Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nature ; 550(7674): 74-79, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28953867

RESUMO

De novo protein design holds promise for creating small stable proteins with shapes customized to bind therapeutic targets. We describe a massively parallel approach for designing, manufacturing and screening mini-protein binders, integrating large-scale computational design, oligonucleotide synthesis, yeast display screening and next-generation sequencing. We designed and tested 22,660 mini-proteins of 37-43 residues that target influenza haemagglutinin and botulinum neurotoxin B, along with 6,286 control sequences to probe contributions to folding and binding, and identified 2,618 high-affinity binders. Comparison of the binding and non-binding design sets, which are two orders of magnitude larger than any previously investigated, enabled the evaluation and improvement of the computational model. Biophysical characterization of a subset of the binder designs showed that they are extremely stable and, unlike antibodies, do not lose activity after exposure to high temperatures. The designs elicit little or no immune response and provide potent prophylactic and therapeutic protection against influenza, even after extensive repeated dosing.


Assuntos
Desenho de Fármacos , Influenza Humana/tratamento farmacológico , Influenza Humana/prevenção & controle , Terapia de Alvo Molecular/métodos , Engenharia de Proteínas/métodos , Proteínas/química , Proteínas/uso terapêutico , Toxinas Botulínicas/classificação , Toxinas Botulínicas/metabolismo , Simulação por Computador , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Temperatura Alta , Humanos , Influenza Humana/metabolismo , Simulação de Dinâmica Molecular , Ligação Proteica , Estabilidade Proteica , Proteínas/imunologia , Proteínas/metabolismo , Temperatura
2.
Genes Dev ; 26(3): 247-58, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22302937

RESUMO

Synapses are the fundamental units of neural circuits that enable complex behaviors. The neuromuscular junction (NMJ), a synapse formed between a motoneuron and a muscle fiber, has contributed greatly to understanding of the general principles of synaptogenesis as well as of neuromuscular disorders. NMJ formation requires neural agrin, a motoneuron-derived protein, which interacts with LRP4 (low-density lipoprotein receptor-related protein 4) to activate the receptor tyrosine kinase MuSK (muscle-specific kinase). However, little is known of how signals are transduced from agrin to MuSK. Here, we present the first crystal structure of an agrin-LRP4 complex, consisting of two agrin-LRP4 heterodimers. Formation of the initial binary complex requires the z8 loop that is specifically present in neuronal, but not muscle, agrin and that promotes the synergistic formation of the tetramer through two additional interfaces. We show that the tetrameric complex is essential for neuronal agrin-induced acetylcholine receptor (AChR) clustering. Collectively, these results provide new insight into the agrin-LRP4-MuSK signaling cascade and NMJ formation and represent a novel mechanism for activation of receptor tyrosine kinases.


Assuntos
Agrina/química , Agrina/metabolismo , Modelos Moleculares , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de LDL/química , Receptores de LDL/metabolismo , Transdução de Sinais , Animais , Sítios de Ligação , Linhagem Celular , Ativação Enzimática , Camundongos , Neurônios/metabolismo , Estrutura Quaternária de Proteína , Ratos
3.
PLoS Pathog ; 9(10): e1003690, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24130488

RESUMO

Botulinum neurotoxins (BoNTs) are produced by Clostridium botulinum and cause the fatal disease botulism, a flaccid paralysis of the muscle. BoNTs are released together with several auxiliary proteins as progenitor toxin complexes (PTCs) to become highly potent oral poisons. Here, we report the structure of a ∼760 kDa 14-subunit large PTC of serotype A (L-PTC/A) and reveal insight into its absorption mechanism. Using a combination of X-ray crystallography, electron microscopy, and functional studies, we found that L-PTC/A consists of two structurally and functionally independent sub-complexes. A hetero-dimeric 290 kDa complex protects BoNT, while a hetero-dodecameric 470 kDa complex facilitates its absorption in the harsh environment of the gastrointestinal tract. BoNT absorption is mediated by nine glycan-binding sites on the dodecameric sub-complex that forms multivalent interactions with carbohydrate receptors on intestinal epithelial cells. We identified monosaccharides that blocked oral BoNT intoxication in mice, which suggests a new strategy for the development of preventive countermeasures for BoNTs based on carbohydrate receptor mimicry.


Assuntos
Toxinas Botulínicas , Botulismo , Complexos Multiproteicos , Animais , Toxinas Botulínicas/química , Toxinas Botulínicas/genética , Toxinas Botulínicas/toxicidade , Clostridium botulinum/genética , Clostridium botulinum/metabolismo , Feminino , Camundongos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/toxicidade , Estrutura Quaternária de Proteína , Relação Estrutura-Atividade
4.
Nat Commun ; 14(1): 2338, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-37095076

RESUMO

Botulinum neurotoxin E (BoNT/E) is one of the major causes of human botulism and paradoxically also a promising therapeutic agent. Here we determined the co-crystal structures of the receptor-binding domain of BoNT/E (HCE) in complex with its neuronal receptor synaptic vesicle glycoprotein 2A (SV2A) and a nanobody that serves as a ganglioside surrogate. These structures reveal that the protein-protein interactions between HCE and SV2 provide the crucial location and specificity information for HCE to recognize SV2A and SV2B, but not the closely related SV2C. At the same time, HCE exploits a separated sialic acid-binding pocket to mediate recognition of an N-glycan of SV2. Structure-based mutagenesis and functional studies demonstrate that both the protein-protein and protein-glycan associations are essential for SV2A-mediated cell entry of BoNT/E and for its potent neurotoxicity. Our studies establish the structural basis to understand the receptor-specificity of BoNT/E and to engineer BoNT/E variants for new clinical applications.


Assuntos
Toxinas Botulínicas Tipo A , Vesículas Sinápticas , Humanos , Vesículas Sinápticas/metabolismo , Toxinas Botulínicas Tipo A/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica
5.
Biochem J ; 438(2): 255-63, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21639859

RESUMO

The AMPA (α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid) subfamily of iGluRs (ionotropic glutamate receptors) is essential for fast excitatory neurotransmission in the central nervous system. The malfunction of AMPARs (AMPA receptors) has been implicated in many neurological diseases, including Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. The active channels of AMPARs and other iGluR subfamilies are tetramers formed exclusively by assembly of subunits within the same subfamily. It has been proposed that the assembly process is controlled mainly by the extracellular ATD (N-terminal domain) of iGluR. In addition, ATD has also been implicated in synaptogenesis, iGluR trafficking and trans-synaptic signalling, through unknown mechanisms. We report in the present study a 2.5 Å (1 Å=0.1 nm) resolution crystal structure of the ATD of GluA1. Comparative analyses of the structure of GluA1-ATD and other subunits sheds light on our understanding of how ATD drives subfamily-specific assembly of AMPARs. In addition, analysis of the crystal lattice of GluA1-ATD suggests a novel mechanism by which the ATD might participate in inter-tetramer AMPAR clustering, as well as in trans-synaptic protein-protein interactions.


Assuntos
Receptores de AMPA/química , Animais , Cristalografia por Raios X , Modelos Moleculares , Maleabilidade , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Receptores de AMPA/isolamento & purificação
6.
Toxicon ; 147: 19-26, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29042313

RESUMO

Botulinum neurotoxins (BoNTs) are causative agents of the life-threatening disease botulism. They are naturally produced by species of the bacteria Clostridium botulinum as stable and non-covalent complexes, in which the BoNT molecule is assembled with several auxiliary non-toxic proteins. Some BoNT serotypes, represented by the well-studied BoNT serotype A (BoNT/A), are produced by Clostridium strains that carry the ha gene cluster, which encodes four neurotoxin-associated proteins (NTNHA, HA17, HA33, and HA70) that play an important role to deliver and protect BoNTs in the gastrointestinal tract during oral intoxication. In contrast, BoNT/E- and BoNT/F-producing strains carry a distinct gene cluster that encodes five proteins (NTNHA, P47, OrfX1, OrfX2, and OrfX3, termed the orfX cluster). The structures and functions of these proteins remain largely unknown. Here, we report the crystal structure of P47 resolved at 2.8 Å resolution. Surprisingly, P47 displays a structural topology that is similar to bactericidal/permeability-increasing (BPI) like proteins, which were previously identified only in eukaryotes. The similarity of a hydrophobic cleft of P47 with the phospholipid-binding groove of BPI suggests that P47 might be involved in lipid association to exert its function. Consistently, P47 associates and induces aggregation of asolectin-containing liposomes in a protein- and lipid-concentration dependent manner. These findings laid the foundation for future structural and functional studies of the potential roles of P47 and OrfX proteins in facilitating oral intoxication of BoNTs.


Assuntos
Toxinas Botulínicas/química , Clostridium botulinum/metabolismo , Sequência de Aminoácidos , Toxinas Botulínicas/classificação , Toxinas Botulínicas/genética , Toxinas Botulínicas/metabolismo , Clonagem Molecular , Clostridium botulinum/química , Regulação Bacteriana da Expressão Gênica/fisiologia , Lipossomos/química , Modelos Moleculares , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Conformação Proteica , Dobramento de Proteína
7.
Toxins (Basel) ; 9(3)2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28282873

RESUMO

Botulinum neurotoxins (BoNTs), which have been exploited as cosmetics and muscle-disorder treatment medicines for decades, are well known for their extreme neurotoxicity to humans. They pose a potential bioterrorism threat because they cause botulism, a flaccid muscular paralysis-associated disease that requires immediate antitoxin treatment and intensive care over a long period of time. In addition to the existing seven established BoNT serotypes (BoNT/A-G), a new mosaic toxin type termed BoNT/HA (aka type FA or H) was reported recently. Sequence analyses indicate that the receptor-binding domain (HC) of BoNT/HA is ~84% identical to that of BoNT/A1. However, BoNT/HA responds differently to some potent BoNT/A-neutralizing antibodies (e.g., CR2) that target the HC. Therefore, it raises a serious concern as to whether BoNT/HA poses a new threat to our biosecurity. In this study, we report the first high-resolution crystal structure of BoNT/HA-HC at 1.8 Å. Sequence and structure analyses reveal that BoNT/HA and BoNT/A1 are different regarding their binding to cell-surface receptors including both polysialoganglioside (PSG) and synaptic vesicle glycoprotein 2 (SV2). Furthermore, the new structure also provides explanations for the ~540-fold decreased affinity of antibody CR2 towards BoNT/HA compared to BoNT/A1. Taken together, these new findings advance our understanding of the structure and function of this newly identified toxin at the molecular level, and pave the way for the future development of more effective countermeasures.


Assuntos
Toxinas Botulínicas/química , Neurotoxinas/química , Toxinas Botulínicas/genética , Toxinas Botulínicas/metabolismo , Neurotoxinas/genética , Neurotoxinas/metabolismo , Ligação Proteica , Domínios Proteicos
8.
Sci Rep ; 7(1): 7438, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28785006

RESUMO

Antibody treatment is currently the only available countermeasure for botulism, a fatal illness caused by flaccid paralysis of muscles due to botulinum neurotoxin (BoNT) intoxication. Among the seven major serotypes of BoNT/A-G, BoNT/A poses the most serious threat to humans because of its high potency and long duration of action. Prior to entering neurons and blocking neurotransmitter release, BoNT/A recognizes motoneurons via a dual-receptor binding process in which it engages both the neuron surface polysialoganglioside (PSG) and synaptic vesicle glycoprotein 2 (SV2). Previously, we identified a potent neutralizing antitoxin against BoNT/A1 termed ciA-C2, derived from a camelid heavy-chain-only antibody (VHH). In this study, we demonstrate that ciA-C2 prevents BoNT/A1 intoxication by inhibiting its binding to neuronal receptor SV2. Furthermore, we determined the crystal structure of ciA-C2 in complex with the receptor-binding domain of BoNT/A1 (HCA1) at 1.68 Å resolution. The structure revealed that ciA-C2 partially occupies the SV2-binding site on HCA1, causing direct interference of HCA1 interaction with both the N-glycan and peptide-moiety of SV2. Interestingly, this neutralization mechanism is similar to that of a monoclonal antibody in clinical trials, despite that ciA-C2 is more than 10-times smaller. Taken together, these results enlighten our understanding of BoNT/A1 interactions with its neuronal receptor, and further demonstrate that inhibiting toxin binding to the host receptor is an efficient countermeasure strategy.


Assuntos
Anticorpos Neutralizantes/farmacologia , Toxinas Botulínicas Tipo A/química , Toxinas Botulínicas Tipo A/metabolismo , Camelidae/imunologia , Proteínas do Tecido Nervoso/metabolismo , Animais , Anticorpos Neutralizantes/química , Sítios de Ligação , Cristalografia por Raios X , Gangliosídeos/metabolismo , Cadeias Pesadas de Imunoglobulinas/química , Cadeias Pesadas de Imunoglobulinas/farmacologia , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Ratos , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/farmacologia
9.
Nat Struct Mol Biol ; 23(7): 656-62, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27294781

RESUMO

Botulinum neurotoxin serotype A1 (BoNT/A1), a licensed drug widely used for medical and cosmetic applications, exerts its action by invading motoneurons. Here we report a 2.0-Å-resolution crystal structure of the BoNT/A1 receptor-binding domain in complex with its neuronal receptor, glycosylated human SV2C. We found that the neuronal tropism of BoNT/A1 requires recognition of both the peptide moiety and an N-linked glycan on SV2. This N-glycan-which is conserved in all SV2 isoforms across vertebrates-is essential for BoNT/A1 binding to neurons and for its potent neurotoxicity. The glycan-binding interface on SV2 is targeted by a human BoNT/A1-neutralizing antibody currently licensed as an antibotulism drug. Our studies reveal a new paradigm of host-pathogen interactions, in which pathogens exploit conserved host post-translational modifications, thereby achieving highly specific receptor binding while also tolerating genetic changes across multiple isoforms of receptors.


Assuntos
Anticorpos Monoclonais/química , Antídotos/química , Toxinas Botulínicas Tipo A/química , Clostridium botulinum/química , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Sítios de Ligação , Transporte Biológico , Toxinas Botulínicas Tipo A/metabolismo , Clonagem Molecular , Clostridium botulinum/patogenicidade , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Glicosilação , Células HEK293 , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Modelos Moleculares , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
10.
Prog Biophys Mol Biol ; 117(2-3): 225-231, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25701633

RESUMO

Botulinum neurotoxins (BoNTs) are among the most deadly toxins known. They act rapidly in a highly specific manner to block neurotransmitter release by cleaving the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) complex at neuromuscular junctions. The extreme toxicity of BoNTs relies predominantly on their neurotropism that is accomplished by recognition of two host receptors, a polysialo-ganglioside and in the majority of cases a synaptic vesicle protein, through their receptor-binding domains. Two proteins, synaptotagmin and synaptic vesicle glycoprotein 2, have been identified as the receptors for various serotypes of BoNTs. Here, we review recent breakthroughs in the structural studies of BoNT-protein receptor recognitions that highlight a range of diverse mechanisms by which BoNTs manipulate host neuronal proteins for highly specific uptake at neuromuscular junctions.


Assuntos
Toxinas Botulínicas/química , Toxinas Botulínicas/ultraestrutura , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/ultraestrutura , Neurotoxinas/química , Sinapses/química , Sítios de Ligação , Simulação por Computador , Modelos Químicos , Modelos Moleculares , Modelos Neurológicos , Ligação Proteica , Sinapses/ultraestrutura
11.
Toxins (Basel) ; 6(2): 624-35, 2014 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-24525478

RESUMO

Botulinum neurotoxins (BoNTs) are potent bacterial toxins. The high oral toxicity of BoNTs is largely attributed to the progenitor toxin complex (PTC), which is assembled from BoNT and nontoxic neurotoxin-associated proteins (NAPs) that are produced together with BoNT in bacteria. Here, we performed ex vivo studies to examine binding of the highly homogeneous recombinant NAPs to mouse small intestine. We also carried out the first comprehensive glycan array screening with the hemagglutinin (HA) component of NAPs. Our data confirmed that intestinal binding of the PTC is partly mediated by the HA moiety through multivalent interactions between HA and host carbohydrates. The specific HA-carbohydrate recognition could be inhibited by receptor-mimicking saccharides.


Assuntos
Toxinas Botulínicas Tipo A/metabolismo , Hemaglutininas/metabolismo , Polissacarídeos/metabolismo , Animais , Intestino Delgado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica
12.
Neurosci Lett ; 548: 90-4, 2013 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-23685131

RESUMO

Alzheimer's disease (AD) is primarily caused by overproduction/deposition of ß-amyloid (Aß) in the brain. Dysregulation of iron in the brain also contributes to AD. Although iron affects ß-amyloid precursor protein (APP) expression and Aß deposition, detailed role of iron in AD requires further elucidation. Aß is produced by sequential proteolytic cleavages of APP by ß-secretase and γ-secretase. The γ-secretase complex comprises presenilins (PS1 or PS2), nicastrin, APH-1, and PEN-2. Herein, we find that PEN-2 can interact with ferritin light chain (FTL), an important component of the iron storage protein ferritin. In addition, we show that overexpression of FTL increases the protein levels of PEN-2 and PS1 amino-terminal fragment (NTF) and promotes γ-secretase activity for more production of Aß and notch intracellular domain (NICD). Furthermore, iron treatments increase the levels of FTL, PEN-2 and PS1 NTF and promote γ-secretase-mediated NICD production. Moreover, downregulation of FTL decreases the levels of PEN-2 and PS1 NTF. Together, our results suggest that iron can increase γ-secretase activity through promoting the level of FTL that interacts with and stabilizes PEN-2, providing a new molecular link between iron, PEN-2/γ-secretase and Aß generation in AD.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Apoferritinas/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais/fisiologia , Ativação Enzimática , Células HEK293 , Humanos , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA