos Químicos , Fragmentos de Péptidos/farmacología , Fosfatidilcolinas/química , Fosfatidilgliceroles/química , Unión Proteica , Relación Estructura-Actividad
10.
PLoS Genet ; 9(7): e1003628, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935508

RESUMEN

Alternative splicing of precursor messenger RNA (pre-mRNA) is common in mammalian cells and enables the production of multiple gene products from a single gene, thus increasing transcriptome and proteome diversity. Disturbance of splicing regulation is associated with many human diseases; however, key splicing factors that control tissue-specific alternative splicing remain largely undefined. In an unbiased genetic screen for essential male fertility genes in the mouse, we identified the RNA binding protein RBM5 (RNA binding motif 5) as an essential regulator of haploid male germ cell pre-mRNA splicing and fertility. Mice carrying a missense mutation (R263P) in the second RNA recognition motif (RRM) of RBM5 exhibited spermatid differentiation arrest, germ cell sloughing and apoptosis, which ultimately led to azoospermia (no sperm in the ejaculate) and male sterility. Molecular modelling suggested that the R263P mutation resulted in compromised mRNA binding. Within the adult mouse testis, RBM5 localises to somatic and germ cells including spermatogonia, spermatocytes and round spermatids. Through the use of RNA pull down coupled with microarrays, we identified 11 round spermatid-expressed mRNAs as putative RBM5 targets. Importantly, the R263P mutation affected pre-mRNA splicing and resulted in a shift in the isoform ratios, or the production of novel spliced transcripts, of most targets. Microarray analysis of isolated round spermatids suggests that altered splicing of RBM5 target pre-mRNAs affected expression of genes in several pathways, including those implicated in germ cell adhesion, spermatid head shaping, and acrosome and tail formation. In summary, our findings reveal a critical role for RBM5 as a pre-mRNA splicing regulator in round spermatids and male fertility. Our findings also suggest that the second RRM of RBM5 is pivotal for appropriate pre-mRNA splicing.


Asunto(s)
Empalme Alternativo/genética , Diferenciación Celular/genética , Infertilidad Masculina/genética , Motivos de Nucleótidos/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células Germinativas/patología , Humanos , Infertilidad Masculina/patología , Masculino , Ratones , Modelos Moleculares , Mutación , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Espermátides/metabolismo , Espermátides/patología , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
11.
Nucleic Acids Res ; 41(5): 3436-45, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23325848

RESUMEN

The retinoic acid inducible gene-I (RIG-I)-like family of receptors is positioned at the front line of our innate cellular defence system. RIG-I detects and binds to foreign duplex RNA in the cytoplasm of both immune and non-immune cells, and initiates the induction of type I interferons and pro-inflammatory cytokines. The mechanism of RIG-I activation by double-stranded RNA (dsRNA) involves a molecular rearrangement proposed to expose the N-terminal pair of caspase activation recruitment domains, enabling an interaction with interferon-beta promoter stimulator 1 (IPS-1) and thereby initiating downstream signalling. dsRNA is particularly stimulatory when longer than 20 bp, potentially through allowing binding of more than one RIG-I molecule. Here, we characterize full-length RIG-I and RIG-I subdomains combined with a stimulatory 29mer dsRNA using multi-angle light scattering and size-exclusion chromatography-coupled small-angle X-ray scattering, to build up a molecular model of RIG-I before and after the formation of a 2:1 protein:dsRNA assembly. We report the small-angle X-ray scattering-derived solution structure of the human apo-RIG-I and observe that on binding of RIG-I to dsRNA in a 2:1 ratio, the complex becomes highly extended and flexible. Hence, here we present the first model of the fully activated oligomeric RIG-I.


Asunto(s)
Apoproteínas/química , ARN Helicasas DEAD-box/química , ARN Bicatenario/química , Cromatografía en Gel , Proteína 58 DEAD Box , Humanos , Modelos Moleculares , Fragmentos de Péptidos/química , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteolisis , Receptores Inmunológicos , Dispersión del Ángulo Pequeño , Tripsina/química , Difracción de Rayos X
12.
J Biol Chem ; 288(22): 15821-9, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23592783

RESUMEN

The complement system is an ancient innate immune defense pathway that plays a front line role in eliminating microbial pathogens. Recognition of foreign targets by antibodies drives sequential activation of two serine proteases, C1r and C1s, which reside within the complement Component 1 (C1) complex. Active C1s propagates the immune response through its ability to bind and cleave the effector molecule complement Component 4 (C4). Currently, the precise structural and biochemical basis for the control of the interaction between C1s and C4 is unclear. Here, using surface plasmon resonance, we show that the transition of the C1s zymogen to the active form is essential for C1s binding to C4. To understand this, we determined the crystal structure of a zymogen C1s construct (comprising two complement control protein (CCP) domains and the serine protease (SP) domain). These data reveal that two loops (492-499 and 573-580) in the zymogen serine protease domain adopt a conformation that would be predicted to sterically abrogate C4 binding. The transition from zymogen to active C1s repositions both loops such that they would be able to interact with sulfotyrosine residues on C4. The structure also shows the junction of the CCP1 and CCP2 domains of C1s for the first time, yielding valuable information about the exosite for C4 binding located at this position. Together, these data provide a structural explanation for the control of the interaction with C1s and C4 and, furthermore, point to alternative strategies for developing therapeutic approaches for controlling activation of the complement cascade.


Asunto(s)
Complemento C1s/química , Complemento C4/química , Precursores Enzimáticos/química , Complemento C1s/genética , Complemento C1s/metabolismo , Complemento C4/genética , Complemento C4/metabolismo , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Humanos , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad
13.
Breast Cancer Res Treat ; 143(3): 593-603, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24464577

RESUMEN

Grb7 is an adapter protein, aberrantly co-overexpressed with HER2 and identified as an independent prognostic marker in breast cancer. It has been established that Grb7 exacerbates the cellular growth and migratory behaviour of HER2+ve breast cancer cells. Less is known about Grb7's role in the context of HER2-ve cells. Here we directly compare the effect of stable Grb7 knockdown in oestrogen sensitive (T47D), HER2+ve (SKBR3) and triple-negative (MDA-MB-468 and MDA-MB-231) breast cancer cell lines on anchorage dependent and independent cell growth, wound healing and chemotaxis. All cell lines showed reduced ability to migrate upon Grb7 knockdown, despite their greatly varied endogenous levels of Grb7. Decreased cell proliferation was not observed in any of the cell lines upon Grb7 knockdown; however, decreased ability to form colonies was observed for all but the oestrogen sensitive cell line, depending upon the stringency of the growth conditions. The data reveal that Grb7 plays an important role in breast cancer progression, beyond the context of HER2+ve cell types.


Asunto(s)
Proteína Adaptadora GRB7/genética , Neoplasias de la Mama Triple Negativas/genética , Proliferación Celular , Estrógenos/genética , Estrógenos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Receptor ErbB-2/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/patología
14.
RNA Biol ; 11(6): 766-76, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24824036

RESUMEN

T-cell intracellular antigen-1 (TIA-1) is a key DNA/RNA binding protein that regulates translation by sequestering target mRNAs in stress granules (SG) in response to stress conditions. TIA-1 possesses three RNA recognition motifs (RRM) along with a glutamine-rich domain, with the central domains (RRM2 and RRM3) acting as RNA binding platforms. While the RRM2 domain, which displays high affinity for U-rich RNA sequences, is primarily responsible for interaction with RNA, the contribution of RRM3 to bind RNA as well as the target RNA sequences that it binds preferentially are still unknown. Here we combined nuclear magnetic resonance (NMR) and surface plasmon resonance (SPR) techniques to elucidate the sequence specificity of TIA-1 RRM3. With a novel approach using saturation transfer difference NMR (STD-NMR) to quantify protein-nucleic acids interactions, we demonstrate that isolated RRM3 binds to both C- and U-rich stretches with micromolar affinity. In combination with RRM2 and in the context of full-length TIA-1, RRM3 significantly enhanced the binding to RNA, particularly to cytosine-rich RNA oligos, as assessed by biotinylated RNA pull-down analysis. Our findings provide new insight into the role of RRM3 in regulating TIA-1 binding to C-rich stretches, that are abundant at the 5' TOPs (5' terminal oligopyrimidine tracts) of mRNAs whose translation is repressed under stress situations.


Asunto(s)
Motivos de Nucleótidos , Proteínas de Unión a Poli(A)/química , Proteínas de Unión a Poli(A)/metabolismo , Dominios y Motivos de Interacción de Proteínas , ARN/química , ARN/genética , Secuencia de Bases , Sitios de Unión , Secuencia Rica en GC , Humanos , Resonancia Magnética Nuclear Biomolecular , Posición Específica de Matrices de Puntuación
15.
Nucleic Acids Res ; 40(11): 5101-14, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22344691

RESUMEN

Poly-C-binding proteins are triple KH (hnRNP K homology) domain proteins with specificity for single stranded C-rich RNA and DNA. They play diverse roles in the regulation of protein expression at both transcriptional and translational levels. Here, we analyse the contributions of individual αCP1 KH domains to binding C-rich oligonucleotides using biophysical and structural methods. Using surface plasmon resonance (SPR), we demonstrate that KH1 makes the most stable interactions with both RNA and DNA, KH3 binds with intermediate affinity and KH2 only interacts detectibly with DNA. The crystal structure of KH1 bound to a 5'-CCCTCCCT-3' DNA sequence shows a 2:1 protein:DNA stoichiometry and demonstrates a molecular arrangement of KH domains bound to immediately adjacent oligonucleotide target sites. SPR experiments, with a series of poly-C-sequences reveals that cytosine is preferred at all four positions in the oligonucleotide binding cleft and that a C-tetrad binds KH1 with 10 times higher affinity than a C-triplet. The basis for this high affinity interaction is finally detailed with the structure determination of a KH1.W.C54S mutant bound to 5'-ACCCCA-3' DNA sequence. Together, these data establish the lead role of KH1 in oligonucleotide binding by αCP1 and reveal the molecular basis of its specificity for a C-rich tetrad.


Asunto(s)
Citosina/química , Ribonucleoproteínas Nucleares Heterogéneas/química , Oligonucleótidos/química , Sitios de Unión , ADN/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/química , ARN Mensajero/metabolismo
16.
Biopolymers ; 100(5): 543-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23505041

RESUMEN

Grb7 is an adapter protein involved in the propagation of signals in cancer cell migration and proliferation, and is thus a target for the development of novel anti-cancer agents. An 11-residue thioether-cyclized peptide known as G7-18NATE has previously been developed, that inhibits Grb7 via specific interactions with its SH2 domain with micromolar affinity. Here we explore whether the peptide binding is enhanced by the addition of a second linkage designed to restrain the peptide in its bound conformation and thus reduce the entropic loss upon binding. The use of an O-ally ser covalent linkage between residue positions 1 and 8 successfully enhanced the affinity, and ITC showed that the entropic loss was reduced. A peptide with thioether-cyclization exchanged for an amide linkage showed reduce affinity, though the formation of a disulfide bond between positions 1 and 8 in this peptide enhanced its binding. This study paves the way for improving the G7-18NATE scaffold for second generation inhibitors of Grb7.


Asunto(s)
Proteína Adaptadora GRB7 , Dominios Homologos src , Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Proteína Adaptadora GRB7/metabolismo , Humanos , Péptidos/farmacología , Unión Proteica
17.
RNA Biol ; 10(4): 579-89, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23603827

RESUMEN

The RNA-binding protein TIAR is an mRNA-binding protein that acts as a translational repressor, particularly important under conditions of cellular stress. It binds to target mRNA and DNA via its RNA recognition motif (RRM) domains and is involved in both splicing regulation and translational repression via the formation of "stress granules." TIAR has also been shown to bind ssDNA and play a role in the regulation of transcription. Here we show, using surface plasmon resonance and nuclear magnetic resonance spectroscopy, specific roles of individual TIAR domains for high-affinity binding to RNA and DNA targets. We confirm that RRM2 of TIAR is the major RNA- and DNA-binding domain. However, the strong nanomolar affinity binding to U-rich RNA and T-rich DNA depends on the presence of the six amino acid residues found in the linker region C-terminal to RRM2. On its own, RRM1 shows preferred binding to DNA over RNA. We further characterize the interaction between RRM2 with the C-terminal extension and an AU-rich target RNA sequence using NMR spectroscopy to identify the amino acid residues involved in binding. We demonstrate that TIAR RRM2, together with its C-terminal extension, is the major contributor for the high-affinity (nM) interactions of TIAR with target RNA sequences.


Asunto(s)
Secuencias de Aminoácidos , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , ADN/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Unión Proteica , Empalme del ARN , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Alineación de Secuencia , Resonancia por Plasmón de Superficie
18.
Nucleic Acids Res ; 39(3): 1117-30, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21233170

RESUMEN

TIAR and HuR are mRNA-binding proteins that play important roles in the regulation of translation. They both possess three RNA recognition motifs (RRMs) and bind to AU-rich elements (AREs), with seemingly overlapping specificity. Here we show using SPR that TIAR and HuR bind to both U-rich and AU-rich RNA in the nanomolar range, with higher overall affinity for U-rich RNA. However, the higher affinity for U-rich sequences is mainly due to faster association with U-rich RNA, which we propose is a reflection of the higher probability of association. Differences between TIAR and HuR are observed in their modes of binding to RNA. TIAR is able to bind deoxy-oligonucleotides with nanomolar affinity, whereas HuR affinity is reduced to a micromolar level. Studies with U-rich DNA reveal that TIAR binding depends less on the 2'-hydroxyl group of RNA than HuR binding. Finally we show that SAXS data, recorded for the first two domains of TIAR in complex with RNA, are more consistent with a flexible, elongated shape and not the compact shape that the first two domains of Hu proteins adopt upon binding to RNA. We thus propose that these triple-RRM proteins, which compete for the same binding sites in cells, interact with their targets in fundamentally different ways.


Asunto(s)
Antígenos de Superficie/química , ADN/química , Proteínas de Unión al ARN/química , ARN/química , Adenina/análisis , Antígenos de Superficie/metabolismo , ADN/metabolismo , Proteínas ELAV , Proteína 1 Similar a ELAV , Cinética , Modelos Moleculares , Unión Proteica , Conformación Proteica , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Dispersión del Ángulo Pequeño , Uracilo/análisis , Difracción de Rayos X
19.
Proc Natl Acad Sci U S A ; 107(12): 5534-9, 2010 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-20212169

RESUMEN

Residues within processed protein fragments bound to major histocompatibility complex class I (MHC-I) glycoproteins have been considered to function as a series of "independent pegs" that either anchor the peptide (p) to the MHC-I and/or interact with the spectrum of alphabeta-T-cell receptors (TCRs) specific for the pMHC-I epitope in question. Mining of the extensive pMHC-I structural database established that many self- and viral peptides show extensive and direct interresidue interactions, an unexpected finding that has led us to the idea of "constrained" peptides. Mutational analysis of two constrained peptides (the HLA B44 restricted self-peptide (B44DPalpha-EEFGRAFSF) and an H2-D(b) restricted influenza peptide (D(b)PA, SSLENFRAYV) demonstrated that the conformation of the prominently exposed arginine in both peptides was governed by interactions with MHC-I-orientated flanking residues from the peptide itself. Using reverse genetics in a murine influenza model, we revealed that mutation of an MHC-I-orientated residue (SSLENFRAYV --> SSLENARAYV) within the constrained PA peptide resulted in a diminished cytotoxic T lymphocyte (CTL) response and the recruitment of a limited pMHC-I specific TCR repertoire. Interactions between individual peptide positions can thus impose fine control on the conformation of pMHC-I epitopes, whereas the perturbation of such constraints can lead to a previously unappreciated mechanism of viral escape.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Linfocitos T/inmunología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Presentación de Antígeno , Epítopos/química , Epítopos/genética , Epítopos/metabolismo , Femenino , Antígenos H-2/química , Antígenos H-2/genética , Antígenos H-2/metabolismo , Antígenos HLA-B/química , Antígenos HLA-B/genética , Antígenos HLA-B/metabolismo , Antígeno HLA-B44 , Antígeno de Histocompatibilidad H-2D , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Conformación Proteica , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/inmunología
20.
Methods Mol Biol ; 2705: 199-210, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37668975

RESUMEN

Biosensor measurement using surface plasmon resonance enables precise evaluation of peptide-protein interactions. It is a sensitive technique that provides kinetic and affinity data with very little sample and without the need for analyte labels. Here, we describe its application for the analysis of peptide interactions with the Grb7-SH2 domain prepared with a GST-tag for tethering to the chip surface. This has been successfully and reliably used for direct comparison of a range of peptides under different solution conditions as well as direct comparison of peptides flowed over different related SH2 domains in real time. We have used the BIAcore system and describe both the methodology for data collection and analysis, with principles also applicable to other biosensor platforms.


Asunto(s)
Resonancia por Plasmón de Superficie , Dominios Homologos src , Recolección de Datos , Cinética , Péptidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA