Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Chembiochem ; 23(1): e202100463, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34647407

RESUMEN

Vascular endothelial growth factors (VEGFs) regulate significant pathways in angiogenesis, myocardial and neuronal protection, metabolism, and cancer progression. The VEGF-B growth factor is involved in cell survival, anti-apoptotic and antioxidant mechanisms, through binding to VEGF receptor 1 and neuropilin-1 (NRP1). We employed surface plasmon resonance technology and X-ray crystallography to analyse the molecular basis of the interaction between VEGF-B and the b1 domain of NRP1, and developed VEGF-B C-terminus derived peptides to be used as chemical tools for studying VEGF-B - NRP1 related pathways. Peptide lipidation was used as a means to stabilise the peptides. VEGF-B-derived peptides containing a C-terminal arginine show potent binding to NRP1-b1. Peptide lipidation increased binding residence time and improved plasma stability. A crystal structure of a peptide with NRP1 demonstrated that VEGF-B peptides bind at the canonical C-terminal arginine binding site. VEGF-B C-terminus imparts higher affinity for NRP1 than the corresponding VEGF-A165 region. This tight binding may impact on the activity and selectivity of the full-length protein. The VEGF-B167 derived peptides were more effective than VEGF-A165 peptides in blocking functional phosphorylation events. Blockers of VEGF-B function have potential applications in diabetes and non-alcoholic fatty liver disease.


Asunto(s)
Neuropilina-1/metabolismo , Péptidos/metabolismo , Factor B de Crecimiento Endotelial Vascular/metabolismo , Humanos , Neuropilina-1/química , Péptidos/química , Unión Proteica , Factor B de Crecimiento Endotelial Vascular/química
2.
Biochemistry ; 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34130453

RESUMEN

Two paralogs of the guanine dissociation inhibitor-like solubilizing factors UNC119, UNC119A and UNC119B, are present in the human genome. UNC119 binds to N-myristoylated proteins and masks the hydrophobic lipid from the hydrophilic cytosol, facilitating trafficking between different membranes. Two classes of UNC119 cargo proteins have been classified: low affinity cargoes, released by the Arf-like proteins ARL2 and ARL3, and high affinity cargoes, which are specifically released by ARL3 and trafficked to either the primary cilium or the immunological synapse. The UNC119 homologues have reported differences in functionality, but the structural and biochemical bases for these differences are unknown. Using myristoylated peptide binding and release assays, we show that peptides sharing the previously identified UNC119A high affinity motif show significant variations of binding affinities to UNC119B of up to 427-fold. Furthermore, we solve the first two crystal structures of UNC119B, one in complex with the high affinity cargo peptide of LCK and a second one in complex with the release factor ARL3. Using these novel structures, we identify a stretch of negatively charged amino acids unique to UNC119B that may undergo a conformational change following binding of a release factor which we propose as an additional release mechanism specific to UNC119B.

3.
Glycoconj J ; 33(3): 387-97, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26059692

RESUMEN

Neuropilins are involved in angiogenesis and neuronal development. The membrane proximal domain of neuropilin-1, called c or MAM domain based on its sequence conservation, has been implicated in neuropilin oligomerization required for its function. The c/MAM domain of human neuropilin-1 has been recombinantly expressed to allow for investigation of its propensity to engage in molecular interactions with other protein or carbohydrate components on a cell surface. We found that the c/MAM domain was heavily O-glycosylated with up to 24 monosaccharide units in the form of disialylated core 1 and core 2 O-glycans. Attachment sites were identified on the chymotryptic c/MAM peptide ETGATEKPTVIDSTIQSEFPTY by electron-transfer dissociation mass spectrometry (ETD-MS/MS). For highly glycosylated species consisting of carbohydrate to about 50 %, useful results could only be obtained upon partial desialylation. ETD-MS/MS revealed a hierarchical order of the initial O-GalNAc addition to the four different glycosylation sites. These findings enable future functional studies about the contribution of the described glycosylations in neuropilin-1 oligomerization and the binding to partner proteins as VEGF or galectin-1.As a spin-off result the sialidase from Clostridium perfringens turned out to discriminate between galactose- and N-acetylgalactosamine-linked sialic acid.


Asunto(s)
Neuropilina-1/química , Procesamiento Proteico-Postraduccional , Acetilglucosamina/análogos & derivados , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Glicosilación , Células HEK293 , Humanos , Neuropilina-1/metabolismo , Dominios Proteicos
4.
Biomolecules ; 12(3)2022 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-35327564

RESUMEN

Vascular endothelial growth factors (VEGFs) are the key regulators of blood and lymphatic vessels' formation and function. Each of the proteins from the homologous family VEGFA, VEGFB, VEGFC and VEGFD employs a core cysteine-knot structural domain for the specific interaction with one or more of the cognate tyrosine kinase receptors. Additional diversity is exhibited by the involvement of neuropilins-transmembrane co-receptors, whose b1 domain contains the binding site for the C-terminal sequence of VEGFs. Although all relevant isoforms of VEGFs that interact with neuropilins contain the required C-terminal Arg residue, there is selectivity of neuropilins and VEGF receptors for the VEGF proteins, which is reflected in the physiological roles that they mediate. To decipher the contribution made by the C-terminal sequences of the individual VEGF proteins to that functional differentiation, we determined structures of molecular complexes of neuropilins and VEGF-derived peptides and examined binding interactions for all neuropilin-VEGF pairs experimentally and computationally. While X-ray crystal structures and ligand-binding experiments highlighted similarities between the ligands, the molecular dynamics simulations uncovered conformational preferences of VEGF-derived peptides beyond the C-terminal arginine that contribute to the ligand selectivity of neuropilins. The implications for the design of the selective antagonists of neuropilins' functions are discussed.


Asunto(s)
Neuropilinas , Factor A de Crecimiento Endotelial Vascular , Ligandos , Neuropilinas/química , Neuropilinas/genética , Neuropilinas/metabolismo , Péptidos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular
5.
J Med Chem ; 65(3): 1898-1914, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35104933

RESUMEN

RAS is a major anticancer drug target which requires membrane localization to activate downstream signal transduction. The direct inhibition of RAS has proven to be challenging. Here, we present a novel strategy for targeting RAS by stabilizing its interaction with the prenyl-binding protein PDE6D and disrupting its localization. Using rationally designed RAS point mutations, we were able to stabilize the RAS:PDE6D complex by increasing the affinity of RAS for PDE6D, which resulted in the redirection of RAS to the cytoplasm and the primary cilium and inhibition of oncogenic RAS/ERK signaling. We developed an SPR fragment screening and identified fragments that bind at the KRAS:PDE6D interface, as shown through cocrystal structures. Finally, we show that the stoichiometric ratios of KRAS:PDE6D vary in different cell lines, suggesting that the impact of this strategy might be cell-type-dependent. This study forms the foundation from which a potential anticancer small-molecule RAS:PDE6D complex stabilizer could be developed.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/análisis , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
6.
Elife ; 102021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33438581

RESUMEN

The ADP-ribosylation factor-like 3 (ARL3) is a ciliopathy G-protein which regulates the ciliary trafficking of several lipid-modified proteins. ARL3 is activated by its guanine exchange factor (GEF) ARL13B via an unresolved mechanism. BART is described as an ARL3 effector which has also been implicated in ciliopathies, although the role of its ARL3 interaction is unknown. Here, we show that, at physiological GTP:GDP levels, human ARL3GDP is weakly activated by ARL13B. However, BART interacts with nucleotide-free ARL3 and, in concert with ARL13B, efficiently activates ARL3. In addition, BART binds ARL3GTP and inhibits GTP dissociation, thereby stabilising the active G-protein; the binding of ARL3 effectors then releases BART. Finally, using live cell imaging, we show that BART accesses the primary cilium and colocalises with ARL13B. We propose a model wherein BART functions as a bona fide co-GEF for ARL3 and maintains the active ARL3GTP, until it is recycled by ARL3 effectors.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Cilios/metabolismo , Factores de Transcripción/genética , Factores de Ribosilacion-ADP/metabolismo , Animales , Guanina/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ratones , Células 3T3 NIH , Factores de Transcripción/metabolismo
7.
Structure ; 29(3): 226-237.e4, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33217330

RESUMEN

Rac1 is a major regulator of actin dynamics, with GTP-bound Rac1 promoting actin assembly via the Scar/WAVE complex. CYRI competes with Scar/WAVE for interaction with Rac1 in a feedback loop regulating actin dynamics. Here, we reveal the nature of the CYRI-Rac1 interaction, through crystal structures of CYRI-B lacking the N-terminal helix (CYRI-BΔN) and the CYRI-BΔN:Rac1Q61L complex, providing the molecular basis for CYRI-B regulation of the Scar/WAVE complex. We reveal CYRI-B as having two subdomains - an N-terminal Rac1 binding subdomain with a unique Rac1-effector interface and a C-terminal Ratchet subdomain that undergoes conformational changes induced by Rac1 binding. Finally, we show that the CYRI protein family, CYRI-A and CYRI-B can produce an autoinhibited hetero- or homodimers, adding an additional layer of regulation to Rac1 signaling.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Proteínas Mitocondriales/química , Proteína de Unión al GTP rac1/química , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Secuencia Conservada , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , Simulación del Acoplamiento Molecular , Unión Proteica , Proteína de Unión al GTP rac1/metabolismo
8.
J Cell Biol ; 220(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34165494

RESUMEN

The Scar/WAVE complex drives actin nucleation during cell migration. Interestingly, the same complex is important in forming membrane ruffles during macropinocytosis, a process mediating nutrient uptake and membrane receptor trafficking. Mammalian CYRI-B is a recently described negative regulator of the Scar/WAVE complex by RAC1 sequestration, but its other paralogue, CYRI-A, has not been characterized. Here, we implicate CYRI-A as a key regulator of macropinosome formation and integrin internalization. We find that CYRI-A is transiently recruited to nascent macropinosomes, dependent on PI3K and RAC1 activity. CYRI-A recruitment precedes RAB5A recruitment but follows sharply after RAC1 and actin signaling, consistent with it being a local inhibitor of actin polymerization. Depletion of both CYRI-A and -B results in enhanced surface expression of the α5ß1 integrin via reduced internalization. CYRI depletion enhanced migration, invasion, and anchorage-independent growth in 3D. Thus, CYRI-A is a dynamic regulator of macropinocytosis, functioning together with CYRI-B to regulate integrin trafficking.


Asunto(s)
Endosomas/metabolismo , Integrina alfa5beta1/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Mitocondriales/genética , Pinocitosis/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética , Actinas/genética , Actinas/metabolismo , Animales , Células COS , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Chlorocebus aethiops , Endosomas/patología , Endosomas/ultraestructura , Regulación de la Expresión Génica , Células HEK293 , Humanos , Integrina alfa5beta1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Mitocondriales/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Polimerizacion , Transporte de Proteínas , Transducción de Señal , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
9.
Nat Commun ; 12(1): 1623, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712589

RESUMEN

The signalling pathways underpinning cell growth and invasion use overlapping components, yet how mutually exclusive cellular responses occur is unclear. Here, we report development of 3-Dimensional culture analyses to separately quantify growth and invasion. We identify that alternate variants of IQSEC1, an ARF GTPase Exchange Factor, act as switches to promote invasion over growth by controlling phosphoinositide metabolism. All IQSEC1 variants activate ARF5- and ARF6-dependent PIP5-kinase to promote PI(3,4,5)P3-AKT signalling and growth. In contrast, select pro-invasive IQSEC1 variants promote PI(3,4,5)P3 production to form invasion-driving protrusions. Inhibition of IQSEC1 attenuates invasion in vitro and metastasis in vivo. Induction of pro-invasive IQSEC1 variants and elevated IQSEC1 expression occurs in a number of tumour types and is associated with higher-grade metastatic cancer, activation of PI(3,4,5)P3 signalling, and predicts long-term poor outcome across multiple cancers. IQSEC1-regulated phosphoinositide metabolism therefore is a switch to induce invasion over growth in response to the same external signal. Targeting IQSEC1 as the central regulator of this switch may represent a therapeutic vulnerability to stop metastasis.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Metástasis de la Neoplasia , Fosfatidilinositoles/metabolismo , Transducción de Señal , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular Tumoral , Factores de Intercambio de Guanina Nucleótido/genética , Xenoinjertos , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/genética , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
10.
Dev Cell ; 47(1): 122-132.e4, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30220567

RESUMEN

Upon engagement of the T cell receptor with an antigen-presenting cell, LCK initiates TCR signaling by phosphorylating its activation motifs. However, the mechanism of LCK activation specifically at the immune synapse is a major question. We show that phosphorylation of the LCK activating Y394, despite modestly increasing its catalytic rate, dramatically focuses LCK localization to the immune synapse. We describe a trafficking mechanism whereby UNC119A extracts membrane-bound LCK by sequestering the hydrophobic myristoyl group, followed by release at the target membrane under the control of the ciliary ARL3/ARL13B. The UNC119A N terminus acts as a "regulatory arm" by binding the LCK kinase domain, an interaction inhibited by LCK Y394 phosphorylation, thus together with the ARL3/ARL13B machinery ensuring immune synapse focusing of active LCK. We propose that the ciliary machinery has been repurposed by T cells to generate and maintain polarized segregation of signals such as activated LCK at the immune synapse.


Asunto(s)
Cilios/fisiología , Sinapsis Inmunológicas/fisiología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Presentadoras de Antígenos/inmunología , Humanos , Células Jurkat , Activación de Linfocitos , Fosforilación , Transporte de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/fisiología
11.
FEBS J ; 285(7): 1290-1304, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29430837

RESUMEN

Neuropilin-1 (NRP1) is a transmembrane co-receptor involved in binding interactions with variety of ligands and receptors, including receptor tyrosine kinases. Expression of NRP1 in several cancers correlates with cancer stages and poor prognosis. Thus, NRP1 has been considered a therapeutic target and is the focus of multiple drug discovery initiatives. Vascular endothelial growth factor (VEGF) binds to the b1 domain of NRP1 through interactions between the C-terminal arginine of VEGF and residues in the NRP1-binding site including Tyr297, Tyr353, Asp320, Ser346 and Thr349. We obtained several complexes of the synthetic ligands and the NRP1-b1 domain and used X-ray crystallography and computational methods to analyse atomic details and hydration profile of this binding site. We observed side chain flexibility for Tyr297 and Asp320 in the six new high-resolution crystal structures of arginine analogues bound to NRP1. In addition, we identified conserved water molecules in binding site regions which can be targeted for drug design. The computational prediction of the VEGF ligand-binding site hydration map of NRP1 was in agreement with the experimentally derived, conserved hydration structure. Displacement of certain conserved water molecules by a ligand's functional groups may contribute to binding affinity, whilst other water molecules perform as protein-ligand bridges. Our report provides a comprehensive description of the binding site for the peptidic ligands' C-terminal arginines in the b1 domain of NRP1, highlights the importance of conserved structural waters in drug design and validates the utility of the computational hydration map prediction method in the context of neuropilin. DATABASE: The structures were deposited to the PDB with accession numbers PDB ID: 5IJR, 5IYY, 5JHK, 5J1X, 5JGQ, 5JGI.


Asunto(s)
Arginina/química , Neuropilina-1/química , Arginina/metabolismo , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Humanos , Hidrógeno/química , Ligandos , Modelos Biológicos , Estructura Molecular , Neuropilina-1/metabolismo , Resonancia por Plasmón de Superficie
12.
J Med Chem ; 61(9): 4135-4154, 2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29648813

RESUMEN

We report the design, synthesis, and biological evaluation of some potent small-molecule neuropilin-1 (NRP1) antagonists. NRP1 is implicated in the immune response to tumors, particularly in Treg cell fragility, required for PD1 checkpoint blockade. The design of these compounds was based on a previously identified compound EG00229. The design of these molecules was informed and supported by X-ray crystal structures. Compound 1 (EG01377) was identified as having properties suitable for further investigation. Compound 1 was then tested in several in vitro assays and was shown to have antiangiogenic, antimigratory, and antitumor effects. Remarkably, 1 was shown to be selective for NRP1 over the closely related protein NRP2. In purified Nrp1+, FoxP3+, and CD25+ populations of Tregs from mice, 1 was able to block a glioma-conditioned medium-induced increase in TGFß production. This comprehensive characterization of a small-molecule NRP1 antagonist provides the basis for future in vivo studies.


Asunto(s)
Inmunomodulación/efectos de los fármacos , Neuropilina-1/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Ratones , Modelos Moleculares , Conformación Molecular , Ácidos Pentanoicos/química , Ácidos Pentanoicos/farmacología , Bibliotecas de Moléculas Pequeñas/química , Linfocitos T Reguladores/inmunología , Factor A de Crecimiento Endotelial Vascular/farmacología
13.
Structure ; 24(11): 2008-2015, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27720589

RESUMEN

Neuropilins (NRPs) are single-pass transmembrane receptors involved in several signaling pathways that regulate key physiological processes such as vascular morphogenesis and axon guidance. The MAM domain of NRP, which has previously been implicated in receptor multimerization, was the only portion of the ectopic domain of the NRPs for which the structure, until now, has been elusive. Using site-directed mutagenesis in the linker region preceding the MAM domain we generated a protein construct amenable to crystallization. Here we present the crystal structure of the MAM domain of human NRP1 at 2.24 Å resolution. The protein exhibits a jellyroll topology, with Ca2+ ions bound at the inter-strand space enhancing the thermostability of the domain. We show that the MAM domain of NRP1 is monomeric in solution and insufficient to drive receptor dimerization, which leads us to propose a different role for this domain in the context of NRP membrane assembly and signaling.


Asunto(s)
Calcio/metabolismo , Mutagénesis Sitio-Dirigida/métodos , Neuropilina-1/química , Neuropilina-1/genética , Sitios de Unión , Cristalografía por Rayos X , Proteínas de Unión al ADN/metabolismo , Dimerización , Humanos , Modelos Moleculares , Neuropilina-1/metabolismo , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Factores de Transcripción/metabolismo
14.
FEBS J ; 283(10): 1921-34, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26991001

RESUMEN

Neuropilin-2 is a transmembrane receptor involved in lymphangiogenesis and neuronal development. In adults, neuropilin-2 and its homologous protein neuropilin-1 have been implicated in cancers and infection. Molecular determinants of the ligand selectivity of neuropilins are poorly understood. We have identified and structurally characterized a zinc ion binding site on human neuropilin-2. The neuropilin-2-specific zinc ion binding site is located near the interface between domains b1 and b2 in the ectopic region of the protein, remote from the neuropilin binding site for its physiological ligand, i.e. vascular endothelial growth factor. We also present an X-ray crystal structure of the neuropilin-2 b1 domain in a complex with the C-terminal sub-domain of VEGF-A. Zn(2+) binding to neuropilin-2 destabilizes the protein structure but this effect was counteracted by heparin, suggesting that modifications by glycans and zinc in the extracellular matrix may affect functional neuropilin-2 ligand binding and signalling activity.


Asunto(s)
Neuropilina-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Zinc/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Humanos , Neuropilina-2/química , Conformación Proteica , Homología de Secuencia de Aminoácido
15.
J Mol Biol ; 426(18): 3134-3147, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25020226

RESUMEN

CPE (Clostridium perfringens enterotoxin) is the major virulence determinant for C. perfringens type-A food poisoning, the second most common bacterial food-borne illness in the UK and USA. After binding to its receptors, which include particular human claudins, the toxin forms pores in the cell membrane. The mature pore apparently contains a hexamer of CPE, claudin and, possibly, occludin. The combination of high binding specificity with cytotoxicity has resulted in CPE being investigated, with some success, as a targeted cytotoxic agent for oncotherapy. In this paper, we present the X-ray crystallographic structure of CPE in complex with a peptide derived from extracellular loop 2 of a modified, CPE-binding Claudin-2, together with high-resolution native and pore-formation mutant structures. Our structure provides the first atomic-resolution data on any part of a claudin molecule and reveals that claudin's CPE-binding fingerprint (NPLVP) is in a tight turn conformation and binds, as expected, in CPE's C-terminal claudin-binding groove. The leucine and valine residues insert into the binding groove while the first residue, asparagine, tethers the peptide via an interaction with CPE's aspartate 225 and the two prolines are required to maintain the tight turn conformation. Understanding the structural basis of the contribution these residues make to binding will aid in engineering CPE to target tumor cells.


Asunto(s)
Claudina-2/química , Clostridium perfringens/química , Enterotoxinas/química , Modelos Moleculares , Sustitución de Aminoácidos , Claudina-2/metabolismo , Clostridium perfringens/genética , Clostridium perfringens/aislamiento & purificación , Clostridium perfringens/metabolismo , Cristalografía por Rayos X , Enterotoxinas/genética , Enterotoxinas/aislamiento & purificación , Enterotoxinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Mutación , Péptidos/genética , Péptidos/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA