Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 8317, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110403

RESUMEN

In this study, we characterize Designed Ankyrin Repeat Proteins (DARPins) as investigative tools to probe botulinum neurotoxin A1 (BoNT/A1) structure and function. We identify DARPin-F5 that completely blocks SNAP25 substrate cleavage by BoNT/A1 in vitro. X-ray crystallography reveals that DARPin-F5 inhibits BoNT/A1 activity by interacting with a substrate-binding region between the α- and ß-exosite. This DARPin does not block substrate cleavage of BoNT/A3, indicating that DARPin-F5 is a subtype-specific inhibitor. BoNT/A1 Glu-171 plays a critical role in the interaction with DARPin-F5 and its mutation to Asp, the residue found in BoNT/A3, results in a loss of inhibition of substrate cleavage. In contrast to the in vitro results, DARPin-F5 promotes faster substrate cleavage of BoNT/A1 in primary neurons and muscle tissue by increasing toxin translocation. Our findings could have important implications for the application of BoNT/A1 in therapeutic areas requiring faster onset of toxin action combined with long persistence.


Asunto(s)
Toxinas Botulínicas Tipo A , Toxinas Botulínicas , Clostridium botulinum , Proteínas de Repetición de Anquirina Diseñadas , Toxinas Botulínicas Tipo A/metabolismo , Clostridium botulinum/genética
2.
Sci Rep ; 13(1): 10159, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37349348

RESUMEN

Structure elucidation of inactive-state GPCRs still mostly relies on X-ray crystallography. The major goal of our work was to create a new GPCR tool that would provide receptor stability and additional soluble surface for crystallization. Towards this aim, we selected the two-stranded antiparallel coiled coil as a domain fold that satisfies both criteria. A selection of antiparallel coiled coils was used for structure-guided substitution of intracellular loop 3 of the ß3 adrenergic receptor. Unexpectedly, only the two GPCR variants containing thermostable coiled coils were expressed. We showed that one GPCR chimera is stable upon purification in detergent, retains ligand-binding properties, and can be crystallized. However, the quality of the crystals was not suitable for structure determination. By using two other examples, 5HTR2C and α2BAR, we demonstrate that our approach is generally suitable for the stabilization of GPCRs. To provide additional surface for promoting crystal contacts, we replaced in a structure-based approach the loop connecting the antiparallel coiled coil by T4L. We found that the engineered GPCR is even more stable than the coiled-coil variant. Negative-staining TEM revealed a homogeneous distribution of particles, indicating that coiled-coil-T4L receptor variants might also be promising candidate proteins for structure elucidation by cryo-EM. Our approach should be of interest for applications that benefit from stable GPCRs.


Asunto(s)
Receptores Acoplados a Proteínas G , Secuencia de Aminoácidos , Estructura Secundaria de Proteína , Cristalografía por Rayos X , Dominios Proteicos , Receptores Acoplados a Proteínas G/genética
3.
J Clin Invest ; 131(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34618682

RESUMEN

We used human monoclonal antibodies (humAbs) to study the mechanism of neuron intoxication by tetanus neurotoxin and to evaluate these antibodies as a safe preventive and therapeutic substitute for hyperimmune sera to treat tetanus in mice. By screening memory B cells from immune donors, we selected 2 tetanus neurotoxin-specific mAbs with exceptionally high neutralizing activities and extensively characterized them both structurally and functionally. We found that these antibodies interfered with the binding and translocation of the neurotoxin into neurons by interacting with 2 epitopes, whose identification pinpoints crucial events in the cellular pathogenesis of tetanus. Our observations explain the neutralization ability of these antibodies, which we found to be exceptionally potent in preventing experimental tetanus when injected into mice long before the toxin. Moreover, their Fab derivatives neutralized tetanus neurotoxin in post-exposure experiments, suggesting their potential for therapeutic use via intrathecal injection. As such, we believe these humAbs, as well as their Fab derivatives, meet the requirements to be considered for prophylactic and therapeutic use in human tetanus and are ready for clinical trials.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Metaloendopeptidasas/antagonistas & inhibidores , Toxina Tetánica/antagonistas & inhibidores , Tétanos/prevención & control , Adulto , Animales , Anticuerpos Monoclonales/química , Complejo Antígeno-Anticuerpo/química , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Metaloendopeptidasas/química , Ratones , Conformación Proteica , Ratas , Tétanos/tratamiento farmacológico , Toxina Tetánica/química
4.
J Biol Chem ; 296: 100684, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33891946

RESUMEN

Botulinum neurotoxins (BoNTs) are among the most widely used therapeutic proteins; however, only two subtypes within the seven serotypes, BoNT/A1 and BoNT/B1, are currently used for medical and cosmetic applications. Distinct catalytic properties, substrate specificities, and duration of enzymatic activities potentially make other subtypes very attractive candidates to outperform conventional BoNTs in particular therapeutic applications. For example, BoNT/A3 has a significantly shorter duration of action than other BoNT/A subtypes. Notably, BoNT/A3 is the subtype with the least conserved catalytic domain among BoNT/A subtypes. This suggests that the sequence differences, many of which concern the α-exosite, contribute to the observed functional differences in toxin persistence by affecting the binding of the substrate SNAP-25 and/or the stability of the catalytic domain fold. To identify the molecular determinants accounting for the differences in the persistence observed for BoNT/A subtypes, we determined the crystal structure of the catalytic domain of BoNT/A3 (LC/A3). The structure of LC/A3 was found to be very similar to that of LC/A1, suggesting that the overall mode of SNAP-25 binding is common between these two proteins. However, circular dichroism (CD) thermal unfolding experiments demonstrated that LC/A3 is significantly less stable than LC/A1, implying that this might contribute to the reduced toxin persistence of BoNT/A3. These findings could be of interest in developing next-generation therapeutic toxins.


Asunto(s)
Toxinas Botulínicas Tipo A/química , Dominio Catalítico , Secuencia de Aminoácidos , Toxinas Botulínicas Tipo A/metabolismo , Cristalografía por Rayos X , Modelos Moleculares , Especificidad por Sustrato
5.
Toxicon ; 175: 36-43, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31783045

RESUMEN

A dual-receptor interaction with a polysialoganglioside and synaptic vesicle glycoprotein 2 (SV2) is required for botulinum neurotoxin A (BoNT) toxicity. Here, we review what is currently known about the BoNT/A-SV2 interaction based on structural studies. Currently, five crystal structures of the receptor-binding domain (Hc) of BoNT subtypes A1 and A2 complexed to the large luminal domain (LD4) of SV2C have been determined. On the basis of the available structures, we will discuss the importance of protein-protein and protein-carbohydrate interactions for BoNT/A toxicity as well as the high plasticity of BoNT/A for receptor recognition by tolerating a variety of side-chain interactions at the interface. A plausible explanation how receptor-binding specificity of BoNT/A may be achieved without an extensive and conserved side chain-side chain interaction network will be provided.


Asunto(s)
Toxinas Botulínicas Tipo A/química , Glicoproteínas de Membrana/química , Proteínas del Tejido Nervioso/química , Gangliósidos , Humanos , Unión Proteica , Elementos Estructurales de las Proteínas , Células Receptoras Sensoriales
6.
Cell Microbiol ; 19(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27404998

RESUMEN

Botulinum and tetanus neurotoxins are the most toxic substances known and form the growing family of clostridial neurotoxins. They are composed of a metalloprotease light chain (L), linked via a disulfide bond to a heavy chain (H). H mediates the binding to nerve terminals and the membrane translocation of L into the cytosol where their substrates, the three SNARE proteins, are localised. L translocation is accompanied by unfolding, and it has to be reduced and reacquire the native fold to exert its neurotoxicity. The Thioredoxin reductase-Thioredoxin system is responsible for the reduction, but it is unknown whether the refolding of L is spontaneous or aided by host chaperones. Here we report that geldanamycin, a specific inhibitor of heat shock protein 90, hampers the refolding of L after membrane translocation and completely prevents the cleavage of SNAREs. We also found that geldanamycin strongly synergises with PX-12, an inhibitor of thioredoxin, suggesting that the processes of L chain refolding and interchain disulfide reduction are strictly coupled. Indeed we found that the heat shock protein 90 and the Thioredoxin reductase-Thioredoxin system physically interact on synaptic vesicle where they orchestrate a chaperone-redox machinery which is exploited by clostridial neurotoxins to deliver their catalytic part into the cytosol.


Asunto(s)
Citosol/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Pliegue de Proteína , Toxina Tetánica/metabolismo , Transporte de Proteínas , Proteolisis , Proteínas SNARE/metabolismo , Vesículas Sinápticas/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/metabolismo
7.
Sci Rep ; 6: 30257, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27443638

RESUMEN

The genome of Weissella oryzae SG25T was recently sequenced and a botulinum neurotoxin (BoNT) like gene was identified by bioinformatics methods. The typical three-domains organization of BoNTs with a N-terminal metalloprotease domain, a translocation and a cell binding domains could be identified. The BoNT family of neurotoxins is rapidly growing, but this was the first indication of the possible expression of a BoNT toxin outside the Clostridium genus. We performed molecular modeling and dynamics simulations showing that the 50 kDa N-terminal domain folds very similarly to the metalloprotease domain of BoNT/B, whilst the binding part is different. However, neither the recombinant metalloprotease nor the binding domains showed cross-reactivity with the standard antisera that define the seven serotypes of BoNTs. We found that the purified Weissella metalloprotease cleaves VAMP at a single site untouched by the other VAMP-specific BoNTs. This site is a unique Trp-Trp peptide bond located within the juxtamembrane segment of VAMP which is essential for neurotransmitter release. Therefore, the present study identifies the first non-Clostridial BoNT-like metalloprotease that cleaves VAMP at a novel and relevant site and we propose to label it BoNT/Wo.


Asunto(s)
Toxinas Botulínicas/química , Metaloproteasas/química , Neurotoxinas/química , Weissella/genética , Secuencia de Aminoácidos/genética , Toxinas Botulínicas/genética , Membrana Celular/química , Membrana Celular/genética , Clostridium botulinum/genética , Genoma Bacteriano , Metaloproteasas/genética , Modelos Moleculares , Simulación de Dinámica Molecular , Neurotoxinas/genética , Unión Proteica , Dominios Proteicos , Pliegue de Proteína , Weissella/química
8.
Biochim Biophys Acta ; 1858(3): 467-74, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26307528

RESUMEN

Tetanus and botulinum neurotoxins are produced by anaerobic bacteria of the genus Clostridium and are the most poisonous toxins known, with 50% mouse lethal dose comprised within the range of 0.1-few nanograms per Kg, depending on the individual toxin. Botulinum neurotoxins are similarly toxic to humans and can therefore be considered for potential use in bioterrorism. At the same time, their neurospecificity and reversibility of action make them excellent therapeutics for a growing and heterogeneous number of human diseases that are characterized by a hyperactivity of peripheral nerve terminals. The complete crystallographic structure is available for some botulinum toxins, and reveals that they consist of four domains functionally related to the four steps of their mechanism of neuron intoxication: 1) binding to specific receptors of the presynaptic membrane; 2) internalization via endocytic vesicles; 3) translocation across the membrane of endocytic vesicles into the neuronal cytosol; 4) catalytic activity of the enzymatic moiety directed towards the SNARE proteins. Despite the many advances in understanding the structure-mechanism relationship of tetanus and botulinum neurotoxins, the molecular events involved in the translocation step have been only partially elucidated. Here we will review recent advances that have provided relevant insights on the process and discuss possible models that can be experimentally tested. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Mauro Dalla Serra and Franco Gambale.


Asunto(s)
Toxinas Botulínicas/metabolismo , Membrana Celular/metabolismo , Endocitosis , Terminales Presinápticos/metabolismo , Proteínas SNARE/metabolismo , Toxina Tetánica/metabolismo , Animales , Toxinas Botulínicas/química , Membrana Celular/química , Humanos , Concentración de Iones de Hidrógeno , Ratones , Terminales Presinápticos/química , Transporte de Proteínas , Proteínas SNARE/química , Relación Estructura-Actividad , Toxina Tetánica/química
9.
Sci Rep ; 5: 17513, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26670952

RESUMEN

Botulinum neurotoxins (BoNTs) form a large class of potent and deadly neurotoxins. Given their growing number, it is of paramount importance to discover novel inhibitors targeting common steps of their intoxication process. Recently, EGA was shown to inhibit the action of bacterial toxins and viruses exhibiting a pH-dependent translocation step in mammalian cells, by interfering with their entry route. As BoNTs act in the cytosol of nerve terminals, the entry into an appropriate compartment wherefrom they translocate the catalytic moiety is essential for toxicity. Herein we propose an optimized procedure to synthesize EGA and we show that, in vitro, it prevents the neurotoxicity of different BoNT serotypes by interfering with their trafficking. Furthermore, in mice, EGA mitigates botulism symptoms induced by BoNT/A and significantly decreases the lethality of BoNT/B and BoNT/D. This opens the possibility of using EGA as a lead compound to develop novel inhibitors of botulinum neurotoxins.


Asunto(s)
Toxinas Botulínicas/antagonistas & inhibidores , Neurotoxinas/antagonistas & inhibidores , Parálisis/fisiopatología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Animales , Transporte Biológico , Toxinas Botulínicas/metabolismo , Diafragma/efectos de los fármacos , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neurotoxinas/metabolismo , Parálisis/tratamiento farmacológico , Parálisis/etiología , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/etiología , Proteínas SNARE/metabolismo , Semicarbazonas/síntesis química , Semicarbazonas/química , Semicarbazonas/farmacología
10.
FEBS J ; 281(9): 2115-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24628974

RESUMEN

UNLABELLED: Diphtheria toxin (DT), the etiological agent of the homonymous disease, like other bacterial toxins, has to undergo a dramatic structural change in order to be internalized into the cytosol, where it finally performs its function. The molecular mechanism of toxin transit across the membrane is not well known, but the available experimental evidence indicates that one of the three domains of the toxin, called the central α-helical domain, inserts into the lipid bilayer, so favoring the translocation of the catalytic domain. This process is driven by the acidic pH of the endosomal lumen. Here, we describe the crystal structure of DT grown at acidic pH in the presence of bicelles. We were unable to freeze the moment of DT insertion into the lipid bilayer, but our crystal structure indicates that the low pH causes the unfolding of the TH2, TH3 and TH4 α-helices. This event gives rise to the exposure of a hydrophobic surface that includes the TH5 and TH8 α-helices, and the loop region connecting the TH8 and TH9 α-helices. Their exposure is probably favored by the presence of lipid bilayers in the crystallization solution, and they appear to be ready to insert into the membrane. DATABASE: Coordinates and structure factors have been deposited in the Protein Data Bank under accession number 4OW6.


Asunto(s)
Ácidos/química , Toxina Diftérica/química , Concentración de Iones de Hidrógeno , Cristalografía por Rayos X , Endocitosis , Membrana Dobles de Lípidos , Modelos Moleculares , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...