Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 186(10): 2219-2237.e29, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37172566

RESUMEN

The Commander complex is required for endosomal recycling of diverse transmembrane cargos and is mutated in Ritscher-Schinzel syndrome. It comprises two sub-assemblies: Retriever composed of VPS35L, VPS26C, and VPS29; and the CCC complex which contains twelve subunits: COMMD1-COMMD10 and the coiled-coil domain-containing (CCDC) proteins CCDC22 and CCDC93. Combining X-ray crystallography, electron cryomicroscopy, and in silico predictions, we have assembled a complete structural model of Commander. Retriever is distantly related to the endosomal Retromer complex but has unique features preventing the shared VPS29 subunit from interacting with Retromer-associated factors. The COMMD proteins form a distinctive hetero-decameric ring stabilized by extensive interactions with CCDC22 and CCDC93. These adopt a coiled-coil structure that connects the CCC and Retriever assemblies and recruits a 16th subunit, DENND10, to form the complete Commander complex. The structure allows mapping of disease-causing mutations and reveals the molecular features required for the function of this evolutionarily conserved trafficking machinery.


Asunto(s)
Anomalías Múltiples , Anomalías Craneofaciales , Complejos Multiproteicos , Humanos , Endosomas/metabolismo , Transporte de Proteínas , Proteínas/metabolismo , Complejos Multiproteicos/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-38886588

RESUMEN

Endosomes are central protein-sorting stations at the crossroads of numerous membrane trafficking pathways in all eukaryotes. They have a key role in protein homeostasis and cellular signalling and are involved in the pathogenesis of numerous diseases. Endosome-associated protein assemblies or coats collect transmembrane cargo proteins and concentrate them into retrieval domains. These domains can extend into tubular carriers, which then pinch off from the endosomal membrane and deliver the cargoes to appropriate subcellular compartments. Here we discuss novel insights into the structure of a number of tubular membrane coats that mediate the recruitment of cargoes into these carriers, focusing on sorting nexin-based coats such as Retromer, Commander and ESCPE-1. We summarize current and emerging views of how selective tubular endosomal carriers form and detach from endosomes by fission, highlighting structural aspects, conceptual challenges and open questions.

3.
PLoS Biol ; 20(4): e3001601, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35417450

RESUMEN

Coat complexes coordinate cargo recognition through cargo adaptors with biogenesis of transport carriers during integral membrane protein trafficking. Here, we combine biochemical, structural, and cellular analyses to establish the mechanistic basis through which SNX27-Retromer, a major endosomal cargo adaptor, couples to the membrane remodeling endosomal SNX-BAR sorting complex for promoting exit 1 (ESCPE-1). In showing that the SNX27 FERM (4.1/ezrin/radixin/moesin) domain directly binds acidic-Asp-Leu-Phe (aDLF) motifs in the SNX1/SNX2 subunits of ESCPE-1, we propose a handover model where SNX27-Retromer captured cargo proteins are transferred into ESCPE-1 transport carriers to promote endosome-to-plasma membrane recycling. By revealing that assembly of the SNX27:Retromer:ESCPE-1 coat evolved in a stepwise manner during early metazoan evolution, likely reflecting the increasing complexity of endosome-to-plasma membrane recycling from the ancestral opisthokont to modern animals, we provide further evidence of the functional diversification of yeast pentameric Retromer in the recycling of hundreds of integral membrane proteins in metazoans.


Asunto(s)
Endosomas , Nexinas de Clasificación , Animales , Membrana Celular/metabolismo , Endosomas/metabolismo , Transporte de Proteínas , Nexinas de Clasificación/metabolismo
4.
Traffic ; 20(7): 465-478, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30993794

RESUMEN

Endosomes are dynamic intracellular compartments that control the sorting of a constant stream of different transmembrane cargos either for ESCRT-mediated degradation or for egress and recycling to compartments such as the Golgi and the plasma membrane. The recycling of cargos occurs within tubulovesicular membrane domains and is facilitated by peripheral membrane protein machineries that control both membrane remodelling and selection of specific transmembrane cargos. One of the primary sorting machineries is the Retromer complex, which controls the recycling of a large array of different cargo molecules in cooperation with various sorting nexin (SNX) adaptor proteins. Recently a Retromer-like complex was also identified that controls plasma membrane recycling of cargos including integrins and lipoprotein receptors. Termed "Retriever," this complex uses a different SNX family member SNX17 for cargo recognition, and cooperates with the COMMD/CCDC93/CCDC22 (CCC) complex to form a larger assembly called "Commander" to mediate endosomal trafficking. In this review we focus on recent advances that have begun to provide a molecular understanding of these two distantly related transport machineries.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Humanos , Nexinas de Clasificación/química , Nexinas de Clasificación/metabolismo
5.
Proc Natl Acad Sci U S A ; 109(25): 9816-21, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22670057

RESUMEN

When nerve cells communicate, vesicles from one neuron fuse with the presynaptic membrane releasing chemicals that signal to the next. Similarly, when insulin binds its receptor on adipocytes or muscle, glucose transporter-4 vesicles fuse with the cell membrane, allowing glucose to be imported. These essential processes require the interaction of SNARE proteins on vesicle and cell membranes, as well as the enigmatic protein Munc18 that binds the SNARE protein Syntaxin. Here, we show that in solution the neuronal protein Syntaxin1a interacts with Munc18-1 whether or not the Syntaxin1a N-peptide is present. Conversely, the adipocyte protein Syntaxin4 does not bind its partner Munc18c unless the N-peptide is present. Solution-scattering data for the Munc18-1:Syntaxin1a complex in the absence of the N-peptide indicates that this complex adopts the inhibitory closed binding mode, exemplified by a crystal structure of the complex. However, when the N-peptide is present, the solution-scattering data indicate both Syntaxin1a and Syntaxin4 adopt extended conformations in complexes with their respective Munc18 partners. The low-resolution solution structure of the open Munc18:Syntaxin binding mode was modeled using data from cross-linking/mass spectrometry, small-angle X-ray scattering, and small-angle neutron scattering with contrast variation, indicating significant differences in Munc18:Syntaxin interactions compared with the closed binding mode. Overall, our results indicate that the neuronal Munc18-1:Syntaxin1a proteins can adopt two alternate and functionally distinct binding modes, closed and open, depending on the presence of the N-peptide, whereas Munc18c:Syntaxin4 adopts only the open binding mode.


Asunto(s)
Proteínas Munc18/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Munc18/química , Fragmentos de Péptidos/química , Unión Proteica , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
6.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 5): 774-84, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23633586

RESUMEN

The caspase recruitment domain (CARD) is present in death-domain superfamily proteins involved in inflammation and apoptosis. BinCARD is named for its ability to interact with Bcl10 and inhibit downstream signalling. Human BinCARD is expressed as two isoforms that encode the same N-terminal CARD region but which differ considerably in their C-termini. Both isoforms are expressed in immune cells, although BinCARD-2 is much more highly expressed. Crystals of the CARD fold common to both had low symmetry (space group P1). Molecular replacement was unsuccessful in this low-symmetry space group and, as the construct contains no methionines, first one and then two residues were engineered to methionine for MAD phasing. The double-methionine variant was produced as a selenomethionine derivative, which was crystallized and the structure was solved using data measured at two wavelengths. The crystal structures of the native and selenomethionine double mutant were refined to high resolution (1.58 and 1.40 Šresolution, respectively), revealing the presence of a cis-peptide bond between Tyr39 and Pro40. Unexpectedly, the native crystal structure revealed that all three cysteines were oxidized. The mitochondrial localization of BinCARD-2 and the susceptibility of its CARD region to redox modification points to the intriguing possibility of a redox-regulatory role.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/química , Proteínas/química , Proteínas/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Cristalografía por Rayos X , Cisteína/metabolismo , Células HeLa , Humanos , Mitocondrias/metabolismo , Modelos Moleculares , Mutación , Oxidación-Reducción , Prolina/química , Conformación Proteica , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas/genética , Selenometionina
7.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 6): 637-48, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22683786

RESUMEN

Fam96a mRNA, which encodes a mammalian DUF59 protein, is enriched in macrophages. Recombinant human Fam96a forms stable monomers and dimers in solution. Crystal structures of these two forms revealed that each adopts a distinct type of domain-swapped dimer, one of which is stabilized by zinc binding. Two hinge loops control Fam96a domain swapping; both are flexible and highly conserved, suggesting that domain swapping may be a common feature of eukaryotic but not bacterial DUF59 proteins. The derived monomer fold of Fam96a diverges from that of bacterial DUF59 counterparts in that the C-terminal region of Fam96a is much longer and is positioned on the opposite side of the N-terminal core fold. The putative metal-binding site of bacterial DUF59 proteins is not conserved in Fam96a, but Fam96a interacts tightly in vitro with Ciao1, the cytosolic iron-assembly protein. Moreover, Fam96a and Ciao1 can be co-immunoprecipitated, suggesting that the interaction also occurs in vivo. Although predicted to have a signal peptide, it is shown that Fam96a is cytoplasmic. The data reveal that eukaryotic DUF59 proteins share intriguing characteristics with amyloidogenic proteins.


Asunto(s)
Proteínas Portadoras/química , Multimerización de Proteína , Animales , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Células HeLa , Humanos , Metalochaperonas/metabolismo , Metaloproteínas , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Homología Estructural de Proteína
8.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 10): 1290-302, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22993083

RESUMEN

The enzyme TcpG is a periplasmic protein produced by the Gram-negative pathogen Vibrio cholerae. TcpG is essential for the production of ToxR-regulated proteins, including virulence-factor pilus proteins and cholera toxin, and is therefore a target for the development of a new class of anti-virulence drugs. Here, the 1.2 Å resolution crystal structure of TcpG is reported using a cryocooled crystal. This structure is compared with a previous crystal structure determined at 2.1 Å resolution from data measured at room temperature. The new crystal structure is the first DsbA crystal structure to be solved at a sufficiently high resolution to allow the inclusion of refined H atoms in the model. The redox properties of TcpG are also reported, allowing comparison of its oxidoreductase activity with those of other DSB proteins. One of the defining features of the Escherichia coli DsbA enzyme is its destabilizing disulfide, and this is also present in TcpG. The data presented here provide new insights into the structure and redox properties of this enzyme, showing that the binding mode identified between E. coli DsbB and DsbA is likely to be conserved in TcpG and that the ß5-α7 loop near the proposed DsbB binding site is flexible, and suggesting that the tense oxidized conformation of TcpG may be the consequence of a short contact at the active site that is induced by disulfide formation and is relieved by reduction.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Portadoras/química , Toxina del Cólera/fisiología , Fimbrias Bacterianas/fisiología , Proteína Disulfuro Isomerasas/química , Vibrio cholerae/enzimología , Proteínas Bacterianas/fisiología , Proteínas Portadoras/fisiología , Toxina del Cólera/biosíntesis , Cristalografía por Rayos X , Fimbrias Bacterianas/enzimología , Hidrógeno/química , Oxidación-Reducción , Proteína Disulfuro Isomerasas/fisiología , Vibrio cholerae/patogenicidad
9.
Nat Commun ; 13(1): 260, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017494

RESUMEN

Advances in peptide and protein therapeutics increased the need for rapid and cost-effective polypeptide prototyping. While in vitro translation systems are well suited for fast and multiplexed polypeptide prototyping, they suffer from misfolding, aggregation and disulfide-bond scrambling of the translated products. Here we propose that efficient folding of in vitro produced disulfide-rich peptides and proteins can be achieved if performed in an aggregation-free and thermodynamically controlled folding environment. To this end, we modify an E. coli-based in vitro translation system to allow co-translational capture of translated products by affinity matrix. This process reduces protein aggregation and enables productive oxidative folding and recycling of misfolded states under thermodynamic control. In this study we show that the developed approach is likely to be generally applicable for prototyping of a wide variety of disulfide-constrained peptides, macrocyclic peptides with non-native bonds and antibody fragments in amounts sufficient for interaction analysis and biological activity assessment.


Asunto(s)
Sistema Libre de Células/efectos de los fármacos , Medicamentos Genéricos/química , Medicamentos Genéricos/farmacología , Péptidos/química , Péptidos/farmacología , Animales , Anticuerpos , Análisis Costo-Beneficio , Interpretación Estadística de Datos , Disulfuros , Drosophila melanogaster , Escherichia coli , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Leishmania , Péptidos/genética , Agregado de Proteínas , Dominios Proteicos , ARN Ribosómico 16S , Biología Sintética , Termodinámica
10.
Curr Res Struct Biol ; 3: 179-186, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34401749

RESUMEN

Chlorotoxin (ClTx) is a 36-residue disulfide-rich peptide isolated from the venom of the scorpion Leiurus quinquestriatus. This peptide has been shown to selectively bind to brain tumours (gliomas), however, with conflicting reports regarding its direct cellular target. Recently, the vascular endothelial growth factor receptor, neuropilin-1 (NRP1) has emerged as a potential target of the peptide. Here, we sought to characterize the details of the binding of ClTx to the b1-domain of NRP1 (NRP1-b1) using solution state nuclear magnetic resonance (NMR) spectroscopy. The 3D structure of the isotope labelled peptide was solved using multidimensional heteronuclear NMR spectroscopy to produce a well-resolved structural ensemble. The structure points to three putative protein-protein interaction interfaces, two basic patches (R14/K15/K23 and R25/K27/R36) and a hydrophobic patch (F6/T7/T8/H10). The NRP1-b1 binding interface of ClTx was elucidated using 15N chemical shift mapping and included the R25/K27/R36 region of the peptide. The thermodynamics of binding was determined using isothermal titration calorimetry (ITC). In both NMR and ITC measurements, the binding was shown to be competitive with a known NRP1-b1 inhibitor. Finally, combining all of this data we generate a model of the ClTx:NRP1-b1 complex. The data shows that the peptide binds to the same region of NRP1 that is used by the SARS-CoV-2 virus for cell entry, however, via a non-canonical binding mode. Our results provide evidence for a non-standard NRP1 binding motif, while also providing a basis for further engineering of ClTx to generate peptides with improved NRP1 binding for future biomedical applications.

11.
Sci Adv ; 7(49): eabg4007, 2021 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-34851660

RESUMEN

The retromer complex (Vps35-Vps26-Vps29) is essential for endosomal membrane trafficking and signaling. Mutation of the retromer subunit Vps35 causes late-onset Parkinson's disease, while viral and bacterial pathogens can hijack the complex during cellular infection. To modulate and probe its function, we have created a novel series of macrocyclic peptides that bind retromer with high affinity and specificity. Crystal structures show that most of the cyclic peptides bind to Vps29 via a Pro-Leu­containing sequence, structurally mimicking known interactors such as TBC1D5 and blocking their interaction with retromer in vitro and in cells. By contrast, macrocyclic peptide RT-L4 binds retromer at the Vps35-Vps26 interface and is a more effective molecular chaperone than reported small molecules, suggesting a new therapeutic avenue for targeting retromer. Last, tagged peptides can be used to probe the cellular localization of retromer and its functional interactions in cells, providing novel tools for studying retromer function.

12.
J Mol Biol ; 433(13): 166964, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-33781758

RESUMEN

Recent years have seen a dramatic improvement in protein-design methodology. Nevertheless, most methods demand expert intervention, limiting their widespread adoption. By contrast, the PROSS algorithm for improving protein stability and heterologous expression levels has been successfully applied to a range of challenging enzymes and binding proteins. Here, we benchmark the application of PROSS as a stand-alone tool for protein scientists with no or limited experience in modeling. Twelve laboratories from the Protein Production and Purification Partnership in Europe (P4EU) challenged the PROSS algorithm with 14 unrelated protein targets without support from the PROSS developers. For each target, up to six designs were evaluated for expression levels and in some cases, for thermal stability and activity. In nine targets, designs exhibited increased heterologous expression levels either in prokaryotic and/or eukaryotic expression systems under experimental conditions that were tailored for each target protein. Furthermore, we observed increased thermal stability in nine of ten tested targets. In two prime examples, the human Stem Cell Factor (hSCF) and human Cadherin-Like Domain (CLD12) from the RET receptor, the wild type proteins were not expressible as soluble proteins in E. coli, yet the PROSS designs exhibited high expression levels in E. coli and HEK293 cells, respectively, and improved thermal stability. We conclude that PROSS may improve stability and expressibility in diverse cases, and that improvement typically requires target-specific expression conditions. This study demonstrates the strengths of community-wide efforts to probe the generality of new methods and recommends areas for future research to advance practically useful algorithms for protein science.


Asunto(s)
Algoritmos , Estabilidad Proteica , Animales , Escherichia coli/metabolismo , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Proteínas/química , Proteínas/metabolismo , Solubilidad , Temperatura , Pez Cebra
13.
Science ; 370(6518): 861-865, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33082294

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), uses the viral spike (S) protein for host cell attachment and entry. The host protease furin cleaves the full-length precursor S glycoprotein into two associated polypeptides: S1 and S2. Cleavage of S generates a polybasic Arg-Arg-Ala-Arg carboxyl-terminal sequence on S1, which conforms to a C-end rule (CendR) motif that binds to cell surface neuropilin-1 (NRP1) and NRP2 receptors. We used x-ray crystallography and biochemical approaches to show that the S1 CendR motif directly bound NRP1. Blocking this interaction by RNA interference or selective inhibitors reduced SARS-CoV-2 entry and infectivity in cell culture. NRP1 thus serves as a host factor for SARS-CoV-2 infection and may potentially provide a therapeutic target for COVID-19.


Asunto(s)
Betacoronavirus/fisiología , Neuropilina-1/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Enzima Convertidora de Angiotensina 2 , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , COVID-19 , Células CACO-2 , Infecciones por Coronavirus/virología , Cristalografía por Rayos X , Furina/metabolismo , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/química , Neuropilina-1/genética , Pandemias , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/virología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética
14.
Nat Commun ; 10(1): 1528, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30948714

RESUMEN

Phox homology (PX) domains are membrane interacting domains that bind to phosphatidylinositol phospholipids or phosphoinositides, markers of organelle identity in the endocytic system. Although many PX domains bind the canonical endosome-enriched lipid PtdIns3P, others interact with alternative phosphoinositides, and a precise understanding of how these specificities arise has remained elusive. Here we systematically screen all human PX domains for their phospholipid preferences using liposome binding assays, biolayer interferometry and isothermal titration calorimetry. These analyses define four distinct classes of human PX domains that either bind specifically to PtdIns3P, non-specifically to various di- and tri-phosphorylated phosphoinositides, bind both PtdIns3P and other phosphoinositides, or associate with none of the lipids tested. A comprehensive evaluation of PX domain structures reveals two distinct binding sites that explain these specificities, providing a basis for defining and predicting the functional membrane interactions of the entire PX domain protein family.


Asunto(s)
Fosfatidilinositoles/química , Sitios de Unión , Calorimetría , Humanos , Interferometría , Modelos Moleculares , Fosfatidilinositoles/metabolismo , Dominios Proteicos , Análisis de Secuencia de Proteína , Nexinas de Clasificación/química , Nexinas de Clasificación/metabolismo
15.
Structure ; 26(12): 1612-1625.e4, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30293811

RESUMEN

Phosphorylation of phosphoinositides by the class II phosphatidylinositol 3-kinase (PI3K) PI3K-C2α is essential for many processes, including neuroexocytosis and formation of clathrin-coated vesicles. A defining feature of the class II PI3Ks is a C-terminal module composed of phox-homology (PX) and C2 membrane interacting domains; however, the mechanisms that control their specific cellular localization remain poorly understood. Here we report the crystal structure of the C2 domain of PI3K-C2α in complex with the phosphoinositide head-group mimic inositol hexaphosphate, revealing two distinct pockets for membrane binding. The C2 domain preferentially binds to phosphatidylinositol 4,5-bisphosphate and phosphatidylinositol (3,4,5)-trisphosphate, and low-resolution structures of the combined PX-C2 module by small-angle X-ray scattering reveal a compact conformation in which cooperative lipid binding by each domain binding can occur. Finally, we demonstrate an unexpected role for calcium in perturbing the membrane interactions of the PX-C2 module, which we speculate may be important for regulating the activity of PI3K-C2α.


Asunto(s)
Membrana Celular/metabolismo , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositoles/metabolismo , Sitios de Unión , Calcio/metabolismo , Línea Celular , Membrana Celular/química , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Fosforilación , Unión Proteica , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
16.
Elife ; 72018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30067224

RESUMEN

The COMMD proteins are a conserved family of proteins with central roles in intracellular membrane trafficking and transcription. They form oligomeric complexes with each other and act as components of a larger assembly called the CCC complex, which is localized to endosomal compartments and mediates the transport of several transmembrane cargos. How these complexes are formed however is completely unknown. Here, we have systematically characterised the interactions between human COMMD proteins, and determined structures of COMMD proteins using X-ray crystallography and X-ray scattering to provide insights into the underlying mechanisms of homo- and heteromeric assembly. All COMMD proteins possess an α-helical N-terminal domain, and a highly conserved C-terminal domain that forms a tightly interlocked dimeric structure responsible for COMMD-COMMD interactions. The COMM domains also bind directly to components of CCC and mediate non-specific membrane association. Overall these studies show that COMMD proteins function as obligatory dimers with conserved domain architectures.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/uso terapéutico , Complejos Multiproteicos/química , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Cristalografía por Rayos X , Endosomas/química , Endosomas/genética , Humanos , Membranas Intracelulares/química , Membranas Intracelulares/metabolismo , Proteínas de Transporte de Membrana/genética , Complejos Multiproteicos/genética , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos , Mapeo de Interacción de Proteínas , Alineación de Secuencia , Transducción de Señal/genética , Transcripción Genética
17.
Structure ; 23(4): 653-64, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25728925

RESUMEN

Epidermal growth factor receptor (EGFR) pathway substrate 15 (Eps15) is a newly identified substrate for protein tyrosine phosphatase N3 (PTPN3), which belongs to the FERM-containing PTP subfamily comprising five members including PTPN3, N4, N13, N14, and N21. We solved the crystal structures of the PTPN3-Eps15 phosphopeptide complex and found that His812 of PTPN3 and Pro850 of Eps15 are responsible for the specific interaction between them. We defined the critical role of the additional residue Tyr676 of PTPN3, which is replaced by Ile939 in PTPN14, in recognition of tyrosine phosphorylated Eps15. The WPD loop necessary for catalysis is present in all members but not PTPN21. We identified that Glu instead of Asp in the WPE loop contributes to the catalytic incapability of PTPN21 due to an extended distance beyond protonation targeting a phosphotyrosine substrate. Together with in vivo validations, our results provide novel insights into the substrate specificity and plasticity of FERM-containing PTPs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Simulación del Acoplamiento Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 3/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Células HEK293 , Humanos , Datos de Secuencia Molecular , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Especificidad por Sustrato
18.
Sci Signal ; 7(347): ra98, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25314968

RESUMEN

The mitogen-activated protein kinase p38γ (also known as MAPK12) and its specific phosphatase PTPN3 (also known as PTPH1) cooperate to promote Ras-induced oncogenesis. We determined the architecture of the PTPN3-p38γ complex by a hybrid method combining x-ray crystallography, small-angle x-ray scattering, and chemical cross-linking coupled to mass spectrometry. A unique feature of the glutamic acid-containing loop (E-loop) of the phosphatase domain defined the substrate specificity of PTPN3 toward fully activated p38γ. The solution structure revealed the formation of an active-state complex between p38γ and the phosphatase domain of PTPN3. The PDZ domain of PTPN3 stabilized the active-state complex through an interaction with the PDZ-binding motif of p38γ. This interaction alleviated autoinhibition of PTPN3, enabling efficient tyrosine dephosphorylation of p38γ. Our findings may enable structure-based drug design targeting the PTPN3-p38γ interaction as an anticancer therapeutic.


Asunto(s)
Proteína Quinasa 12 Activada por Mitógenos/química , Dominios PDZ , Proteína Tirosina Fosfatasa no Receptora Tipo 3/química , Regulación Alostérica , Antineoplásicos/química , Clonación Molecular , Reactivos de Enlaces Cruzados/química , Cristalografía por Rayos X , Diseño de Fármacos , Glutatión Transferasa/metabolismo , Humanos , Espectrometría de Masas , Mutagénesis Sitio-Dirigida , Neopterin/química , Péptidos/química , Fosforilación , Unión Proteica , Espectrometría de Masa por Ionización de Electrospray , Especificidad por Sustrato , Tripsina/química , Tirosina/química , Ultracentrifugación
19.
Biomol NMR Assign ; 7(2): 117-20, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22618863

RESUMEN

Proteins containing a domain of unknown function 59 (DUF59) appear to have a variety of physiological functions, ranging from iron-sulfur cluster assembly to DNA repair. DUF59 proteins have been found in bacteria, archaea and eukaryotes, however Fam96a and Fam96b are the only mammalian proteins predicted to contain a DUF59 domain. Fam96a is an 18 kDa protein comprised primarily of a DUF59 domain (residues 31-157) and an N-terminal signal peptide (residues 1-27). Interestingly, the DUF59 domain of Fam96a exists as monomeric and dimeric forms in solution, and X-ray crystallography studies of both forms unexpectedly revealed two different domain-swapped dimer structures. Here we report the backbone resonance assignments and secondary structure of the monomeric form of the 127 residue DUF59 domain of human Fam96a. This study provides the basis for further understanding the structural variability exhibited by Fam96a and the mechanism for domain swapping.


Asunto(s)
Proteínas Portadoras/química , Resonancia Magnética Nuclear Biomolecular , Secuencia de Aminoácidos , Humanos , Metaloproteínas , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Terciaria de Proteína
20.
PLoS One ; 4(11): e7851, 2009 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-19924245

RESUMEN

BACKGROUND: Protein crystallisation screening involves the parallel testing of large numbers of candidate conditions with the aim of identifying conditions suitable as a starting point for the production of diffraction quality crystals. Generally, condition screening is performed in 96-well plates. While previous studies have examined the effects of protein construct, protein purity, or crystallisation condition ingredients on protein crystallisation, few have examined the effect of the crystallisation plate. METHODOLOGY/PRINCIPAL FINDINGS: We performed a statistically rigorous examination of protein crystallisation, and evaluated interactions between crystallisation success and plate row/column, different plates of same make, different plate makes and different proteins. From our analysis of protein crystallisation, we found a significant interaction between plate make and the specific protein being crystallised. CONCLUSIONS/SIGNIFICANCE: Protein crystal structure determination is the principal method for determining protein structure but is limited by the need to produce crystals of the protein under study. Many important proteins are difficult to crystallize, so that identification of factors that assist crystallisation could open up the structure determination of these more challenging targets. Our findings suggest that protein crystallisation success may be improved by matching a protein with its optimal plate make.


Asunto(s)
Cristalización/métodos , Proteínas/química , Animales , Antígenos/química , Catalasa/química , Bovinos , Pollos , Cristalografía por Rayos X/instrumentación , Cristalografía por Rayos X/métodos , Clara de Huevo/química , Escherichia coli/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Hígado/enzimología , Ratones , Muramidasa/química , Proyectos Piloto , Streptomyces/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA