Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
PLoS Pathog ; 18(7): e1010733, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35849637

RESUMEN

Emerging SARS-CoV-2 variants are creating major challenges in the ongoing COVID-19 pandemic. Being able to predict mutations that could arise in SARS-CoV-2 leading to increased transmissibility or immune evasion would be extremely valuable in development of broad-acting therapeutics and vaccines, and prioritising viral monitoring and containment. Here we use in vitro evolution to seek mutations in SARS-CoV-2 receptor binding domain (RBD) that would substantially increase binding to ACE2. We find a double mutation, S477N and Q498H, that increases affinity of RBD for ACE2 by 6.5-fold. This affinity gain is largely driven by the Q498H mutation. We determine the structure of the mutant-RBD:ACE2 complex by cryo-electron microscopy to reveal the mechanism for increased affinity. Addition of Q498H to SARS-CoV-2 RBD variants is found to boost binding affinity of the variants for human ACE2 and confer a new ability to bind rat ACE2 with high affinity. Surprisingly however, in the presence of the common N501Y mutation, Q498H inhibits binding, due to a clash between H498 and Y501 side chains. To achieve an intermolecular bonding network, affinity gain and cross-species binding similar to Q498H alone, RBD variants with the N501Y mutation must acquire instead the related Q498R mutation. Thus, SARS-CoV-2 RBD can access large affinity gains and cross-species binding via two alternative mutational routes involving Q498, with route selection determined by whether a variant already has the N501Y mutation. These mutations are now appearing in emerging SARS-CoV-2 variants where they have the potential to influence human-to-human and cross-species transmission.


Asunto(s)
COVID-19 , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/genética , Animales , COVID-19/genética , Microscopía por Crioelectrón , Humanos , Mutación , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Unión Proteica , Ratas , Receptores Virales/metabolismo , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
2.
J Biol Chem ; 297(2): 100888, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34153320

RESUMEN

Angiopoietins Ang1 and Ang2 are secreted ligands for the endothelial receptor tyrosine kinase Tie2 essential for vascular development and maintenance. Ang1 acts as an agonist to maintain normal vessel function, whereas Ang2 acts as a Tie2 antagonist. Ang2 is increased in macular edema, sepsis, and other conditions, in which it blocks Ang1-mediated signaling, causing vascular dysfunction and contributing to disease pathology. Therefore, Ang2 is an attractive therapeutic target. Previously, we reported a Tie2 ectodomain variant that selectively binds Ang2 and acts as soluble ligand trap to sequester Ang2; however, the mechanism of Ang2-binding selectivity is unknown. In the present study, we used directed protein evolution to enhance Ang2-binding affinity of this Tie2 ectodomain trap. We examined contributions of individual residues in the ligand-binding interface of Tie2 to Ang1 and Ang2 binding. Surprisingly, different combinations of Tie2 residues were found to bind each ligand, with hydrophobic residues binding both ligands and polar residues contributing selectively to either Ang1 or Ang2 binding. Our analysis also identified a single Tie2 residue, His168, with a pivotal role in both Ang1 and Ang2 binding, enabling competition between binding ligands. In summary, this study reports an enhanced-affinity Ang2-selective ligand trap with potential for therapeutic development and reveals the mechanism behind its selectivity. It also provides the first analysis of contributions of individual Tie2 residues to Ang1 and Ang2 binding and identifies selectivity-determining residues that could be targeted in the future design of small molecule and other inhibitors of Ang2 for the treatment of vascular dysfunction.


Asunto(s)
Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Evolución Molecular Dirigida/métodos , Receptor TIE-2/metabolismo , Angiopoyetina 1/química , Angiopoyetina 2/química , Células Cultivadas , Humanos , Ligandos , Neovascularización Fisiológica , Unión Proteica , Receptor TIE-2/química , Transducción de Señal
3.
Biochemistry ; 57(18): 2611-2622, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29505720

RESUMEN

The paralogues TRPV5 and TRPV6 belong to the vanilloid subfamily of the transient receptor potential (TRP) superfamily of ion channels, and both play an important role in overall Ca2+ homeostasis. The functioning of the channels centers on a tightly controlled Ca2+-dependent feedback mechanism in which the direct binding of the universal Ca2+-binding protein calmodulin (CaM) to the channel's C-terminal tail is required for channel inactivation. We have investigated this interaction at the atomic level and propose that under basal cellular Ca2+ concentrations CaM is constitutively bound to the channel's C-tail via CaM C-lobe only contacts. When the cytosolic Ca2+ concentration increases charging the apo CaM N-lobe with Ca2+, the CaM:TRPV6 complex rearranges and the TRPV6 C-tail further engages the CaM N-lobe via a crucial interaction involving L707. In a cellular context, mutation of L707 significantly increased the rate of channel inactivation. Finally, we present a model for TRPV6 CaM-dependent inactivation, which involves a novel so-called "two-tail" mechanism whereby CaM bridges two TRPV6 monomers resulting in closure of the channel pore.


Asunto(s)
Calcio/química , Calmodulina/química , Complejos Multiproteicos/química , Canales Catiónicos TRPV/química , Secuencia de Aminoácidos/genética , Animales , Sitios de Unión , Calcio/metabolismo , Señalización del Calcio/genética , Calmodulina/metabolismo , Células HEK293 , Humanos , Complejos Multiproteicos/genética , Mutación , Unión Proteica , Conformación Proteica , Ratas , Canales Catiónicos TRPV/genética
4.
Biochemistry ; 57(18): 2623-2635, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29584409

RESUMEN

The transient receptor potential vanilloid channel subfamily member 5 (TRPV5) is a highly selective calcium ion channel predominately expressed in the kidney epithelium that plays an essential role in calcium reabsorption from renal infiltrate. In order to maintain Ca2+ homeostasis, TRPV5 possesses a tightly regulated negative feedback mechanism, where the ubiquitous Ca2+ binding protein calmodulin (CaM) directly binds to the intracellular TRPV5 C-terminus, thus regulating TRPV5. Here we report on the characterization of the TRPV5 C-terminal CaM binding site and its interaction with CaM at an atomistic level. We have solved the de novo solution structure of the TRPV5 C-terminus in complex with a CaM mutant, creating conditions that mimic the cellular basal Ca2+ state. We demonstrate that under these conditions the TRPV5 C-terminus is exclusively bound to the CaM C-lobe only, while it confers conformational freedom to the CaM N-lobe. We also show that at elevated calcium levels, additional interactions between the TRPV5 C-terminus and CaM N-lobe occur, resulting in formation of a tight 1:1 complex, effectively making the N-lobe the calcium sensor. Together, these data are consistent with and support the novel model for Ca2+/CaM-dependent inactivation of TRPV channels as proposed by Bate and co-workers [ Bate , N. , et al. ( 2018 ) Biochemistry , ( 57), DOI: 10.1021/acs.biochem.7b01286 ].


Asunto(s)
Canales de Calcio/química , Calmodulina/química , Complejos Multiproteicos/química , Canales Catiónicos TRPV/química , Secuencia de Aminoácidos , Animales , Calcio/química , Canales de Calcio/genética , Calmodulina/genética , Humanos , Complejos Multiproteicos/genética , Unión Proteica , Ratas , Canales Catiónicos TRPV/genética
5.
Mol Pharm ; 15(9): 3962-3968, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30036484

RESUMEN

Angiopoietin-1 (Ang1) is a vascular protective ligand that acts through the receptor tyrosine kinase Tie2 to enhance endothelial survival and quiescence. In sepsis, diabetic retinopathy, and a range of other diseases, Ang2, an antagonist of Tie2, increases markedly. This antagonist suppresses Ang1 protective effects leading to vascular destabilization, inflammation, and endothelial death. Administration of recombinant Ang1 can counter Ang2 antagonism and restore vascular function. However, recombinant Ang1 is needed at sufficiently high concentrations to block Ang2, and the protein is difficult to produce, requires mammalian expression systems, and is prone to aggregation. Here we present an engineered synthetic Tie2 ligand that is not antagonized by Ang2 but is easy to produce and more robust than Ang1. Using a peptide phage display, we isolated a heptameric sequence that binds Tie2-ectodomain and fused this to the coiled:coil domain of cartilage oligomeric matrix protein. This pentameric protein is 60 kDa in size, expressed in E. coli, and facile to purify. The protein, designated TSL1, binds to Tie2-ectodomain in vitro and on the cell surface. TSL1 inhibits endothelial apoptosis. Crucially, TSL1 binds at a site on Tie2 distinct from the angiopoietin-binding site and is resistant to antagonism by Ang2. This engineered ligand has several advantages over recombinant Ang1 for potential therapeutic applications. The study also highlights the value of orthogonal ligands for regulating cellular receptors without being subject to antagonism or modulation by endogenous ligands.


Asunto(s)
Angiopoyetina 2/metabolismo , Biblioteca de Péptidos , Receptor TIE-2/metabolismo , Angiopoyetina 2/genética , Apoptosis/genética , Apoptosis/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Citometría de Flujo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoprecipitación , Receptor TIE-2/genética
6.
Proc Natl Acad Sci U S A ; 112(35): E4864-73, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26283369

RESUMEN

Insight into how molecular machines perform their biological functions depends on knowledge of the spatial organization of the components, their connectivity, geometry, and organizational hierarchy. However, these parameters are difficult to determine in multicomponent assemblies such as integrin-based focal adhesions (FAs). We have previously applied 3D superresolution fluorescence microscopy to probe the spatial organization of major FA components, observing a nanoscale stratification of proteins between integrins and the actin cytoskeleton. Here we combine superresolution imaging techniques with a protein engineering approach to investigate how such nanoscale architecture arises. We demonstrate that talin plays a key structural role in regulating the nanoscale architecture of FAs, akin to a molecular ruler. Talin diagonally spans the FA core, with its N terminus at the membrane and C terminus demarcating the FA/stress fiber interface. In contrast, vinculin is found to be dispensable for specification of FA nanoscale architecture. Recombinant analogs of talin with modified lengths recapitulated its polarized orientation but altered the FA/stress fiber interface in a linear manner, consistent with its modular structure, and implicating the integrin-talin-actin complex as the primary mechanical linkage in FAs. Talin was found to be ∼97 nm in length and oriented at ∼15° relative to the plasma membrane. Our results identify talin as the primary determinant of FA nanoscale organization and suggest how multiple cellular forces may be integrated at adhesion sites.


Asunto(s)
Adhesiones Focales/metabolismo , Nanoestructuras , Talina/fisiología , Humanos , Microscopía Fluorescente
7.
J Biol Chem ; 288(12): 8238-8249, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23389036

RESUMEN

Talin activates integrins, couples them to F-actin, and recruits vinculin to focal adhesions (FAs). Here, we report the structural characterization of the talin rod: 13 helical bundles (R1-R13) organized into a compact cluster of four-helix bundles (R2-R4) within a linear chain of five-helix bundles. Nine of the bundles contain vinculin-binding sites (VBS); R2R3 are atypical, with each containing two VBS. Talin R2R3 also binds synergistically to RIAM, a Rap1 effector involved in integrin activation. Biochemical and structural data show that vinculin and RIAM binding to R2R3 is mutually exclusive. Moreover, vinculin binding requires domain unfolding, whereas RIAM binds the folded R2R3 double domain. In cells, RIAM is enriched in nascent adhesions at the leading edge whereas vinculin is enriched in FAs. We propose a model in which RIAM binding to R2R3 initially recruits talin to membranes where it activates integrins. As talin engages F-actin, force exerted on R2R3 disrupts RIAM binding and exposes the VBS, which recruit vinculin to stabilize the complex.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Adhesiones Focales/metabolismo , Proteínas de la Membrana/química , Talina/química , Vinculina/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Unión Competitiva , Cristalografía por Rayos X , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas de la Membrana/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Talina/metabolismo , Vinculina/metabolismo
8.
EMBO J ; 29(6): 1069-80, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20150896

RESUMEN

Talin is a 270-kDa protein that activates integrins and couples them to cytoskeletal actin. Talin contains an N-terminal FERM domain comprised of F1, F2 and F3 domains, but it is atypical in that F1 contains a large insert and is preceded by an extra domain F0. Although F3 contains the binding site for beta-integrin tails, F0 and F1 are also required for activation of beta1-integrins. Here, we report the solution structures of F0, F1 and of the F0F1 double domain. Both F0 and F1 have ubiquitin-like folds joined in a novel fixed orientation by an extensive charged interface. The F1 insert forms a loop with helical propensity, and basic residues predicted to reside on one surface of the helix are required for binding to acidic phospholipids and for talin-mediated activation of beta1-integrins. This and the fact that basic residues on F2 and F3 are also essential for integrin activation suggest that extensive interactions between the talin FERM domain and acidic membrane phospholipids are required to orientate the FERM domain such that it can activate integrins.


Asunto(s)
Integrinas/metabolismo , Talina/química , Ubiquitina/química , Secuencia de Aminoácidos , Sitios de Unión , Adhesión Celular , Integrinas/química , Modelos Moleculares , Datos de Secuencia Molecular , Pliegue de Proteína , Estructura Terciaria de Proteína , Talina/metabolismo , Ubiquitina/metabolismo
9.
PLoS Biol ; 9(12): e1001223, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22205879

RESUMEN

Mechanotransduction is a critical function for cells, in terms of cell viability, shaping of tissues, and cellular behavior. In vitro, cellular level forces can stretch adhesion proteins that link extracellular matrix to the actin cytoskeleton exposing hidden binding sites. However, there is no evidence that in vivo forces produce significant in vivo stretching to cause domain unfolding. We now report that the adhesion protein, talin, is repeatedly stretched by 100-350 nm in vivo by myosin contraction of actin filaments. Using a functional EGFP-N-Talin1-C-mCherry to measure the length of single talin molecules, we observed that the C-terminal mCherry was normally displaced in the direction of actin flow by 90 to >250 nm from N-EGFP but only by 50-60 nm (talin's length in vitro) after myosin inhibition. Individual talin molecules transiently stretched and relaxed. Peripheral, multimolecular adhesions had green outside and red proximal edges. They also exhibited transient, myosin-dependent stretching of 50-350 nm for 6-16 s; however, expression of the talin-binding head of vinculin increased stretching to about 400 nm and suppressed dynamics. We suggest that rearward moving actin filaments bind, stretch, and release talin in multiple, stochastic stick-slip cycles and that multiple vinculin binding and release cycles integrate pulling on matrices into biochemical signals.


Asunto(s)
Mecanotransducción Celular , Talina/biosíntesis , Vinculina/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Adhesión Celular , Línea Celular , Chlorocebus aethiops , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/metabolismo , Unión Proteica , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Talina/metabolismo
10.
J Struct Biol ; 184(1): 21-32, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23726984

RESUMEN

Talin is a large adaptor protein that activates integrins and couples them to cytoskeletal actin. Talin contains an N-terminal FERM (band 4.1, ezrin, radixin, moesin) domain (the head) linked to a flexible rod comprised of 13 amphipathic helical bundles (R1-R13) that terminate in a C-terminal helix (DD) that forms an anti-parallel dimer. We derived a three-dimensional structural model of full-length talin at a resolution of approximately 2.5nm using EM reconstruction of full-length talin and the known shapes of the individual domains and inter-domain angles as derived from small angle X-ray scattering. Talin adopts a compact conformation consistent with a dimer in which the two talin rods form a donut-shaped structure, with the two talin heads packed side by side occupying the hole at the center of this donut. In this configuration, the integrin binding site in the head domain and the actin-binding site at the carboxy-terminus of the rod are masked, implying that talin must unravel before it can support integrin activation and engage the actin cytoskeleton.


Asunto(s)
Talina/química , Talina/metabolismo , Actinas/química , Actinas/metabolismo , Sitios de Unión , Citoesqueleto/química , Citoesqueleto/metabolismo , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
11.
J Biol Chem ; 287(17): 13799-812, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22351767

RESUMEN

Talin, which is composed of head (THD) and rod domains, plays an important role in cell adhesion events in diverse species including most metazoans and Dictyostelium discoideum. Talin is abundant in the cytosol; however, it mediates adhesion by associating with integrins in the plasma membrane where it forms a primary link between integrins and the actin cytoskeleton. Cells modulate the partitioning of talin between the plasma membrane and the cytosol to control cell adhesion. Here, we combine nuclear magnetic resonance spectroscopy (NMR) with subcellular fractionation to characterize two distinct THD-rod domain interactions that control the interaction of talin with the actin cytoskeleton or its localization to the plasma membrane. An interaction between a discrete vinculin-binding region of the rod (VBS1/2a; Tln1(482-787)), and the THD restrains talin from interacting with the plasma membrane. Furthermore, we show that vinculin binding to VBS1/2a results in talin recruitment to the plasma membrane. Thus, we have structurally defined specific inter-domain interactions between THD and the talin rod domain that regulate the subcellular localization of talin.


Asunto(s)
Regulación de la Expresión Génica , Talina/biosíntesis , Actinas/metabolismo , Animales , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Citoesqueleto/metabolismo , Citosol/metabolismo , Humanos , Espectroscopía de Resonancia Magnética/métodos , Modelos Moleculares , Conformación Proteica , Estructura Terciaria de Proteína , Fracciones Subcelulares/metabolismo , Talina/química
12.
J Biol Chem ; 287(10): 6979-90, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22235127

RESUMEN

The activation of heterodimeric integrin adhesion receptors from low to high affinity states occurs in response to intracellular signals that act on the short cytoplasmic tails of integrin ß subunits. Binding of the talin FERM (four-point-one, ezrin, radixin, moesin) domain to the integrin ß tail provides one key activation signal, but recent data indicate that the kindlin family of FERM domain proteins also play a central role. Kindlins directly bind integrin ß subunit cytoplasmic domains at a site distinct from the talin-binding site, and target to focal adhesions in adherent cells. However, the mechanisms by which kindlins impact integrin activation remain largely unknown. A notable feature of kindlins is their similarity to the integrin-binding and activating talin FERM domain. Drawing on this similarity, here we report the identification of an unstructured insert in the kindlin F1 FERM domain, and provide evidence that a highly conserved polylysine motif in this loop supports binding to negatively charged phospholipid head groups. We further show that the F1 loop and its membrane-binding motif are required for kindlin-1 targeting to focal adhesions, and for the cooperation between kindlin-1 and -2 and the talin head in αIIbß3 integrin activation, but not for kindlin binding to integrin ß tails. These studies highlight the structural and functional similarities between kindlins and the talin head and indicate that as for talin, FERM domain interactions with acidic membrane phospholipids as well ß-integrin tails contribute to the ability of kindlins to activate integrins.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Adhesiones Focales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Secuencias de Aminoácidos , Animales , Células CHO , Proteínas Portadoras/genética , Adhesión Celular/fisiología , Cricetinae , Cricetulus , Proteínas del Citoesqueleto/genética , Adhesiones Focales/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Proteínas Musculares/genética , Proteínas de Neoplasias/genética , Fosfolípidos/genética , Fosfolípidos/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Estructura Terciaria de Proteína , Talina/genética , Talina/metabolismo
13.
Pflugers Arch ; 465(11): 1507-19, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23609407

RESUMEN

The Ca²âº-binding protein calmodulin (CaM) is a well-known regulator of ion-channel activity. Consequently, the Protein Data Bank contains many structures of CaM in complex with different fragments of ion channels that together display a variety of binding modes. In addition to the canonical interaction, in which CaM engages its target with both its domains, many of the ion-channel-CaM complexes demonstrate alternative non-canonical binding modes that depend on the target and experimental conditions. Based on these findings, several mechanisms of ion-channel regulation by CaM have been proposed, all exploiting its plasticity and flexibility in interacting with its targets. In this review, we focus on complexes of CaM with either the voltage-gated calcium channels; the voltage-gated sodium channels or the small conductance calcium-activated potassium channels, for which both structural and functional data are available. For each channel, the functional relevance of these structural data and possible mechanism of calcium-dependent (in)activation and/or facilitation are discussed in detail.


Asunto(s)
Canales de Calcio/metabolismo , Calmodulina/química , Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Calmodulina/metabolismo , Humanos , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica
14.
EMBO J ; 28(22): 3623-32, 2009 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-19798053

RESUMEN

Fundamental to cell adhesion and migration, integrins are large heterodimeric membrane proteins that uniquely mediate inside-out signal transduction, whereby adhesion to the extracellular matrix is activated from within the cell by direct binding of talin to the cytoplasmic tail of the beta integrin subunit. Here, we report the first structure of talin bound to an authentic full-length beta integrin tail. Using biophysical and whole cell measurements, we show that a specific ionic interaction between the talin F3 domain and the membrane-proximal helix of the beta tail disrupts an integrin alpha/beta salt bridge that helps maintain the integrin inactive state. Second, we identify a positively charged surface on the talin F2 domain that precisely orients talin to disrupt the heterodimeric integrin transmembrane (TM) complex. These results show key structural features that explain the ability of talin to mediate inside-out TM signalling.


Asunto(s)
Integrinas/química , Sustancias Macromoleculares/química , Transducción de Señal/fisiología , Talina/química , Secuencia de Aminoácidos , Animales , Células CHO , Membrana Celular/metabolismo , Polaridad Celular/fisiología , Cricetinae , Cricetulus , Integrinas/metabolismo , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Talina/metabolismo
15.
EMBO J ; 27(2): 458-69, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18157087

RESUMEN

Talin is a large dimeric protein that couples integrins to cytoskeletal actin. Here, we report the structure of the C-terminal actin-binding domain of talin, the core of which is a five-helix bundle linked to a C-terminal helix responsible for dimerisation. The NMR structure of the bundle reveals a conserved surface-exposed hydrophobic patch surrounded by positively charged groups. We have mapped the actin-binding site to this surface and shown that helix 1 on the opposite side of the bundle negatively regulates actin binding. The crystal structure of the dimerisation helix reveals an antiparallel coiled-coil with conserved residues clustered on the solvent-exposed face. Mutagenesis shows that dimerisation is essential for filamentous actin (F-actin) binding and indicates that the dimerisation helix itself contributes to binding. We have used these structures together with small angle X-ray scattering to derive a model of the entire domain. Electron microscopy provides direct evidence for binding of the dimer to F-actin and indicates that it binds to three monomers along the long-pitch helix of the actin filament.


Asunto(s)
Actinas/metabolismo , Proteínas Recombinantes/química , Talina/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Procesamiento de Imagen Asistido por Computador , Espectroscopía de Resonancia Magnética , Ratones , Microscopía Electrónica de Rastreo , Modelos Biológicos , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Conejos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Talina/genética , Talina/metabolismo
16.
Biochem Biophys Res Commun ; 426(2): 190-5, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22925892

RESUMEN

Mice homozygous for several Tln2 gene targeted alleles are viable and fertile. Here we show that although the expression of talin2 protein is drastically reduced in muscle from these mice, other tissues continue to express talin2 albeit at reduced levels. We therefore generated a Tln2 allele lacking the entire coding sequence (Tln2(cd)). Tln2(cd/cd) mice were viable and fertile, and the genotypes of Tln2(cd/+) intercrosses were at the expected Mendelian ratio. Tln2(cd/cd) mice showed no major difference in body mass or the weight of the major organs compared to wild-type, although they displayed a mildly dystrophic phenotype. Moreover, Tln2(cd/cd) mouse embryo fibroblasts showed no obvious defects in cell adhesion, migration or proliferation. However, the number of Tln2(cd/cd) pups surviving to adulthood was variable suggesting that such mice have an underlying defect.


Asunto(s)
Desarrollo Embrionario/genética , Fertilidad , Talina/fisiología , Animales , Peso Corporal , Adhesión Celular , Movimiento Celular , Proliferación Celular , Femenino , Fibroblastos/fisiología , Eliminación de Gen , Masculino , Ratones , Ratones Noqueados , Distrofias Musculares/genética , Distrofias Musculares/patología , Talina/genética
17.
J Biol Chem ; 285(38): 29577-87, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20610383

RESUMEN

Talin is an adaptor protein that couples integrins to F-actin. Structural studies show that the N-terminal talin head contains an atypical FERM domain, whereas the N- and C-terminal parts of the talin rod include a series of α-helical bundles. However, determining the structure of the central part of the rod has proved problematic. Residues 1359-1659 are homologous to the MESDc1 gene product, and we therefore expressed this region of talin in Escherichia coli. The crystal structure shows a unique fold comprised of a 5- and 4-helix bundle. The 5-helix bundle is composed of nonsequential helices due to insertion of the 4-helix bundle into the loop at the C terminus of helix α3. The linker connecting the bundles forms a two-stranded anti-parallel ß-sheet likely limiting the relative movement of the two bundles. Because the 5-helix bundle contains the N and C termini of this module, we propose that it is linked by short loops to adjacent bundles, whereas the 4-helix bundle protrudes from the rod. This suggests the 4-helix bundle has a unique role, and its pI (7.8) is higher than other rod domains. Both helical bundles contain vinculin-binding sites but that in the isolated 5-helix bundle is cryptic, whereas that in the isolated 4-helix bundle is constitutively active. In contrast, both bundles are required for actin binding. Finally, we show that the MESDc1 protein, which is predicted to have a similar fold, is a novel actin-binding protein.


Asunto(s)
Actinas/química , Actinas/metabolismo , Talina/química , Talina/metabolismo , Vinculina/química , Vinculina/metabolismo , Actinas/genética , Animales , Sitios de Unión , Pollos , Dicroismo Circular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Ratones , Células 3T3 NIH , Unión Proteica/genética , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Talina/genética , Vinculina/genética
18.
Sci Rep ; 8(1): 14689, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30279502

RESUMEN

The Sin3A complex acts as a transcriptional hub, integrating the function of diverse transcription factors with histone modifying enzymes, notably, histone deacetylases (HDAC) 1 and 2. The Sin3A protein sits at the centre of the complex, mediating multiple simultaneous protein-protein interactions via its four paired-amphipathic helix (PAH) domains (PAH1-4). The PAH domains contain a conserved four helical bundle, generating a hydrophobic cleft into which the single-helix of a Sin3-interaction domain (SID) is able to insert and bind with high affinity. Although they share a similar mode of interaction, the SIDs of different repressor proteins bind to only one of four potential PAH domains, due to the specific combination of hydrophobic residues at the interface. Here we report the identification of a highly conserved SID in the 5-methylcytosine dioxygenase, Tet1 (Tet1-SID), which interacts directly with the PAH1 domain of Sin3A. Using a combination of NMR spectroscopy and homology modelling we present a model of the PAH1/Tet1-SID complex, which binds in a Type-II orientation similar to Sap25. Mutagenesis of key residues show that the 11-amino acid Tet1-SID is necessary and sufficient for the interaction with Sin3A and is absolutely required for Tet1 to repress transcription in cells.


Asunto(s)
Oxigenasas de Función Mixta/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Sustitución de Aminoácidos , Análisis Mutacional de ADN , Células HEK293 , Humanos , Espectroscopía de Resonancia Magnética , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/genética , Modelos Moleculares , Unión Proteica , Conformación Proteica , Dominios Proteicos , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/química , Proteínas Represoras/genética , Complejo Correpresor Histona Desacetilasa y Sin3
19.
Nat Biotechnol ; 20(7): 713-6, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12089557

RESUMEN

The antibacterial properties of macrolide antibiotics (such as erythromycin, tylosin, and narbomycin) depend ultimately on the glycosylation of otherwise inactive polyketide lactones. Among the sugars commonly found in such macrolides are various 6-deoxyhexoses including the 3-dimethylamino sugars mycaminose and desosamine (4-deoxymycaminose). Some macrolides (such as tylosin) possess multiple sugar moieties, whereas others (such as narbomycin) have only single sugar substituents. As patterns of glycosylation markedly influence a macrolide's drug activity, there is considerable interest in the possibility of using combinatorial biosynthesis to generate new pairings of polyketide lactones with sugars, especially 6-deoxyhexoses. Here, we report a successful attempt to alter the aminodeoxyhexose-biosynthetic capacity of Streptomyces fradiae (a producer of tylosin) by importing genes from the narbomycin producer Streptomyces narbonensis. This engineered S. fradiae produced substantial amounts of two potentially useful macrolides that had not previously been obtained by fermentation.


Asunto(s)
Amino Azúcares/biosíntesis , Amino Azúcares/genética , Glucosamina/análogos & derivados , Glucosamina/biosíntesis , Glucosamina/genética , Streptomyces/genética , Streptomyces/metabolismo , Regulación Bacteriana de la Expresión Génica , Ingeniería Genética/métodos , Macrólidos/metabolismo , Transformación Bacteriana
20.
Nat Commun ; 6: 10038, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26634421

RESUMEN

The link between extracellular-matrix-bound integrins and intracellular F-actin is essential for cell spreading and migration. Here, we demonstrate how the actin-binding proteins talin and vinculin cooperate to provide this link. By expressing structure-based talin mutants in talin null cells, we show that while the C-terminal actin-binding site (ABS3) in talin is required for adhesion complex assembly, the central ABS2 is essential for focal adhesion (FA) maturation. Thus, although ABS2 mutants support cell spreading, the cells lack FAs, fail to polarize and exert reduced force on the surrounding matrix. ABS2 is inhibited by the preceding mechanosensitive vinculin-binding R3 domain, and deletion of R2R3 or expression of constitutively active vinculin generates stable force-independent FAs, although cell polarity is compromised. Our data suggest a model whereby force acting on integrin-talin complexes via ABS3 promotes R3 unfolding and vinculin binding, activating ABS2 and locking talin into an actin-binding configuration that stabilizes FAs.


Asunto(s)
Actomiosina/metabolismo , Talina/metabolismo , Vinculina/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actinas/genética , Actomiosina/genética , Animales , Polaridad Celular , Adhesiones Focales/química , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Ratones , Células 3T3 NIH , Unión Proteica , Estructura Terciaria de Proteína , Talina/química , Talina/genética , Vinculina/química , Vinculina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA