Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 226
Filtrar
1.
Blood ; 137(21): 2970-2980, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33569592

RESUMO

Interaction of factor VIII (FVIII) with von Willebrand factor (VWF) is mediated by the VWF D'D3 domains and thrombin-mediated release is essential for hemostasis after vascular injury. VWF-D'D3 mutations resulting in loss of FVIII binding are the underlying cause of von Willebrand disease (VWD) type 2N. Furthermore, the FVIII-VWF interaction has significant implications for the development of therapeutics for bleeding disorders, particularly hemophilia A, in which endogenous VWF clearance imposes a half-life ceiling on replacement FVIII therapy. To understand the structural basis of FVIII engagement by VWF, we solved the structure of BIVV001 by cryo-electron microscopy to 2.9 Å resolution. BIVV001 is a bioengineered clinical-stage FVIII molecule for the treatment of hemophilia A. In BIVV001, VWF-D'D3 is covalently linked to an Fc domain of a B domain-deleted recombinant FVIII (rFVIII) Fc fusion protein, resulting in a stabilized rFVIII/VWF-D'D3 complex. Our rFVIII/VWF structure resolves BIVV001 architecture and provides a detailed spatial understanding of previous biochemical and clinical observations related to FVIII-VWF engagement. Notably, the FVIII acidic a3 peptide region (FVIII-a3), established as a critical determinant of FVIII/VWF complex formation, inserts into a basic groove formed at the VWF-D'/rFVIII interface. Our structure shows direct interaction of sulfated Y1680 in FVIII-a3 and VWF-R816 that, when mutated, leads to severe hemophilia A or VWD type 2N, respectively. These results provide insight on this key coagulation complex, explain the structural basis of many hemophilia A and VWD type 2N mutations, and inform studies to further elucidate how VWF dissociates rapidly from FVIII upon activation.


Assuntos
Microscopia Crioeletrônica/métodos , Fator VIII/química , Proteínas Recombinantes de Fusão/química , Fator de von Willebrand/química , Combinação de Medicamentos , Humanos , Modelos Moleculares , Conformação Proteica , Domínios Proteicos , Mapeamento de Interação de Proteínas , Proteínas Recombinantes de Fusão/ultraestrutura
2.
Int J Mol Sci ; 22(1)2020 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-33374407

RESUMO

Thymosin α1 (Tα1) is an immunostimulatory peptide for the treatment of hepatitis B virus (HBV) and hepatitis C virus (HCV) infections and used as an immune enhancer, which also offers prospects in the context of COVID-19 infections and cancer. Manufacturing of this N-terminally acetylated 28-residue peptide is demanding, and its short plasma half-life limits in vivo efficacy and requires frequent dosing. Here, we combined the PASylation technology with enzymatic in situ N-acetylation by RimJ to produce a long-acting version of Tα1 in Escherichia coli at high yield. ESI-MS analysis of the purified fusion protein indicated the expected composition without any signs of proteolysis. SEC analysis revealed a 10-fold expanded hydrodynamic volume resulting from the fusion with a conformationally disordered Pro/Ala/Ser (PAS) polypeptide of 600 residues. This size effect led to a plasma half-life in rats extended by more than a factor 8 compared to the original synthetic peptide due to retarded kidney filtration. Our study provides the basis for therapeutic development of a next generation thymosin α1 with prolonged circulation. Generally, the strategy of producing an N-terminally protected PASylated peptide solves three major problems of peptide drugs: (i) instability in the expression host, (ii) rapid degradation by serum exopeptidases, and (iii) low bioactivity because of fast renal clearance.


Assuntos
Adjuvantes Imunológicos/farmacocinética , Timalfasina/farmacocinética , Acetilação , Acetiltransferases/metabolismo , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/farmacologia , Animais , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Feminino , Meia-Vida , Espectrometria de Massas , Microscopia Eletrônica de Varredura , Neoplasias/tratamento farmacológico , Peptídeos/química , Proteólise , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/sangue , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas Ribossômicas/metabolismo , Timalfasina/sangue , Timalfasina/química , Timalfasina/genética , Viroses/tratamento farmacológico , Tratamento Farmacológico da COVID-19
3.
PLoS Biol ; 18(11): e3000925, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33216759

RESUMO

Lifeact is a short actin-binding peptide that is used to visualize filamentous actin (F-actin) structures in live eukaryotic cells using fluorescence microscopy. However, this popular probe has been shown to alter cellular morphology by affecting the structure of the cytoskeleton. The molecular basis for such artefacts is poorly understood. Here, we determined the high-resolution structure of the Lifeact-F-actin complex using electron cryo-microscopy (cryo-EM). The structure reveals that Lifeact interacts with a hydrophobic binding pocket on F-actin and stretches over 2 adjacent actin subunits, stabilizing the DNase I-binding loop (D-loop) of actin in the closed conformation. Interestingly, the hydrophobic binding site is also used by actin-binding proteins, such as cofilin and myosin and actin-binding toxins, such as the hypervariable region of TccC3 (TccC3HVR) from Photorhabdus luminescens and ExoY from Pseudomonas aeruginosa. In vitro binding assays and activity measurements demonstrate that Lifeact indeed competes with these proteins, providing an explanation for the altering effects of Lifeact on cell morphology in vivo. Finally, we demonstrate that the affinity of Lifeact to F-actin can be increased by introducing mutations into the peptide, laying the foundation for designing improved actin probes for live cell imaging.


Assuntos
Actinas/química , Proteínas dos Microfilamentos/química , Actinas/metabolismo , Actinas/ultraestrutura , Animais , Toxinas Bacterianas/química , Sítios de Ligação , Ligação Competitiva , Cofilina 1/química , Cofilina 1/ultraestrutura , Microscopia Crioeletrônica , Corantes Fluorescentes/química , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Técnicas In Vitro , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/ultraestrutura , Microscopia Confocal , Modelos Moleculares , Miosinas/química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Engenharia de Proteínas , Domínios e Motivos de Interação entre Proteínas , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestrutura
4.
Drug Metab Dispos ; 48(12): 1380-1392, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33037045

RESUMO

The most commonly used oral antidiabetic drug, metformin, is a substrate of the hepatic uptake transporter OCT1 (gene name SLC22A1). However, OCT1 deficiency leads to more pronounced reductions of metformin concentrations in mouse than in human liver. Similarly, the effects of OCT1 deficiency on the pharmacokinetics of thiamine were reported to differ between human and mouse. Here, we compared the uptake characteristics of metformin and thiamine between human and mouse OCT1 using stably transfected human embryonic kidney 293 cells. The affinity for metformin was 4.9-fold lower in human than in mouse OCT1, resulting in a 6.5-fold lower intrinsic clearance. Therefore, the estimated liver-to-blood partition coefficient is only 3.34 in human compared with 14.4 in mouse and may contribute to higher intrahepatic concentrations in mice. Similarly, the affinity for thiamine was 9.5-fold lower in human than in mouse OCT1. Using human-mouse chimeric OCT1, we showed that simultaneous substitution of transmembrane helices TMH2 and TMH3 resulted in the reversal of affinity for metformin. Using homology modeling, we suggest several explanations, of which a different interaction of Leu155 (human TMH2) compared with Val156 (mouse TMH2) with residues in TMH3 had the strongest experimental support. In conclusion, the contribution of human OCT1 to the cellular uptake of thiamine and especially of metformin may be much lower than that of mouse OCT1. This may lead to an overestimation of the effects of OCT1 on hepatic concentrations in humans when using mouse as a model. In addition, comparative analyses of human and mouse orthologs may help reveal mechanisms of OCT1 transport. SIGNIFICANCE STATEMENT: OCT1 is a major hepatic uptake transporter of metformin and thiamine, but this study reports strong differences in the affinity for both compounds between human and mouse OCT1. Consequently, intrahepatic metformin concentrations could be much higher in mice than in humans, impacting metformin actions and representing a strong limitation of using rodent animal models for predictions of OCT1-related pharmacokinetics and efficacy in humans. Furthermore, OCT1 transmembrane helices TMH2 and TMH3 were identified to confer the observed species-specific differences in metformin affinity.


Assuntos
Metformina/farmacocinética , Transportador 1 de Cátions Orgânicos/metabolismo , Tiamina/farmacocinética , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Células HEK293 , Hepatócitos , Humanos , Fígado/enzimologia , Masculino , Camundongos , Transportador 1 de Cátions Orgânicos/genética , Transportador 1 de Cátions Orgânicos/ultraestrutura , Conformação Proteica em alfa-Hélice/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Relação Estrutura-Atividade
5.
Biochem Biophys Res Commun ; 532(1): 127-133, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-32828540

RESUMO

Evidence of a complex formation is a crucial step in the structural studies of ligand-receptor interactions. Here we presented a simple and fast approach for qualitative screening of the complex formation between the chimeric extracellular domain of the nicotinic acetylcholine receptor (α7-ECD) and three-finger proteins. Complex formation of snake toxins α-Bgtx and WTX, as well as of recombinant analogs of human proteins Lynx1 and SLURP-1, with α7-ECD was confirmed using fluorescently labeled ligands and size-exclusion chromatography with simultaneous absorbance and fluorescence detection. WTX/α7-ECD complex formation also was confirmed by cryo-EM. The proposed approach could easily be adopted to study the interaction of other receptors with their ligands.


Assuntos
Receptor Nicotínico de Acetilcolina alfa7/química , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Bungarotoxinas/química , Bungarotoxinas/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Transporte/ultraestrutura , Cromatografia em Gel , Microscopia Crioeletrônica , Venenos Elapídicos/química , Venenos Elapídicos/metabolismo , Corantes Fluorescentes , Humanos , Ligantes , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Ressonância de Plasmônio de Superfície , Receptor Nicotínico de Acetilcolina alfa7/ultraestrutura
6.
Methods Mol Biol ; 2159: 55-65, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32529363

RESUMO

MxB/Mx2 is an interferon-induced dynamin-like GTPase, which restricts a number of life-threatening viruses. Because of its N-terminal region, predicted to be intrinsically disordered, and its propensity to self-oligomerize, purification of the full-length protein has not been successful in conventional E. coli expression systems. In this chapter, we describe an expression and purification procedure to obtain pure full-length wild-type MxB from suspension-adapted mammalian cells. We further describe how to characterize its GTPase activity and oligomerization function.


Assuntos
Expressão Gênica , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/isolamento & purificação , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Animais , Cromatografia de Afinidade/métodos , Cromatografia em Gel , Ativação Enzimática , GTP Fosfo-Hidrolases/química , GTP Fosfo-Hidrolases/isolamento & purificação , GTP Fosfo-Hidrolases/metabolismo , Células HEK293 , Humanos , Proteínas de Resistência a Myxovirus/metabolismo , Plasmídeos/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Transfecção
7.
Int J Mol Sci ; 20(6)2019 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-30909376

RESUMO

Myocardial tissue damage that occurs during an ischemic event leads to a spiraling deterioration of cardiac muscle structural and functional integrity. Reperfusion is the only known efficacious strategy and is the most commonly used treatment to reduce injury and prevent remodeling. However, timing is critical, and the procedure is not always feasible for a variety of reasons. The complex molecular basis for cardioprotection has been studied for decades but formulation of a viable therapeutic that can significantly attenuate myocardial injury remains elusive. In this review, we address barriers to the development of a fruitful approach that will substantially improve the prognosis of those suffering from this widespread and largely unmitigated disease. Furthermore, we proffer that ephrinA1, a candidate molecule that satisfies many of the important criteria discussed, possesses robust potential to overcome these hurdles and thus offers protection that surpasses the limitations currently observed.


Assuntos
Infarto do Miocárdio/terapia , Animais , Cardiotônicos , Terapia Combinada , Gerenciamento Clínico , Efrina-A1/genética , Efrina-A1/metabolismo , Efrina-A1/uso terapêutico , Efrina-A1/ultraestrutura , Humanos , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Fragmentos Fc das Imunoglobulinas/ultraestrutura , Ligantes , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Proteínas Recombinantes de Fusão/ultraestrutura , Pesquisa Translacional Biomédica , Resultado do Tratamento
8.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1410-1420, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30790619

RESUMO

Synucleinopathies comprise a diverse group of neurodegenerative diseases including Parkinson's disease (PD), dementia with Lewy bodies, and multiple system atrophy. These share a common pathological feature, the deposition of alpha-synuclein (a-syn) in neurons or oligodendroglia. A-syn is highly conserved in vertebrates, but the primary sequence of mouse a-syn differs from that of human at seven positions. However, structural differences of their aggregates remain to be fully characterized. In this study, we found that human and mouse a-syn aggregated in vitro formed morphologically distinct amyloid fibrils exhibiting twisted and straight structures, respectively. Furthermore, we identified different protease-resistant core regions, long and short, in human and mouse a-syn aggregates. Interestingly, among the seven unconserved amino acids, only A53T substitution, one of the familial PD mutations, was responsible for structural conversion to the straight-type. Finally, we checked whether the structural differences are transmissible by seeding and found that human a-syn seeded with A53T aggregates formed straight-type fibrils with short protease-resistant cores. These results suggest that a-syn aggregates form sequence-dependent polymorphic fibrils upon spontaneous aggregation but become seed structure-dependent upon seeding.


Assuntos
Amiloide/ultraestrutura , Agregados Proteicos , alfa-Sinucleína/ultraestrutura , Sequência de Aminoácidos , Substituição de Aminoácidos , Amiloide/genética , Amiloide/metabolismo , Animais , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
9.
Nat Protoc ; 14(2): 616-638, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30675035

RESUMO

Protein dimerization and oligomerization are essential to most cellular functions, yet measurement of the size of these oligomers in live cells, especially when their size changes over time and space, remains a challenge. A commonly used approach for studying protein aggregates in cells is number and brightness (N&B), a fluorescence microscopy method that is capable of measuring the apparent average number of molecules and their oligomerization (brightness) in each pixel from a series of fluorescence microscopy images. We have recently expanded this approach in order to allow resampling of the raw data to resolve the statistical weighting of coexisting species within each pixel. This feature makes enhanced N&B (eN&B) optimal for capturing the temporal aspects of protein oligomerization when a distribution of oligomers shifts toward a larger central size over time. In this protocol, we demonstrate the application of eN&B by quantifying receptor clustering dynamics using electron-multiplying charge-coupled device (EMCCD)-based total internal reflection microscopy (TIRF) imaging. TIRF provides a superior signal-to-noise ratio, but we also provide guidelines for implementing eN&B in confocal microscopes. For each time point, eN&B requires the acquisition of 200 frames, and it takes a few seconds up to 2 min to complete a single time point. We provide an eN&B (and standard N&B) MATLAB software package amenable to any standard confocal or TIRF microscope. The software requires a high-RAM computer (64 Gb) to run and includes a photobleaching detrending algorithm, which allows extension of the live imaging for more than an hour.


Assuntos
Efrina-B1/ultraestrutura , Processamento de Imagem Assistida por Computador/estatística & dados numéricos , Microscopia de Fluorescência/métodos , Proteínas Recombinantes de Fusão/ultraestrutura , Software , Efrina-B1/genética , Efrina-B1/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Transferência Ressonante de Energia de Fluorescência , Expressão Gênica , Células HEK293 , Humanos , Microscopia Confocal/métodos , Agregados Proteicos , Multimerização Proteica , Receptor EphB2/genética , Receptor EphB2/metabolismo , Receptor EphB2/ultraestrutura , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Razão Sinal-Ruído
10.
Small ; 13(36)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28748658

RESUMO

Vesicles assembled from folded, globular proteins have potential for functions different from traditional lipid or polymeric vesicles. However, they also present challenges in understanding the assembly process and controlling vesicle properties. From detailed investigation of the assembly behavior of recombinant fusion proteins, this work reports a simple strategy to engineer protein vesicles containing functional, globular domains. This is achieved through tunable self-assembly of recombinant globular fusion proteins containing leucine zippers and elastin-like polypeptides. The fusion proteins form complexes in solution via high affinity binding of the zippers, and transition through dynamic coacervates to stable hollow vesicles upon warming. The thermal driving force, which can be tuned by protein concentration or temperature, controls both vesicle size and whether vesicles are single or bi-layered. These results provide critical information to engineer globular protein vesicles via self-assembly with desired size and membrane structure.


Assuntos
Elastina/química , Peptídeos/química , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/química , Elastina/ultraestrutura , Proteínas de Membrana/química , Nefelometria e Turbidimetria , Difração de Nêutrons , Transição de Fase , Proteínas Recombinantes de Fusão/ultraestrutura , Espalhamento a Baixo Ângulo , Tensoativos/química , Temperatura
11.
Microsc Microanal ; 23(1): 56-68, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28112080

RESUMO

The bulk of the major core protein VP7 in African horse sickness virus (AHSV) self-assembles into flat, hexagonal crystalline particles in a process appearing unrelated to viral replication. Why this unique characteristic of AHSV VP7 is genetically conserved, and whether VP7 aggregation and particle formation have an effect on cellular biology or the viral life cycle, is unknown. Here we investigated how different small peptide and enhanced green fluorescent protein (eGFP) insertions into the VP7 top domain affected VP7 localization, aggregation, and particle formation. This was done using a dual laser scanning confocal and transmission electron microscopy approach in conjunction with analyses of the solubility, aggregation, and fluorescence profiles of the proteins. VP7 top domain modifications did not prevent trimerization, or intracellular trafficking, to one or two discrete sites in the cell. However, modifications that resulted in a misfolded and insoluble VP7-eGFP component blocked trafficking, and precluded protein accumulation at a single cellular site, perhaps by interfering with normal trimer-trimer interactions. Furthermore, the modifications disrupted the stable layering of the trimers into characteristic AHSV VP7 crystalline particles. It was concluded that VP7 trafficking is driven by a balance between VP7 solubility, trimer forming ability, and trimer-trimer interactions.


Assuntos
Vírus da Doença Equina Africana/metabolismo , Microscopia Confocal/métodos , Microscopia Eletrônica de Transmissão/métodos , Proteínas do Core Viral/fisiologia , Proteínas do Core Viral/ultraestrutura , Vírus da Doença Equina Africana/genética , Animais , Baculoviridae/genética , Regulação Viral da Expressão Gênica , Vetores Genéticos , Proteínas de Fluorescência Verde , Estágios do Ciclo de Vida , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Células Sf9 , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Proteínas Virais de Fusão/fisiologia , Proteínas Virais de Fusão/ultraestrutura , Replicação Viral
12.
mBio ; 7(2): e00257, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-27006464

RESUMO

UNLABELLED: Influenza viruses expressing chimeric hemagglutinins (HAs) are important tools in the quest for a universal vaccine. Using cryo-electron tomography, we have determined the structures of a chimeric HA variant that comprises an H1 stalk and an H5 globular head domain (cH5/1 HA) in native and antibody-bound states. We show that cH5/1 HA is structurally different from native HA, displaying a 60° rotation between the stalk and head groups, leading to a novel and unexpected "open" arrangement of HA trimers. cH5/1N1 viruses also display higher glycoprotein density than pH1N1 or H5N1 viruses, but despite these differences, antibodies that target either the stalk or head domains of hemagglutinins still bind to cH5/1 HA with the same consequences as those observed with native H1 or H5 HA. Our results show that a large range of structural plasticity can be tolerated in the chimeric spike scaffold without disrupting structural and geometric aspects of antibody binding. IMPORTANCE: Chimeric hemagglutinin proteins are set to undergo human clinical trials as a universal influenza vaccine candidate, yet no structural information for these proteins is available. Using cryo-electron tomography, we report the first three-dimensional (3D) visualization of chimeric hemagglutinin proteins displayed on the surface of the influenza virus. We show that, unexpectedly, the chimeric hemagglutinin structure differs from those of naturally occurring hemagglutinins by displaying a more open head domain and a dramatically twisted head/stalk arrangement. Despite this unusual spatial relationship between head and stalk regions, virus preparations expressing the chimeric hemagglutinin are fully infectious and display a high glycoprotein density, which likely helps induction of a broadly protective immune response.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/ultraestrutura , Vírus da Influenza A/ultraestrutura , Proteínas Recombinantes de Fusão/ultraestrutura , Anticorpos Antivirais/metabolismo , Microscopia Crioeletrônica , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A/genética , Vacinas contra Influenza/genética , Modelos Moleculares , Ligação Proteica , Proteínas Recombinantes de Fusão/genética
13.
Nat Nanotechnol ; 9(10): 858-66, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25240674

RESUMO

Many natural underwater adhesives harness hierarchically assembled amyloid nanostructures to achieve strong and robust interfacial adhesion under dynamic and turbulent environments. Despite recent advances, our understanding of the molecular design, self-assembly and structure-function relationships of these natural amyloid fibres remains limited. Thus, designing biomimetic amyloid-based adhesives remains challenging. Here, we report strong and multi-functional underwater adhesives obtained from fusing mussel foot proteins (Mfps) of Mytilus galloprovincialis with CsgA proteins, the major subunit of Escherichia coli amyloid curli fibres. These hybrid molecular materials hierarchically self-assemble into higher-order structures, in which, according to molecular dynamics simulations, disordered adhesive Mfp domains are exposed on the exterior of amyloid cores formed by CsgA. Our fibres have an underwater adhesion energy approaching 20.9 mJ m(-2), which is 1.5 times greater than the maximum of bio-inspired and bio-derived protein-based underwater adhesives reported thus far. Moreover, they outperform Mfps or curli fibres taken on their own and exhibit better tolerance to auto-oxidation than Mfps at pH ≥ 7.0.


Assuntos
Adesivos/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Mytilus/química , Nanofibras/química , Proteínas/química , Adesividade , Animais , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/ultraestrutura , Modelos Moleculares , Mytilus/genética , Nanofibras/ultraestrutura , Proteínas/genética , Proteínas/ultraestrutura , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Água/química
14.
Biochemistry ; 53(41): 6452-62, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25232897

RESUMO

Self-assembling proteins represent potential scaffolds for the organization of enzymatic activities. The alkaline protease repeats-in-toxin (RTX) domain from Pseudomonas aeruginosa undergoes multiple structural transitions in the presence and absence of calcium, a native structural cofactor. In the absence of calcium, this domain is capable of spontaneous, ordered polymerization, producing amyloid-like fibrils and large two-dimensional protein sheets. This polymerization occurs under near-physiological conditions, is rapid, and can be controlled by regulating calcium in solution. Fusion of the RTX domain to a soluble protein results in the incorporation of engineered protein function into these macromolecular assemblies. Applications of this protein sequence in bacterial adherence and colonization and the generation of biomaterials are discussed.


Assuntos
Amiloide/química , Proteínas de Bactérias/química , Cálcio/química , Metaloexopeptidases/química , Modelos Moleculares , Pseudomonas aeruginosa/enzimologia , Fosfatase Alcalina/química , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Fosfatase Alcalina/ultraestrutura , Amiloide/genética , Amiloide/metabolismo , Amiloide/ultraestrutura , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Cálcio/metabolismo , Dicroísmo Circular , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Fluorescência Verde/ultraestrutura , Cinética , Metaloexopeptidases/genética , Metaloexopeptidases/metabolismo , Metaloexopeptidases/ultraestrutura , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Polimerização , Agregação Patológica de Proteínas , Engenharia de Proteínas , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Sequências Repetitivas de Aminoácidos , Serina Endopeptidases/química , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Serina Endopeptidases/ultraestrutura
15.
PLoS One ; 9(9): e106936, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25233114

RESUMO

Purification of suitable quantity of homogenous protein is very often the bottleneck in protein structural studies. Overexpression of a desired gene and attachment of enzymatically cleavable affinity tags to the protein of interest made a breakthrough in this field. Here we describe the structure of Galleria mellonella silk proteinase inhibitor 2 (GmSPI-2) determined both by X-ray diffraction and NMR spectroscopy methods. GmSPI-2 was purified using a new method consisting in non-enzymatic His-tag removal based on a highly specific peptide bond cleavage reaction assisted by Ni(II) ions. The X-ray crystal structure of GmSPI-2 was refined against diffraction data extending to 0.98 Å resolution measured at 100 K using synchrotron radiation. Anisotropic refinement with the removal of stereochemical restraints for the well-ordered parts of the structure converged with R factor of 10.57% and Rfree of 12.91%. The 3D structure of GmSPI-2 protein in solution was solved on the basis of 503 distance constraints, 10 hydrogen bonds and 26 torsion angle restraints. It exhibits good geometry and side-chain packing parameters. The models of the protein structure obtained by X-ray diffraction and NMR spectroscopy are very similar to each other and reveal the same ß2αß fold characteristic for Kazal-family serine proteinase inhibitors.


Assuntos
Proteínas de Insetos/ultraestrutura , Mariposas/enzimologia , Proteínas Recombinantes de Fusão/ultraestrutura , Marcadores de Afinidade/química , Sequência de Aminoácidos , Animais , Simulação por Computador , Cristalografia por Raios X/métodos , Endopeptidase K/antagonistas & inibidores , Proteínas de Insetos/análise , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular/métodos , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/análise , Alinhamento de Sequência , Subtilisina/antagonistas & inibidores
16.
Nat Commun ; 5: 4807, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25185702

RESUMO

Dynactin is an essential cofactor for most cellular functions of the microtubule motor cytoplasmic dynein, but the mechanism by which dynactin activates dynein remains unclear. Here we use single molecule approaches to investigate dynein regulation by the dynactin subunit p150(Glued). We investigate the formation and motility of a dynein-p150(Glued) co-complex using dual-colour total internal reflection fluorescence microscopy. p150(Glued) recruits and tethers dynein to the microtubule in a concentration-dependent manner. Single molecule imaging of motility in cell extracts demonstrates that the CAP-Gly domain of p150(Glued) decreases the detachment rate of the dynein-dynactin complex from the microtubule and also acts as a brake to slow the dynein motor. Consistent with this important role, two neurodegenerative disease-causing mutations in the CAP-Gly domain abrogate these functions in our assays. Together, these observations support a model in which dynactin enhances the initial recruitment of dynein onto microtubules and promotes the sustained engagement of dynein with its cytoskeletal track.


Assuntos
Dineínas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Sítios de Ligação , Células COS , Movimento Celular/fisiologia , Chlorocebus aethiops , Complexo Dinactina , Dineínas/antagonistas & inibidores , Dineínas/genética , Dineínas/ultraestrutura , Feminino , Humanos , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/ultraestrutura , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura
17.
Mol Microbiol ; 91(5): 1022-35, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24417346

RESUMO

Curli are functional amyloids expressed as fibres on the surface of Enterobacteriaceae. Contrary to the protein misfolding events associated with pathogenic amyloidosis, curli are the result of a dedicated biosynthetic pathway. A specialized transporter in the outer membrane, CsgG, operates in conjunction with the two accessory proteins CsgE and CsgF to secrete curlin subunits to the extracellular surface, where they nucleate into cross-beta strand fibres. Here we investigate the substrate tolerance of the CsgG transporter and the capability of heterologous sequences to be built into curli fibres. Non-native polypeptides ranging up to at least 260 residues were exported when fused to the curli subunit CsgA. Secretion efficiency depended on the folding properties of the passenger sequences, with substrates exceeding an approximately 2 nm transverse diameter blocking passage through the transport channel. Secretion of smaller passengers was compatible with prior DsbA-mediated disulphide bridge formation in the fusion partner, indicating that CsgG is capable of translocating non-linear polypeptide stretches. Using fusions we further demonstrate the exported or secreted heterologous passenger proteins can attain their native, active fold, establishing curli biogenesis pathway as a platform for the secretion and surface display of small heterologous proteins.


Assuntos
Amiloide/metabolismo , Sistemas de Secreção Bacterianos , Vias Biossintéticas , Escherichia coli/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Amiloide/ultraestrutura , Western Blotting , Membrana Celular/metabolismo , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Peptídeos/metabolismo , Estrutura Secundária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/ultraestrutura , Especificidade por Substrato
18.
Proteins ; 82(9): 1694-707, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24464835

RESUMO

Ion channel-coupled receptors (ICCR) are artificial proteins built from a G protein-coupled receptor and an ion channel. Their use as molecular biosensors is promising in diagnosis and high-throughput drug screening. The concept of ICCR was initially validated with the combination of the muscarinic receptor M2 with the inwardly rectifying potassium channel Kir6.2. A long protein engineering phase has led to the biochemical characterization of the M2-Kir6.2 construct. However, its molecular mechanism remains to be elucidated. In particular, it is important to determine how the activation of M2 by its agonist acetylcholine triggers the modulation of the Kir6.2 channel via the M2-Kir6.2 linkage. In the present study, we have developed and validated a computational approach to rebuild models of the M2-Kir6.2 chimera from the molecular structure of M2 and Kir6.2. The protocol was first validated on the known protein complexes of the µ-opioid Receptor, the CXCR4 receptor and the Kv1.2 potassium channel. When applied to M2-Kir6.2, our protocol produced two possible models corresponding to two different orientations of M2. Both models highlights the role of the M2 helices I and VIII in the interaction with Kir6.2, as well as the role of the Kir6.2 N-terminus in the channel opening. Those two hypotheses will be explored in a future experimental study of the M2-Kir6.2 construct.


Assuntos
Complexos Multiproteicos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptor Muscarínico M2/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Técnicas Biossensoriais , Ativação do Canal Iônico , Simulação de Acoplamento Molecular , Complexos Multiproteicos/ultraestrutura , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/ultraestrutura , Engenharia de Proteínas , Receptor Muscarínico M2/ultraestrutura , Receptores CXCR4/metabolismo , Receptores Opioides mu/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura
19.
Methods Cell Biol ; 117: 1-19, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24143969

RESUMO

This chapter presents a general approach for the application of spatial intensity distribution analysis (SpIDA) to pharmacodynamic quantification of receptor tyrosine kinase homodimerization in response to direct ligand activation or transactivation by G protein-coupled receptors. A custom graphical user interface developed for MATLAB is used to extract quantal brightness and receptor density information from intensity histograms calculated from single fluorescence microscopy images. This approach allows measurement of monomer/oligomer protein mixtures within subcellular compartments using conventional confocal laser scanning microscopy. Application of quantitative pharmacological analysis to data obtained using SpIDA provides a universal method for comparing studies between cell lines and receptor systems. In addition, because of its compatibility with conventional immunostaining approaches, SpIDA is suitable not only for use in recombinant systems but also for the characterization of mechanisms involving endogenous proteins. Therefore, SpIDA enables these biological processes to be monitored directly in their native cellular environment.


Assuntos
Receptores ErbB/metabolismo , Imagem Molecular/métodos , Neurônios/metabolismo , Receptor trkB/metabolismo , Receptores Dopaminérgicos/metabolismo , Software , Apomorfina/farmacologia , Linhagem Celular , Receptores ErbB/genética , Receptores ErbB/ultraestrutura , Imunofluorescência , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Ligantes , Microscopia Confocal , Microscopia de Fluorescência , Imagem Molecular/estatística & dados numéricos , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Multimerização Proteica , Quinazolinas/farmacologia , Receptor trkB/genética , Receptor trkB/ultraestrutura , Receptores Dopaminérgicos/genética , Receptores Dopaminérgicos/ultraestrutura , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Ativação Transcricional , Tirfostinas/farmacologia
20.
ACS Nano ; 7(9): 7734-43, 2013 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-23987654

RESUMO

Self-assembly of amyloid nanofiber is associated with both functional biological and pathological processes such as those in neurodegenerative diseases. Despite intensive studies, the stochastic nature of the process has made it difficult to elucidate a molecular mechanism for the key amyloid nucleation event. Here we investigated nucleation of the silk-elastin-like peptide (SELP) amyloid using time-lapse lateral force microscopy (LFM). By repeated scanning of a single line on a SELP-coated mica surface, we observed a sudden stepwise height increase. This corresponds to nucleation of an amyloid fiber, which subsequently grew perpendicular to the scanning direction. The lateral force profiles followed either a worm-like chain model or an exponential function, suggesting that the atomic force microscopy (AFM) tip stretches a single or multiple SELP molecules along the scanning direction. The probability of nucleation correlated with the maximum stretching force and extension, implying that stretching of SELP molecules is a key molecular event for amyloid nucleation. The mechanically induced nucleation allows for positional and directional control of amyloid assembly in vitro, which we demonstrate by generating single nanofibers at predetermined nucleation sites.


Assuntos
Proteínas Amiloidogênicas/química , Proteínas Amiloidogênicas/ultraestrutura , Cristalização/métodos , Micromanipulação/métodos , Microscopia de Força Atômica/métodos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/ultraestrutura , Dimerização , Módulo de Elasticidade , Conformação Proteica , Estresse Mecânico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...