Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 171(7): 1638-1648.e7, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29224781

RESUMEN

Cleavage of membrane-anchored proteins by ADAM (a disintegrin and metalloproteinase) endopeptidases plays a key role in a wide variety of biological signal transduction and protein turnover processes. Among ADAM family members, ADAM10 stands out as particularly important because it is both responsible for regulated proteolysis of Notch receptors and catalyzes the non-amyloidogenic α-secretase cleavage of the Alzheimer's precursor protein (APP). We present here the X-ray crystal structure of the ADAM10 ectodomain, which, together with biochemical and cellular studies, reveals how access to the enzyme active site is regulated. The enzyme adopts an unanticipated architecture in which the C-terminal cysteine-rich domain partially occludes the enzyme active site, preventing unfettered substrate access. Binding of a modulatory antibody to the cysteine-rich domain liberates the catalytic domain from autoinhibition, enhancing enzymatic activity toward a peptide substrate. Together, these studies reveal a mechanism for regulation of ADAM activity and offer a roadmap for its modulation.


Asunto(s)
Proteína ADAM10/química , Secretasas de la Proteína Precursora del Amiloide/química , Proteínas de la Membrana/química , Proteolisis , Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Cristalografía por Rayos X , Humanos , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Receptores Notch/metabolismo , Transducción de Señal
2.
Genes Dev ; 29(8): 785-90, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25838500

RESUMEN

The amyloid precursor protein (APP) has garnered considerable attention due to its genetic links to Alzheimer's disease. Death receptor 6 (DR6) was recently shown to bind APP via the protein extracellular regions, stimulate axonal pruning, and inhibit synapse formation. Here, we report the crystal structure of the DR6 ectodomain in complex with the E2 domain of APP and show that it supports a model for APP-induced dimerization and activation of cell surface DR6.


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Moleculares , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Cristalización , Dimerización , Células HEK293 , Humanos , Ratones , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Transducción de Señal
3.
Proc Natl Acad Sci U S A ; 116(41): 20707-20715, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31548390

RESUMEN

Cedar virus (CedV) is a bat-borne henipavirus related to Nipah virus (NiV) and Hendra virus (HeV), zoonotic agents of fatal human disease. CedV receptor-binding protein (G) shares only ∼30% sequence identity with those of NiV and HeV, although they can all use ephrin-B2 as an entry receptor. We demonstrate that CedV also enters cells through additional B- and A-class ephrins (ephrin-B1, ephrin-A2, and ephrin-A5) and report the crystal structure of the CedV G ectodomain alone and in complex with ephrin-B1 or ephrin-B2. The CedV G receptor-binding site is structurally distinct from other henipaviruses, underlying its capability to accommodate additional ephrin receptors. We also show that CedV can enter cells through mouse ephrin-A1 but not human ephrin-A1, which differ by 1 residue in the key contact region. This is evidence of species specific ephrin receptor usage by a henipavirus, and implicates additional ephrin receptors in potential zoonotic transmission.


Asunto(s)
Efrina-B1/metabolismo , Efrina-B2/metabolismo , Efrina-B3/metabolismo , Infecciones por Henipavirus/virología , Henipavirus/fisiología , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/química , Animales , Fusión Celular , Efrina-B1/genética , Efrina-B2/genética , Efrina-B3/genética , Infecciones por Henipavirus/genética , Infecciones por Henipavirus/metabolismo , Humanos , Ratones , Mutación , Unión Proteica , Conformación Proteica , Receptores Virales/genética , Especificidad de la Especie , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
4.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34638814

RESUMEN

The Eph receptor tyrosine kinases and their ephrin ligands direct axon pathfinding and neuronal cell migration, as well as mediate many other cell-cell communication events. Their dysfunctional signaling has been shown to lead to various diseases, including cancer. The Ephs and ephrins both localize to the plasma membrane and, upon cell-cell contact, form extensive signaling assemblies at the contact sites. The Ephs and the ephrins are divided into A and B subclasses based on their sequence conservation and affinities for each other. The molecular details of Eph-ephrin recognition have been previously revealed and it has been documented that ephrin binding induces higher-order Eph assemblies, which are essential for full biological activity, via multiple, distinct Eph-Eph interfaces. One Eph-Eph interface type is characterized by a homotypic, head-to-tail interaction between the ligand-binding and the fibronectin domains of two adjacent Eph molecules. While the previous Eph ectodomain structural studies were focused on A class receptors, we now report the crystal structure of the full ectodomain of EphB2, revealing distinct and unique head-to-tail receptor-receptor interactions. The EphB2 structure and structure-based mutagenesis document that EphB2 uses the head-to-tail interactions as a novel autoinhibitory control mechanism for regulating downstream signaling and that these interactions can be modulated by posttranslational modifications.


Asunto(s)
Receptor EphB2/química , Receptor EphB2/metabolismo , Transducción de Señal , Animales , Células HEK293 , Humanos , Ratones , Dominios Proteicos , Receptor EphB2/genética , Relación Estructura-Actividad
5.
Mol Cell ; 37(5): 643-55, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20227369

RESUMEN

The Tie family of endothelial-specific receptor tyrosine kinases is essential for cell proliferation, migration, and survival during angiogenesis. Despite considerable similarity, experiments with Tie1- or Tie2-deficient mice highlight distinct functions for these receptors in vivo. The Tie2 receptor is further unique with respect to its structurally homologous ligands. Angiopoietin-2 and -3 can function as agonists or antagonists; angiopoietin-1 and -4 are constitutive agonists. To address the role of Tie1 in angiopoietin-mediated Tie2 signaling and determine the basis for the behavior of the individual angiopoietins, we used an in vivo FRET-based proximity assay to monitor Tie1 and -2 localization and association. We provide evidence for Tie1-Tie2 complex formation on the cell surface and identify molecular surface areas essential for receptor-receptor recognition. We further demonstrate that the Tie1-Tie2 interactions are dynamic, inhibitory, and differentially modulated by angiopoietin-1 and -2. Based on the available data, we propose a unified model for angiopoietin-induced Tie2 signaling.


Asunto(s)
Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Células Endoteliales/enzimología , Receptor TIE-1/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal , Línea Celular , Membrana Celular/enzimología , Transferencia Resonante de Energía de Fluorescencia , Humanos , Ligandos , Modelos Moleculares , Mutación , Conformación Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Interferencia de ARN , Receptor Cross-Talk , Receptor TIE-1/química , Receptor TIE-1/genética , Receptor TIE-2/química , Receptor TIE-2/genética , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Factores de Tiempo , Transfección
6.
PLoS Pathog ; 11(12): e1005322, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26646856

RESUMEN

Nipah virus (NiV) is a paramyxovirus that infects host cells through the coordinated efforts of two envelope glycoproteins. The G glycoprotein attaches to cell receptors, triggering the fusion (F) glycoprotein to execute membrane fusion. Here we report the first crystal structure of the pre-fusion form of the NiV-F glycoprotein ectodomain. Interestingly this structure also revealed a hexamer-of-trimers encircling a central axis. Electron tomography of Nipah virus-like particles supported the hexameric pre-fusion model, and biochemical analyses supported the hexamer-of-trimers F assembly in solution. Importantly, structure-assisted site-directed mutagenesis of the interfaces between F trimers highlighted the functional relevance of the hexameric assembly. Shown here, in both cell-cell fusion and virus-cell fusion systems, our results suggested that this hexamer-of-trimers assembly was important during fusion pore formation. We propose that this assembly would stabilize the pre-fusion F conformation prior to cell attachment and facilitate the coordinated transition to a post-fusion conformation of all six F trimers upon triggering of a single trimer. Together, our data reveal a novel and functional pre-fusion architecture of a paramyxoviral fusion glycoprotein.


Asunto(s)
Infecciones por Henipavirus/metabolismo , Virus Nipah/química , Proteínas del Envoltorio Viral/química , Internalización del Virus , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Virus Nipah/metabolismo , Conformación Proteica , Proteínas del Envoltorio Viral/metabolismo
7.
Proc Natl Acad Sci U S A ; 110(36): 14634-9, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959867

RESUMEN

Eph receptor tyrosine kinases and their ephrin ligands mediate cell signaling during normal and oncogenic development. Eph signaling is initiated in a multistep process leading to the assembly of higher-order Eph/ephrin clusters that set off bidirectional signaling in interacting cells. Eph and ephrins are divided in two subclasses based on their abilities to bind and activate each other and on sequence conservation. EphA4 is an exception to the general rule because it can be activated by both A- and B-class ephrin ligands. Here we present high-resolution structures of the complete EphA4 ectodomain and its complexes with ephrin-A5. The structures reveal how ligand binding promotes conformational changes in the EphA4 ligand-binding domain allowing the formation of signaling clusters at the sites of cell-cell contact. In addition, the structural data, combined with structure-based mutagenesis, reveal a previously undescribed receptor-receptor interaction between the EphA4 ligand-binding and membrane-proximal fibronectin domains, which is functionally important for efficient receptor activation.


Asunto(s)
Efrina-A5/química , Estructura Terciaria de Proteína , Receptor EphA4/química , Transducción de Señal , Sitios de Unión/genética , Western Blotting , Cristalografía por Rayos X , Activación Enzimática , Efrina-A5/genética , Efrina-A5/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Receptor EphA4/genética , Receptor EphA4/metabolismo
8.
Proc Natl Acad Sci U S A ; 110(18): 7205-10, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23592718

RESUMEN

Angiogenesis is a complex cellular process involving multiple regulatory growth factors and growth factor receptors. Among them, the ligands for the endothelial-specific tunica intima endothelial receptor tyrosine kinase 2 (Tie2) receptor kinase, angiopoietin-1 (Ang1) and Ang2, play essential roles in balancing vessel stability and regression during both developmental and tumor-induced angiogenesis. Despite possessing a high degree of sequence identity, Ang1 and Ang2 have distinct functional roles and cell-signaling characteristics. Here, we present the crystal structures of Ang1 both unbound and in complex with the Tie2 ectodomain. Comparison of the Ang1-containing structures with their Ang2-containing counterparts provide insight into the mechanism of receptor activation and reveal molecular surfaces important for interactions with Tie2 coreceptors and associated signaling proteins. Using structure-based mutagenesis, we identify a loop within the angiopoietin P domain, adjacent to the receptor-binding interface, which confers the specific agonist/antagonist properties of the molecule. We demonstrate using cell-based assays that an Ang2 chimera containing the Ang1 loop sequence behaves functionally similarly to Ang1 as a constitutive Tie2 agonist, able to efficiently dissociate the inhibitory Tie1/Tie2 complex and elicit Tie2 clustering and downstream signaling.


Asunto(s)
Angiopoyetina 1/química , Angiopoyetina 1/metabolismo , Transducción de Señal , Angiopoyetina 2/química , Angiopoyetina 2/metabolismo , Secuencia Conservada , Cristalografía por Rayos X , Células HEK293 , Humanos , Modelos Moleculares , Estructura Terciaria de Proteína , Receptor TIE-1/química , Receptor TIE-1/metabolismo , Receptor TIE-2/química , Receptor TIE-2/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Electricidad Estática , Relación Estructura-Actividad
9.
PLoS Pathog ; 9(10): e1003684, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24130486

RESUMEN

The henipaviruses, represented by Hendra (HeV) and Nipah (NiV) viruses are highly pathogenic zoonotic paramyxoviruses with uniquely broad host tropisms responsible for repeated outbreaks in Australia, Southeast Asia, India and Bangladesh. The high morbidity and mortality rates associated with infection and lack of licensed antiviral therapies make the henipaviruses a potential biological threat to humans and livestock. Henipavirus entry is initiated by the attachment of the G envelope glycoprotein to host cell membrane receptors. Previously, henipavirus-neutralizing human monoclonal antibodies (hmAb) have been isolated using the HeV-G glycoprotein and a human naïve antibody library. One cross-reactive and receptor-blocking hmAb (m102.4) was recently demonstrated to be an effective post-exposure therapy in two animal models of NiV and HeV infection, has been used in several people on a compassionate use basis, and is currently in development for use in humans. Here, we report the crystal structure of the complex of HeV-G with m102.3, an m102.4 derivative, and describe NiV and HeV escape mutants. This structure provides detailed insight into the mechanism of HeV and NiV neutralization by m102.4, and serves as a blueprint for further optimization of m102.4 as a therapeutic agent and for the development of entry inhibitors and vaccines.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/química , Virus Hendra/química , Proteínas Virales de Fusión/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Línea Celular , Reacciones Cruzadas , Cristalografía por Rayos X , Virus Hendra/genética , Virus Hendra/inmunología , Infecciones por Henipavirus/genética , Infecciones por Henipavirus/inmunología , Humanos , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
10.
Semin Cell Dev Biol ; 23(1): 116-23, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22227101

RESUMEN

Members of the ephrin cell-surface protein family interact with the Eph receptors, the largest family of receptor tyrosine kinases, mediating bi-directional signaling during tumorogenesis and various developmental events. Surprisingly, ephrin-B2 and -B3 were recently identified as entry receptors for henipaviruses, emerging zoonotic paramyxoviruses responsible for repeated outbreaks in humans and animals in Australia, Southeast Asia, India and Bangladesh. Nipah virus (NiV) and Hendra virus (HeV) are the only two identified members in the henipavirus genus. While the initial human infection cases came from contact with infected pigs (NiV) or horses (HeV), in the more recent outbreaks of NiV both food-borne and human-to-human transmission were reported. These characteristics, together with high mortality and morbidity rates and lack of effective anti-viral therapies, make the henipaviruses a potential biological-agent threat. Viral entry is an important target for the development of anti-viral drugs. The entry of henipavirus is initiated by the attachment of the viral G envelope glycoprotein to the host cell receptors ephrin-B2 and/or -B3, followed by activation of the F fusion protein, which triggers fusion between the viral envelop and the host membrane. We review recent progress in the study of henipavirus entry, particularly the identification of ephrins as their entry receptors, and the structural characterization of the ephrin/Henipa-G interactions.


Asunto(s)
Efrina-B2/metabolismo , Efrina-B3/metabolismo , Henipavirus/fisiología , Receptores Virales/metabolismo , Acoplamiento Viral , Animales , Anticuerpos Neutralizantes/química , Efrina-B2/química , Efrina-B3/química , Henipavirus/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Transducción de Señal , Proteínas Virales/química , Proteínas Virales/metabolismo
11.
J Biol Chem ; 288(25): 18448-57, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23661698

RESUMEN

The EphA2 receptor tyrosine kinase is overexpressed in a number of malignancies and is activated by ephrin ligands, most commonly by ephrin-A1. The crystal structure of the ligand-receptor complex revealed a glycosylation on the Asn-26 of ephrin-A1. Here we report for the first time the significance of the glycosylation in the biology of EphA2 and ephrin-A1. Ephrin-A1 was enzymatically deglycosylated, and its activity was evaluated in several assays using glioblastoma (GBM) cells and recombinant EphA2. We found that deglycosylated ephrin-A1 does not efficiently induce EphA2 receptor internalization and degradation, and does not activate the downstream signaling pathways involved in cell migration and proliferation. Data obtained by surface plasmon resonance confirms that deglycosylated ephrin-A1 does not bind EphA2 with high affinity. Mutations in the glycosylation site on ephrin-A1 result in protein aggregation and mislocalization. Analysis of Eph/ephrin crystal structures reveals an interaction between the ligand's carbohydrates and two residues of EphA2: Asp-78 and Lys-136. These findings suggest that the glycosylation on ephrin-A1 plays a critical role in the binding and activation of the EphA2 receptor.


Asunto(s)
Efrina-A1/metabolismo , Receptor EphA2/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Sitios de Unión/genética , Unión Competitiva , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Cristalografía por Rayos X , Efrina-A1/química , Efrina-A1/genética , Efrina-A2/genética , Efrina-A2/metabolismo , Glicosilación , Humanos , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Receptor EphA2/química , Receptor EphA2/genética , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
12.
Biochim Biophys Acta ; 1834(10): 2160-5, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23628727

RESUMEN

The Eph receptors and their ephrin ligands play crucial roles in a large number of cell-cell interaction events, including those associated with axon pathfinding, neuronal cell migration and vasculogenesis. They are also involved in the patterning of most tissues and overall cell positioning in the development of the vertebrate body plan. The Eph/ephrin signaling system manifests several unique features that differentiate it from other receptor tyrosine kinases, including initiation of bi-directional signaling cascades and the existence of ligand and receptor subclasses displaying promiscuous intra-subclass interactions, but very rare inter-subclass interactions. In this review we briefly discuss these features and focus on recent studies of the unique and expansive high-affinity Eph/ephrin assemblies that form at the sites of cell-cell contact and are required for Eph signaling initiation. This article is part of a Special Issue entitled: Emerging recognition and activation mechanisms of receptor tyrosine kinases.


Asunto(s)
Comunicación Celular/fisiología , Efrinas/metabolismo , Receptores de la Familia Eph/metabolismo , Transducción de Señal , Adhesión Celular , Línea Celular , Movimiento Celular , Análisis por Conglomerados , Efrinas/química , Efrinas/genética , Regulación de la Expresión Génica , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Receptores de la Familia Eph/química , Receptores de la Familia Eph/genética
13.
Growth Factors ; 32(6): 214-22, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25494541

RESUMEN

Recombinant antibody phage library technology provides multiple advantages, including that human antibodies can be generated against proteins that are highly conserved between species. We used this technology to isolate and characterize an anti-EphA2 single-chain antibody. We show that the antibody binds the antigen with 1:1 stoichiometry and has high specificity for EphA2. The crystal structure of the complex reveals that the antibody targets the same receptor surface cavity as the ephrin ligand. Specifically, a lengthy CDR-H3 loop protrudes deep into the ligand-binding cavity, with several hydrophobic residues at its tip forming an anchor-like structure buried within the hydrophobic Eph pocket, in a way similar to the ephrin receptor-binding loop in the Eph/ephrin structures. Consequently, the antibody blocks ephrin binding to EphA2. Furthermore, it induces apoptosis and reduces cell proliferation in lymphoma cells lines. Since Ephs are important mediators of tumorigenesis, such antibodies could have applications both in research and therapy.


Asunto(s)
Receptor EphA2/inmunología , Anticuerpos de Cadena Única/inmunología , Secuencia de Aminoácidos , Animales , Afinidad de Anticuerpos , Sitios de Unión de Anticuerpos , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Humanos , Datos de Secuencia Molecular , Anticuerpos de Cadena Única/química
14.
J Cell Sci ; 125(Pt 24): 6084-93, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23108669

RESUMEN

The ADAM10 transmembrane metalloprotease cleaves a variety of cell surface proteins that are important in disease, including ligands for receptor tyrosine kinases of the erbB and Eph families. ADAM10-mediated cleavage of ephrins, the ligands for Eph receptors, is suggested to control Eph/ephrin-mediated cell-cell adhesion and segregation, important during normal developmental processes, and implicated in tumour neo-angiogenesis and metastasis. We previously identified a substrate-binding pocket in the ADAM10 C domain that binds the EphA/ephrin-A complex thereby regulating ephrin cleavage. We have now generated monoclonal antibodies specifically recognising this region of ADAM10, which inhibit ephrin cleavage and Eph/ephrin-mediated cell function, including ephrin-induced Eph receptor internalisation, phosphorylation and Eph-mediated cell segregation. Our studies confirm the important role of ADAM10 in cell-cell interactions mediated by both A- and B-type Eph receptors, and suggest antibodies against the ADAM10 substrate-recognition pocket as promising therapeutic agents, acting by inhibiting cleavage of ephrins and potentially other ADAM10 substrates.


Asunto(s)
Proteínas ADAM/metabolismo , Anticuerpos Monoclonales/metabolismo , Efrinas/metabolismo , Receptores de la Familia Eph/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Sitios de Unión de Anticuerpos , Bovinos , Adhesión Celular , Células HEK293 , Humanos , Ratones , Unión Proteica , Transducción de Señal
15.
Curr Opin Cell Biol ; 19(5): 534-42, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17928214

RESUMEN

Eph receptors are the largest subfamily of receptor tyrosine kinases regulating cell shape, movements, and attachment. The interactions of the Ephs with their ephrin ligands are restricted to the sites of cell-cell contact since both molecules are membrane attached. This review summarizes recent advances in our understanding of the molecular mechanisms underlining the diverse functions of the molecules during development and in the adult organism. The unique properties of this signaling system that are of highest interest and have been the focus of intense investigations are as follows: (i) the signal is simultaneously transduced in both ligand-expressing cells and receptor-expressing cells, (ii) signaling via the same molecules can generate opposing cellular reactions depending on the context, and (iii) the Ephs and the ephrins are divided into two subclasses with promiscuous intrasubclass interactions, but rarely observed intersubclass interactions.


Asunto(s)
Comunicación Celular/fisiología , Efrinas/metabolismo , Receptor EphA1/metabolismo , Transducción de Señal/fisiología , Adhesión Celular/fisiología , Endocitosis/fisiología , Activación Enzimática , Inhibidores Enzimáticos/metabolismo , Efrinas/ultraestructura , Humanos , Sistema Nervioso , Péptidos/metabolismo , Estructura Terciaria de Proteína , Receptor EphA1/ultraestructura
16.
FASEB J ; 27(2): 437-45, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23070608

RESUMEN

Recovery from stroke is limited, in part, by an inhibitory environment in the postischemic brain, but factors preventing successful remodeling are not well known. Using cultured cortical neurons from mice, brain endothelial cells, and a mouse model of ischemic stroke, we show that signaling from the axon guidance molecule Sema3A via eicosanoid second messengers can contribute to this inhibitory environment. Either 90 nM recombinant Sema3A, or the 12/15-lipoxygenase (12/15-LOX) metabolites 12-HETE and 12-HPETE at 300 nM, block axon extension in neurons compared to solvent controls, and decrease tube formation in endothelial cells. The Sema3A effect is reversed by inhibiting 12/15-LOX, and neurons derived from 12/15-LOX-knockout mice are insensitive to Sema3A. Following middle cerebral artery occlusion to induce stroke in mice, immunohistochemistry shows both Sema3A and 12/15-LOX are increased in the cortex up to 2 wk. To determine whether a Sema3A-dependent damage pathway is activated following ischemia, we injected recombinant Sema3A into the striatum. Sema3A alone did not cause injury in normal brains. But when injected into postischemic brains, Sema3A increased cortical damage by 79%, and again, this effect was reversed by 12/15-LOX inhibition. Our findings suggest that blocking the semaphorin pathway should be investigated as a therapeutic strategy to improve stroke recovery.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Encéfalo/metabolismo , Semaforina-3A/metabolismo , Accidente Cerebrovascular/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/metabolismo , Animales , Araquidonato 12-Lipooxigenasa/deficiencia , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/deficiencia , Araquidonato 15-Lipooxigenasa/genética , Encéfalo/irrigación sanguínea , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Inmunohistoquímica , Leucotrienos/metabolismo , Masculino , Ratones , Ratones Noqueados , Neovascularización Fisiológica , Neuronas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sistemas de Mensajero Secundario , Semaforina-3A/antagonistas & inhibidores , Semaforina-3A/genética , Transducción de Señal , Accidente Cerebrovascular/patología
17.
J Virol ; 86(21): 11457-71, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22915804

RESUMEN

The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are paramyxoviruses discovered in the mid- to late 1990s that possess a broad host tropism and are known to cause severe and often fatal disease in both humans and animals. HeV and NiV infect cells by a pH-independent membrane fusion mechanism facilitated by their attachment (G) and fusion (F) glycoproteins. Here, several soluble forms of henipavirus F (sF) were engineered and characterized. Recombinant sF was produced by deleting the transmembrane (TM) and cytoplasmic tail (CT) domains and appending a glycosylphosphatidylinositol (GPI) anchor signal sequence followed by GPI-phospholipase D digestion, appending a trimeric coiled-coil (GCNt) domain (sF(GCNt)), or deleting the TM, CT, and fusion peptide domain. These sF glycoproteins were produced as F(0) precursors, and all were apparent stable trimers recognized by NiV-specific antisera. Surprisingly, however, only the GCNt-appended constructs (sF(GCNt)) could elicit cross-reactive henipavirus-neutralizing antibody in mice. In addition, sF(GCNt) constructs could be triggered in vitro by protease cleavage and heat to transition from an apparent prefusion to postfusion conformation, transitioning through an intermediate that could be captured by a peptide corresponding to the C-terminal heptad repeat domain of F. The pre- and postfusion structures of sF(GCNt) and non-GCNt-appended sF could be revealed by electron microscopy and were distinguishable by F-specific monoclonal antibodies. These data suggest that only certain sF constructs could serve as potential subunit vaccine immunogens against henipaviruses and also establish important tools for further structural, functional, and diagnostic studies on these important emerging viruses.


Asunto(s)
Henipavirus/inmunología , Henipavirus/ultraestructura , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/ultraestructura , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Reacciones Cruzadas , Células HeLa , Henipavirus/genética , Humanos , Ratones , Microscopía Electrónica , Conformación Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo
18.
Curr Top Microbiol Immunol ; 359: 197-223, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22481140

RESUMEN

Hendra virus and Nipah virus are recently discovered and closely related emerging viruses that now comprise the genus henipavirus within the sub-family Paramyxoviridae and are distinguished by their broad species tropism and in addition to bats can infect and cause fatal disease in a wide variety of mammalian hosts including humans. The high mortality associated with human and animal henipavirus infections has highlighted the importance and necessity of developing effective immunization strategies. The development of suitable animal models of henipavirus infection and pathogenesis has been critical for testing the efficacy of potential therapeutic approaches. Several henipavirus challenge models have been used and recent successes in both active and passive immunization strategies against henipaviruses have been reported which have all targeted the viral envelope glycoproteins.


Asunto(s)
Virus Hendra/inmunología , Infecciones por Henipavirus/prevención & control , Inmunización Pasiva , Virus Nipah/inmunología , Vacunación , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Virus Hendra/patogenicidad , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/patología , Humanos , Virus Nipah/patogenicidad , Vacunas de Subunidad , Vacunas Sintéticas , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/biosíntesis , Tropismo Viral , Vacunas Virales/administración & dosificación , Vacunas Virales/biosíntesis
19.
Proc Natl Acad Sci U S A ; 107(24): 10860-5, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20505120

RESUMEN

Eph receptor tyrosine kinases and their ephrin ligands regulate cell navigation during normal and oncogenic development. Signaling of Ephs is initiated in a multistep process leading to the assembly of higher-order signaling clusters that set off bidirectional signaling in interacting cells. However, the structural and mechanistic details of this assembly remained undefined. Here we present high-resolution structures of the complete EphA2 ectodomain and complexes with ephrin-A1 and A5 as the base unit of an Eph cluster. The structures reveal an elongated architecture with novel Eph/Eph interactions, both within and outside of the Eph ligand-binding domain, that suggest the molecular mechanism underlying Eph/ephrin clustering. Structure-function analysis, by using site-directed mutagenesis and cell-based signaling assays, confirms the importance of the identified oligomerization interfaces for Eph clustering.


Asunto(s)
Receptor EphA1/química , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Efrina-A1/química , Efrina-A1/genética , Efrina-A1/metabolismo , Efrina-A5/química , Efrina-A5/genética , Efrina-A5/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor EphA1/genética , Receptor EphA1/metabolismo , Receptor EphA2/química , Receptor EphA2/genética , Receptor EphA2/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
20.
Microorganisms ; 11(12)2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38138084

RESUMEN

The viral agent SARS-CoV-2 clearly affects several organ systems, including the cardiovascular system. Angiopoietins are involved in vascular integrity and angiogenesis. Angiopoietin-1 (Ang1) promotes vessel stabilization, while angiopoietin-2 (Ang2), which is usually expressed at low levels, is significantly elevated in inflammatory and angiogenic conditions. Interleukin-6 (IL-6) is known to induce defective angiogenesis via the activation of the Ang2 pathway. Vasculitis and vasculopathy are some of the defining features of moderate to severe COVID-19-associated systemic disease. We investigated the serum levels of angiopoietins, as well as interleukin-6 levels and anti-SARS-CoV2 IgG titers, in hospitalized COVID-19 patients across disease severity and healthy controls. Ang2 levels were elevated in COVID-19 patients across all severity compared to healthy controls, while Ang1 levels were decreased. The patients with adverse outcomes (death and/or prolonged hospitalization) had relatively lower and stable Ang1 levels but continuously elevated Ang2 levels, while those who had no adverse outcomes had increasing levels of both Ang1 and Ang2, followed by a decrease in both. These results suggest that the dynamic levels of Ang1 and Ang2 during the clinical course may predict adverse outcomes in COVID-19 patients. Ang1 seems to play an important role in controlling Ang2-related inflammatory mechanisms in COVID-19 patients. IL-6 and anti-SARS-CoV2 spike protein IgG levels were significantly elevated in patients with severe disease. Our findings represent an informative pilot assessment into the role of the angiopoietin signaling pathway in the inflammatory response in COVID-19.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA