Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
1.
Alzheimers Res Ther ; 11(1): 97, 2019 12 01.
Article de Anglais | MEDLINE | ID: mdl-31787113

RÉSUMÉ

BACKGROUND: Accumulation of amyloid ß (Aß) in the brain is proposed as a cause of Alzheimer's disease (AD), with Aß oligomers hypothesized to be the primary mediators of neurotoxicity. Crenezumab is a humanized immunoglobulin G4 monoclonal antibody that has been shown to bind to synthetic monomeric and aggregated Aß in vitro; however, less is known about the binding characteristic in vivo. In this study, we evaluated the binding patterns of crenezumab to synthetic and native forms of Aß both in vitro and in vivo. METHODS: Crenezumab was used to immunoprecipitate Aß from synthetic Aß preparations or brain homogenates from a PS2APP mouse model of AD to determine the forms of Aß that crenezumab interacts with. Following systemic dosing in PS2APP or nontransgenic control mice, immunohistochemistry was used to localize crenezumab and assess its relative distribution in the brain, compared with amyloid plaques and markers of neuritic dystrophies (BACE1; LAMP1). Pharmacodynamic correlations were performed to investigate the relationship between peripheral and central target engagement. RESULTS: In vitro, crenezumab immunoprecipitated Aß oligomers from both synthetic Aß preparations and endogenous brain homogenates from PS2APP mice. In vivo studies in the PS2APP mouse showed that crenezumab localizes to regions surrounding the periphery of amyloid plaques in addition to the hippocampal mossy fibers. These regions around the plaques are reported to be enriched in oligomeric Aß, actively incorporate soluble Aß, and contribute to Aß-induced neurotoxicity and axonal dystrophy. In addition, crenezumab did not appear to bind to the dense core region of plaques or vascular amyloid. CONCLUSIONS: Crenezumab binds to multiple forms of amyloid ß (Aß), particularly oligomeric forms, and localizes to brain areas rich in Aß oligomers, including the halo around plaques and hippocampal mossy fibers, but not to vascular Aß. These insights highlight a unique mechanism of action for crenezumab of engaging Aß oligomers.


Sujet(s)
Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Anticorps monoclonaux humanisés/pharmacologie , Encéphale/effets des médicaments et des substances chimiques , Animaux , Encéphale/métabolisme , Souris , Souris transgéniques , Plaque amyloïde/métabolisme , Liaison aux protéines
2.
Br J Pharmacol ; 174(22): 4173-4185, 2017 Nov.
Article de Anglais | MEDLINE | ID: mdl-28859225

RÉSUMÉ

BACKGROUND AND PURPOSE: The potential for therapeutic antibody treatment of neurological diseases is limited by poor penetration across the blood-brain barrier. I.c.v. delivery is a promising route to the brain; however, it is unclear how efficiently antibodies delivered i.c.v. penetrate the cerebrospinal spinal fluid (CSF)-brain barrier and distribute throughout the brain parenchyma. EXPERIMENTAL APPROACH: We evaluated the pharmacokinetics and pharmacodynamics of an inhibitory monoclonal antibody against ß-secretase 1 (anti-BACE1) following continuous infusion into the left lateral ventricle of healthy adult cynomolgus monkeys. KEY RESULTS: Animals infused with anti-BACE1 i.c.v. showed a robust and sustained reduction (~70%) of CSF amyloid-ß (Aß) peptides. Antibody distribution was near uniform across the brain parenchyma, ranging from 20 to 40 nM, resulting in a ~50% reduction of Aß in the cortical parenchyma. In contrast, animals administered anti-BACE1 i.v. showed no significant change in CSF or cortical Aß levels and had a low (~0.6 nM) antibody concentration in the brain. CONCLUSION AND IMPLICATIONS: I.c.v. administration of anti-BACE1 resulted in enhanced BACE1 target engagement and inhibition, with a corresponding dramatic reduction in CNS Aß concentrations, due to enhanced brain exposure to antibody.


Sujet(s)
Amyloid precursor protein secretases/antagonistes et inhibiteurs , Peptides bêta-amyloïdes/métabolisme , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/pharmacocinétique , Aspartic acid endopeptidases/antagonistes et inhibiteurs , Amyloid precursor protein secretases/immunologie , Peptides bêta-amyloïdes/sang , Peptides bêta-amyloïdes/liquide cérébrospinal , Animaux , Anticorps monoclonaux/sang , Anticorps monoclonaux/liquide cérébrospinal , Aspartic acid endopeptidases/immunologie , Encéphale/métabolisme , Femelle , Perfusions intraventriculaires , Macaca fascicularis
3.
Sci Transl Med ; 9(403)2017 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-28814543

RÉSUMÉ

Hallmarks of chronic neurodegenerative disease include progressive synaptic loss and neuronal cell death, yet the cellular pathways that underlie these processes remain largely undefined. We provide evidence that dual leucine zipper kinase (DLK) is an essential regulator of the progressive neurodegeneration that occurs in amyotrophic lateral sclerosis and Alzheimer's disease. We demonstrate that DLK/c-Jun N-terminal kinase signaling was increased in mouse models and human patients with these disorders and that genetic deletion of DLK protected against axon degeneration, neuronal loss, and functional decline in vivo. Furthermore, pharmacological inhibition of DLK activity was sufficient to attenuate the neuronal stress response and to provide functional benefit even in the presence of ongoing disease. These findings demonstrate that pathological activation of DLK is a conserved mechanism that regulates neurodegeneration and suggest that DLK inhibition may be a potential approach to treat multiple neurodegenerative diseases.


Sujet(s)
Glissières à leucine , MAP Kinase Kinase Kinases/métabolisme , Maladies neurodégénératives/enzymologie , Maladies neurodégénératives/anatomopathologie , Transduction du signal , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/anatomopathologie , Sclérose latérale amyotrophique/enzymologie , Sclérose latérale amyotrophique/anatomopathologie , Animaux , Modèles animaux de maladie humaine , Délétion de gène , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , JNK Mitogen-Activated Protein Kinases/métabolisme , Système de signalisation des MAP kinases , Souris transgéniques , Neuroprotection , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Moelle spinale/enzymologie , Moelle spinale/anatomopathologie , Superoxide dismutase/métabolisme
4.
Sci Rep ; 7: 44249, 2017 03 10.
Article de Anglais | MEDLINE | ID: mdl-28281673

RÉSUMÉ

Assessing BACE1 (ß-site APP cleaving enzyme 1) knockout mice for general health and neurological function may be useful in predicting risks associated with prolonged pharmacological BACE1 inhibition, a treatment approach currently being developed for Alzheimer's disease. To determine whether BACE1 deletion-associated effects in mice generalize to another species, we developed a novel Bace1-/- rat line using zinc-finger nuclease technology and compared Bace1-/- mice and rats with their Bace1+/+ counterparts. Lack of BACE1 was confirmed in Bace1-/- animals from both species. Removal of BACE1 affected startle magnitude, balance beam performance, pain response, and nerve myelination in both species. While both mice and rats lacking BACE1 have shown increased mortality, the increase was smaller and restricted to early developmental stages for rats. Bace1-/- mice and rats further differed in body weight, spontaneous locomotor activity, and prepulse inhibition of startle. While the effects of species and genetic background on these phenotypes remain difficult to distinguish, our findings suggest that BACE1's role in myelination and some sensorimotor functions is consistent between mice and rats and may be conserved in other species. Other phenotypes differ between these models, suggesting that some effects of BACE1 inhibition vary with the biological context (e.g. species or background strain).


Sujet(s)
Amyloid precursor protein secretases/génétique , Aspartic acid endopeptidases/génétique , Délétion de gène , Réflexe de sursaut/génétique , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/physiopathologie , Amyloid precursor protein secretases/métabolisme , Animaux , Aspartic acid endopeptidases/métabolisme , Poids/génétique , Poids/physiologie , Humains , Souris de lignée C57BL , Souris knockout , Activité motrice/génétique , Activité motrice/physiologie , Inhibition du réflexe de sursaut/génétique , Inhibition du réflexe de sursaut/physiologie , Rats , Réflexe de sursaut/physiologie , Spécificité d'espèce
5.
Nat Med ; 20(12): 1452-7, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25419706

RÉSUMÉ

We have identified a rare coding mutation, T835M (rs137875858), in the UNC5C netrin receptor gene that segregated with disease in an autosomal dominant pattern in two families enriched for late-onset Alzheimer's disease and that was associated with disease across four large case-control cohorts (odds ratio = 2.15, Pmeta = 0.0095). T835M alters a conserved residue in the hinge region of UNC5C, and in vitro studies demonstrate that this mutation leads to increased cell death in human HEK293T cells and in rodent neurons. Furthermore, neurons expressing T835M UNC5C are more susceptible to cell death from multiple neurotoxic stimuli, including ß-amyloid (Aß), glutamate and staurosporine. On the basis of these data and the enriched hippocampal expression of UNC5C in the adult nervous system, we propose that one possible mechanism in which T835M UNC5C contributes to the risk of Alzheimer's disease is by increasing susceptibility to neuronal cell death, particularly in vulnerable regions of the Alzheimer's disease brain.


Sujet(s)
Maladie d'Alzheimer/génétique , Neurones/métabolisme , Récepteurs de surface cellulaire/génétique , Récepteurs facteur croissance nerf/génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Peptides bêta-amyloïdes , Animaux , Région CA3 de l'hippocampe/cytologie , Mort cellulaire/génétique , Femelle , Prédisposition génétique à une maladie , Acide glutamique , Cellules HEK293 , Humains , Mâle , Souris , Récepteurs de la nétrine , Rats , Staurosporine
6.
Sci Transl Med ; 6(261): 261ra154, 2014 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-25378646

RÉSUMÉ

Using therapeutic antibodies that need to cross the blood-brain barrier (BBB) to treat neurological disease is a difficult challenge. We have shown that bispecific antibodies with optimized binding to the transferrin receptor (TfR) that target ß-secretase (BACE1) can cross the BBB and reduce brain amyloid-ß (Aß) in mice. Can TfR enhance antibody uptake in the primate brain? We describe two humanized TfR/BACE1 bispecific antibody variants. Using a human TfR knock-in mouse, we observed that anti-TfR/BACE1 antibodies could cross the BBB and reduce brain Aß in a TfR affinity-dependent fashion. Intravenous dosing of monkeys with anti-TfR/BACE1 antibodies also reduced Aß both in cerebral spinal fluid and in brain tissue, and the degree of reduction correlated with the brain concentration of anti-TfR/BACE1 antibody. These results demonstrate that the TfR bispecific antibody platform can robustly and safely deliver therapeutic antibody across the BBB in the primate brain.


Sujet(s)
Amyloid precursor protein secretases/immunologie , Anticorps bispécifiques/pharmacocinétique , Antigènes CD/immunologie , Aspartic acid endopeptidases/immunologie , Barrière hémato-encéphalique/métabolisme , Perméabilité capillaire , Récepteurs à la transferrine/immunologie , Administration par voie intraveineuse , Amyloid precursor protein secretases/antagonistes et inhibiteurs , Amyloid precursor protein secretases/métabolisme , Peptides bêta-amyloïdes/liquide cérébrospinal , Animaux , Anticorps bispécifiques/administration et posologie , Anticorps bispécifiques/sang , Anticorps bispécifiques/immunologie , Spécificité des anticorps , Antigènes CD/génétique , Antigènes CD/métabolisme , Aspartic acid endopeptidases/antagonistes et inhibiteurs , Aspartic acid endopeptidases/métabolisme , Transport biologique , Cellules CHO , Cricetulus , Réactions croisées , Régulation négative , Cellules HEK293 , Humains , Macaca fascicularis , Souris de lignée BALB C , Souris de lignée C57BL , Souris transgéniques , Fragments peptidiques/liquide cérébrospinal , Récepteurs à la transferrine/génétique , Récepteurs à la transferrine/métabolisme , Transfection
7.
J Biol Chem ; 289(45): 30990-1000, 2014 Nov 07.
Article de Anglais | MEDLINE | ID: mdl-25253696

RÉSUMÉ

Pathogenic mutations in the amyloid precursor protein (APP) gene have been described as causing early onset familial Alzheimer disease (AD). We recently identified a rare APP variant encoding an alanine-to-threonine substitution at residue 673 (A673T) that confers protection against development of AD (Jonsson, T., Atwal, J. K., Steinberg, S., Snaedal, J., Jonsson, P. V., Bjornsson, S., Stefansson, H., Sulem, P., Gudbjartsson, D., Maloney, J., Hoyte, K., Gustafson, A., Liu, Y., Lu, Y., Bhangale, T., Graham, R. R., Huttenlocher, J., Bjornsdottir, G., Andreassen, O. A., Jönsson, E. G., Palotie, A., Behrens, T. W., Magnusson, O. T., Kong, A., Thorsteinsdottir, U., Watts, R. J., and Stefansson, K. (2012) Nature 488, 96-99). The Ala-673 residue lies within the ß-secretase recognition sequence and is part of the amyloid-ß (Aß) peptide cleavage product (position 2 of Aß). We previously demonstrated that the A673T substitution makes APP a less favorable substrate for cleavage by BACE1. In follow-up studies, we confirm that A673T APP shows reduced cleavage by BACE1 in transfected mouse primary neurons and in isogenic human induced pluripotent stem cell-derived neurons. Using a biochemical approach, we show that the A673T substitution modulates the catalytic turnover rate (V(max)) of APP by the BACE1 enzyme, without affecting the affinity (K(m)) of the APP substrate for BACE1. We also show a reduced level of Aß(1-42) aggregation with A2T Aß peptides, an observation not conserved in Aß(1-40) peptides. When combined in a ratio of 1:9 Aß(1-42)/Aß(1-40) to mimic physiologically relevant mixtures, A2T retains a trend toward slowed aggregation kinetics. Microglial uptake of the mutant Aß(1-42) peptides correlated with their aggregation level. Cytotoxicity of the mutant Aß peptides was not dramatically altered. Taken together, our findings demonstrate that A673T, a protective allele of APP, reproducibly reduces amyloidogenic processing of APP and also mildly decreases Aß aggregation. These effects could together have an additive or even synergistic impact on the risk of developing AD.


Sujet(s)
Maladie d'Alzheimer/génétique , Précurseur de la protéine bêta-amyloïde/génétique , Allèles , Amyloid precursor protein secretases/métabolisme , Peptides bêta-amyloïdes/génétique , Animaux , Aspartic acid endopeptidases/métabolisme , Catalyse , ADN complémentaire/métabolisme , Transfert d'énergie par résonance de fluorescence , Cellules HEK293 , Hétérozygote , Humains , Concentration inhibitrice 50 , Cinétique , Souris , Souris de lignée C57BL , Microglie/métabolisme , Mutation , Neurones/métabolisme , Fragments peptidiques/génétique , Liaison aux protéines
8.
J Neurosci ; 32(39): 13439-53, 2012 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-23015435

RÉSUMÉ

In addition to being a hallmark of neurodegenerative disease, axon degeneration is used during development of the nervous system to prune unwanted connections. In development, axon degeneration is tightly regulated both temporally and spatially. Here, we provide evidence that degeneration cues are transduced through various kinase pathways functioning in spatially distinct compartments to regulate axon degeneration. Intriguingly, glycogen synthase kinase-3 (GSK3) acts centrally, likely modulating gene expression in the cell body to regulate distally restricted axon degeneration. Through a combination of genetic and pharmacological manipulations, including the generation of an analog-sensitive kinase allele mutant mouse for GSK3ß, we show that the ß isoform of GSK3, not the α isoform, is essential for developmental axon pruning in vitro and in vivo. Additionally, we identify the dleu2/mir15a/16-1 cluster, previously characterized as a regulator of B-cell proliferation, and the transcription factor tbx6, as likely downstream effectors of GSK3ß in axon degeneration.


Sujet(s)
Axones/métabolisme , Glycogen Synthase Kinase 3/métabolisme , Dégénérescence nerveuse/enzymologie , Dégénérescence nerveuse/anatomopathologie , Neurones/anatomopathologie , Phosphotransferases/métabolisme , Transduction du signal/physiologie , Animaux , Animaux nouveau-nés , Cellules cultivées , Électroporation , Embryon de mammifère , Antienzymes/pharmacologie , Femelle , Ganglions sensitifs des nerfs spinaux/cytologie , Analyse de profil d'expression de gènes/méthodes , Régulation de l'expression des gènes au cours du développement/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes au cours du développement/génétique , Régulation de l'expression des gènes au cours du développement/physiologie , Génotype , Glycogen Synthase Kinase 3/génétique , Glycogen synthase kinase 3 beta , Protéines à fluorescence verte/génétique , Hippocampe/cytologie , Humains , Immunoprécipitation , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/génétique , Système de signalisation des MAP kinases/physiologie , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Mutation/génétique , Dégénérescence nerveuse/traitement médicamenteux , Dégénérescence nerveuse/prévention et contrôle , Facteur de croissance nerveuse/déficit , Protéines de tissu nerveux/métabolisme , Séquençage par oligonucléotides en batterie , Techniques de culture d'organes , Phosphorylation/physiologie , Petit ARN interférent/administration et posologie , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Cellules ganglionnaires rétiniennes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transfection ,
9.
Nature ; 488(7409): 96-9, 2012 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-22801501

RÉSUMÉ

The prevalence of dementia in the Western world in people over the age of 60 has been estimated to be greater than 5%, about two-thirds of which are due to Alzheimer's disease. The age-specific prevalence of Alzheimer's disease nearly doubles every 5 years after age 65, leading to a prevalence of greater than 25% in those over the age of 90 (ref. 3). Here, to search for low-frequency variants in the amyloid-ß precursor protein (APP) gene with a significant effect on the risk of Alzheimer's disease, we studied coding variants in APP in a set of whole-genome sequence data from 1,795 Icelanders. We found a coding mutation (A673T) in the APP gene that protects against Alzheimer's disease and cognitive decline in the elderly without Alzheimer's disease. This substitution is adjacent to the aspartyl protease ß-site in APP, and results in an approximately 40% reduction in the formation of amyloidogenic peptides in vitro. The strong protective effect of the A673T substitution against Alzheimer's disease provides proof of principle for the hypothesis that reducing the ß-cleavage of APP may protect against the disease. Furthermore, as the A673T allele also protects against cognitive decline in the elderly without Alzheimer's disease, the two may be mediated through the same or similar mechanisms.


Sujet(s)
Vieillissement/génétique , Maladie d'Alzheimer/génétique , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Troubles de la cognition/génétique , Troubles de la cognition/physiopathologie , Mutation/génétique , Allèles , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/physiopathologie , Maladie d'Alzheimer/prévention et contrôle , Amyloid precursor protein secretases/métabolisme , Précurseur de la protéine bêta-amyloïde/composition chimique , Aspartic acid endopeptidases/métabolisme , Cognition/physiologie , Troubles de la cognition/prévention et contrôle , Prédisposition génétique à une maladie , Cellules HEK293 , Humains , Plaque amyloïde/génétique , Plaque amyloïde/métabolisme
10.
J Neurosci ; 32(28): 9677-89, 2012 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-22787053

RÉSUMÉ

Passive immunization against ß-amyloid (Aß) has become an increasingly desirable strategy as a therapeutic treatment for Alzheimer's disease (AD). However, traditional passive immunization approaches carry the risk of Fcγ receptor-mediated overactivation of microglial cells, which may contribute to an inappropriate proinflammatory response leading to vasogenic edema and cerebral microhemorrhage. Here, we describe the generation of a humanized anti-Aß monoclonal antibody of an IgG4 isotype, known as MABT5102A (MABT). An IgG4 subclass was selected to reduce the risk of Fcγ receptor-mediated overactivation of microglia. MABT bound with high affinity to multiple forms of Aß, protected against Aß1-42 oligomer-induced cytotoxicity, and increased uptake of neurotoxic Aß oligomers by microglia. Furthermore, MABT-mediated amyloid plaque removal was demonstrated using in vivo live imaging in hAPP((V717I))/PS1 transgenic mice. When compared with a human IgG1 wild-type subclass, containing the same antigen-binding variable domains and with equal binding to Aß, MABT showed reduced activation of stress-activated p38MAPK (p38 mitogen-activated protein kinase) in microglia and induced less release of the proinflammatory cytokine TNFα. We propose that a humanized IgG4 anti-Aß antibody that takes advantage of a unique Aß binding profile, while also possessing reduced effector function, may provide a safer therapeutic alternative for passive immunotherapy for AD. Data from a phase I clinical trial testing MABT is consistent with this hypothesis, showing no signs of vasogenic edema, even in ApoE4 carriers.


Sujet(s)
Maladie d'Alzheimer/thérapie , Peptides bêta-amyloïdes/immunologie , Immunoglobuline G/pharmacologie , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Neuroprotecteurs/pharmacologie , Fragments peptidiques/métabolisme , Sujet âgé , Sujet âgé de 80 ans ou plus , Maladie d'Alzheimer/sang , Maladie d'Alzheimer/immunologie , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Animaux , Animaux nouveau-nés , Récepteur-1 de la chimiokine CX3C , Cellules cultivées , Cortex cérébral/cytologie , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Relation dose-réponse (immunologie) , Méthode en double aveugle , Test ELISA , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/génétique , Régulation de l'expression des gènes/immunologie , Protéines à fluorescence verte/génétique , Hippocampe/cytologie , Humains , Immunoglobuline G/métabolisme , Mâle , Souris , Souris transgéniques , Microscopie confocale , Adulte d'âge moyen , Mutation/génétique , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neuroprotecteurs/métabolisme , Plaque amyloïde/immunologie , Plaque amyloïde/métabolisme , Plaque amyloïde/anatomopathologie , Préséniline-1/génétique , Liaison aux protéines/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Récepteurs aux chimiokines/génétique , Statistique non paramétrique , Facteurs temps , Facteur de nécrose tumorale alpha/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme
11.
EMBO J ; 26(23): 4902-12, 2007 Nov 28.
Article de Anglais | MEDLINE | ID: mdl-17989695

RÉSUMÉ

Neuropilins (Nrps) are co-receptors for class 3 semaphorins and vascular endothelial growth factors and important for the development of the nervous system and the vasculature. The extracellular portion of Nrp is composed of two domains that are essential for semaphorin binding (a1a2), two domains necessary for VEGF binding (b1b2), and one domain critical for receptor dimerization (c). We report several crystal structures of Nrp1 and Nrp2 fragments alone and in complex with antibodies that selectively block either semaphorin or vascular endothelial growth factor (VEGF) binding. In these structures, Nrps adopt an unexpected domain arrangement in which the a2, b1, and b2 domains form a tightly packed core that is only loosely connected to the a1 domain. The locations of the antibody epitopes together with in vitro experiments indicate that VEGF and semaphorin do not directly compete for Nrp binding. Based upon our structural and functional data, we propose possible models for ligand binding to neuropilins.


Sujet(s)
Neuropilines/composition chimique , Sémaphorine-3A/composition chimique , Facteur de croissance endothéliale vasculaire de type A/composition chimique , Séquence d'acides aminés , Anticorps/composition chimique , Sites de fixation , Cristallographie aux rayons X/méthodes , Dimérisation , Conformation moléculaire , Données de séquences moléculaires , Neuropilines/physiologie , Liaison aux protéines , Conformation des protéines , Structure tertiaire des protéines , Sémaphorine-3A/métabolisme , Sémaphorines/métabolisme , Similitude de séquences d'acides aminés , Facteur de croissance endothéliale vasculaire de type A/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE