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.
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
3.
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
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.
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
6.
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
7.
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
8.
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
9.
Proc Natl Acad Sci U S A ; 110(3): 960-5, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23277578

RESUMO

In eukaryotes, the nucleocytoplasmic transport of macromolecules is mainly mediated by soluble nuclear transport receptors of the karyopherin-ß superfamily termed importins and exportins. The highly versatile exportin chromosome region maintenance 1 (CRM1) is essential for nuclear depletion of numerous structurally and functionally unrelated protein and ribonucleoprotein cargoes. CRM1 has been shown to adopt a toroidal structure in several functional transport complexes and was thought to maintain this conformation throughout the entire nucleocytoplasmic transport cycle. We solved crystal structures of free CRM1 from the thermophilic eukaryote Chaetomium thermophilum. Surprisingly, unbound CRM1 exhibits an overall extended and pitched superhelical conformation. The two regulatory regions, namely the acidic loop and the C-terminal α-helix, are dramatically repositioned in free CRM1 in comparison with the ternary CRM1-Ran-Snurportin1 export complex. Single-particle EM analysis demonstrates that, in a noncrystalline environment, free CRM1 exists in equilibrium between extended, superhelical and compact, ring-like conformations. Molecular dynamics simulations show that the C-terminal helix plays an important role in regulating the transition from an extended to a compact conformation and reveal how the binding site for nuclear export signals of cargoes is modulated by different CRM1 conformations. Combining these results, we propose a model for the cooperativity of CRM1 export complex assembly involving the long-range allosteric communication between the distant binding sites of GTP-bound Ran and cargo.


Assuntos
Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Carioferinas/química , Carioferinas/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Regulação Alostérica , Sequência de Aminoácidos , Sítios de Ligação , Chaetomium/química , Chaetomium/genética , Chaetomium/metabolismo , Cristalografia por Raios X , Proteínas Fúngicas/genética , Proteínas Fúngicas/ultraestrutura , Carioferinas/genética , Carioferinas/ultraestrutura , Microscopia Eletrônica , Modelos Biológicos , Modelos Moleculares , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Conformação Proteica , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/ultraestrutura , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Proteína Exportina 1
10.
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
11.
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
12.
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
13.
Plant Cell ; 23(6): 2302-13, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21693695

RESUMO

In eukaryotic cells, the actin and microtubule (MT) cytoskeletal networks are dynamic structures that organize intracellular processes and facilitate their rapid reorganization. In plant cells, actin filaments (AFs) and MTs are essential for cell growth and morphogenesis. However, dynamic interactions between these two essential components in live cells have not been explored. Here, we use spinning-disc confocal microscopy to dissect interaction and cooperation between cortical AFs and MTs in Arabidopsis thaliana, utilizing fluorescent reporter constructs for both components. Quantitative analyses revealed altered AF dynamics associated with the positions and orientations of cortical MTs. Reorganization and reassembly of the AF array was dependent on the MTs following drug-induced depolymerization, whereby short AFs initially appeared colocalized with MTs, and displayed motility along MTs. We also observed that light-induced reorganization of MTs occurred in concert with changes in AF behavior. Our results indicate dynamic interaction between the cortical actin and MT cytoskeletons in interphase plant cells.


Assuntos
Citoesqueleto de Actina/metabolismo , Arabidopsis/citologia , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Microtúbulos/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Antineoplásicos/farmacologia , Arabidopsis/efeitos dos fármacos , Arabidopsis/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Citoesqueleto/efeitos dos fármacos , Depsipeptídeos/farmacologia , Microtúbulos/ultraestrutura , Células Vegetais/efeitos dos fármacos , Células Vegetais/metabolismo , Células Vegetais/ultraestrutura , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Proteínas de Plantas/ultraestrutura , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Tiazolidinas/farmacologia
14.
PLoS Pathog ; 7(5): e1002030, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21589902

RESUMO

Rift Valley fever virus (RVFV), a Phlebovirus with a genome consisting of three single-stranded RNA segments, is spread by infected mosquitoes and causes large viral outbreaks in Africa. RVFV encodes a nucleoprotein (N) that encapsidates the viral RNA. The N protein is the major component of the ribonucleoprotein complex and is also required for genomic RNA replication and transcription by the viral polymerase. Here we present the 1.6 Å crystal structure of the RVFV N protein in hexameric form. The ring-shaped hexamers form a functional RNA binding site, as assessed by mutagenesis experiments. Electron microscopy (EM) demonstrates that N in complex with RNA also forms rings in solution, and a single-particle EM reconstruction of a hexameric N-RNA complex is consistent with the crystallographic N hexamers. The ring-like organization of the hexamers in the crystal is stabilized by circular interactions of the N terminus of RVFV N, which forms an extended arm that binds to a hydrophobic pocket in the core domain of an adjacent subunit. The conformation of the N-terminal arm differs from that seen in a previous crystal structure of RVFV, in which it was bound to the hydrophobic pocket in its own core domain. The switch from an intra- to an inter-molecular interaction mode of the N-terminal arm may be a general principle that underlies multimerization and RNA encapsidation by N proteins from Bunyaviridae. Furthermore, slight structural adjustments of the N-terminal arm would allow RVFV N to form smaller or larger ring-shaped oligomers and potentially even a multimer with a super-helical subunit arrangement. Thus, the interaction mode between subunits seen in the crystal structure would allow the formation of filamentous ribonucleocapsids in vivo. Both the RNA binding cleft and the multimerization site of the N protein are promising targets for the development of antiviral drugs.


Assuntos
Proteínas do Nucleocapsídeo/química , Multimerização Proteica , RNA Viral/química , Ribonucleoproteínas/química , Vírus da Febre do Vale do Rift/fisiologia , Sequência de Aminoácidos , Animais , Cristalografia por Raios X/métodos , DNA Complementar/genética , Humanos , Microscopia Eletrônica , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/isolamento & purificação , Proteínas do Nucleocapsídeo/ultraestrutura , Domínios e Motivos de Interação entre Proteínas , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/ultraestrutura , Vírus da Febre do Vale do Rift/química , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/ultraestrutura , Alinhamento de Sequência , Ressonância de Plasmônio de Superfície/métodos , Montagem de Vírus
15.
Proc Natl Acad Sci U S A ; 107(26): 11769-74, 2010 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-20547879

RESUMO

Rift Valley fever virus (RVFV) is a negative-sense RNA virus (genus Phlebovirus, family Bunyaviridae) that infects livestock and humans and is endemic to sub-Saharan Africa. Like all negative-sense viruses, the segmented RNA genome of RVFV is encapsidated by a nucleocapsid protein (N). The 1.93-A crystal structure of RVFV N and electron micrographs of ribonucleoprotein (RNP) reveal an encapsidated genome of substantially different organization than in other negative-sense RNA virus families. The RNP polymer, viewed in electron micrographs of both virus RNP and RNP reconstituted from purified N with a defined RNA, has an extended structure without helical symmetry. N-RNA species of approximately 100-kDa apparent molecular weight and heterogeneous composition were obtained by exhaustive ribonuclease treatment of virus RNP, by recombinant expression of N, and by reconstitution from purified N and an RNA oligomer. RNA-free N, obtained by denaturation and refolding, has a novel all-helical fold that is compact and well ordered at both the N and C termini. Unlike N of other negative-sense RNA viruses, RVFV N has no positively charged surface cleft for RNA binding and no protruding termini or loops to stabilize a defined N-RNA oligomer or RNP helix. A potential protein interaction site was identified in a conserved hydrophobic pocket. The nonhelical appearance of phlebovirus RNP, the heterogeneous approximately 100-kDa N-RNA multimer, and the N fold differ substantially from the RNP and N of other negative-sense RNA virus families and provide valuable insights into the structure of the encapsidated phlebovirus genome.


Assuntos
Proteínas do Nucleocapsídeo/química , RNA Viral/química , Vírus da Febre do Vale do Rift/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Sequência Conservada , Cristalografia por Raios X , Primers do DNA/genética , Humanos , Substâncias Macromoleculares/química , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/ultraestrutura , Phlebovirus/genética , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , RNA Viral/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/fisiologia , Vírus da Febre do Vale do Rift/ultraestrutura , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Montagem de Vírus
16.
Biomacromolecules ; 13(2): 304-12, 2012 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-22176138

RESUMO

Spider silk is renowned for its extraordinary mechanical properties, having a balance of high tensile strength and extensibility. To date, the majority of studies have focused on the production of dragline silks from synthetic spider silk gene products. Here we report the first mechanical analysis of synthetic egg case silk fibers spun from the Latrodectus hesperus tubuliform silk proteins, TuSp1 and ECP-2. We provide evidence that recombinant ECP-2 proteins can be spun into fibers that display mechanical properties similar to other synthetic spider silks. We also demonstrate that silks spun from recombinant thioredoxin-TuSp1 fusion proteins that contain the conserved C-terminal domain exhibit increased extensibility and toughness when compared to the identical fibers spun from fusion proteins lacking the C-terminus. Mechanical analyses reveal that the properties of synthetic tubuliform silks can be modulated by altering the postspin draw ratios of the fibers. Fibers subject to increased draw ratios showed elevated tensile strength and decreased extensibility but maintained constant toughness. Wide-angle X-ray diffraction studies indicate that postdrawn fibers containing the C-terminal domain of TuSp1 have more amorphous content when compared to fibers lacking the C-terminus. Taken together, these studies demonstrate that recombinant tubuliform spidroins that contain the conserved C-terminal domain with embedded protein tags can be effectively spun into fibers, resulting in similar tensile strength but increased extensibility relative to nontagged recombinant dragline silk proteins spun from equivalently sized proteins.


Assuntos
Fibroínas/química , Proteínas Recombinantes de Fusão/química , Seda/biossíntese , Sequência de Aminoácidos , Animais , Clonagem Molecular , Sequência Conservada , DNA Complementar/química , DNA Complementar/genética , Elasticidade , Escherichia coli , Fibroínas/genética , Microscopia Eletrônica de Varredura , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Homologia de Sequência de Aminoácidos , Seda/ultraestrutura , Aranhas , Resistência à Tração , Difração de Raios X
17.
Proc Natl Acad Sci U S A ; 106(34): 14339-44, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19706519

RESUMO

We demonstrate that accurate values of mass-per-length (MPL), which serve as strong constraints on molecular structure, can be determined for amyloid fibrils by quantification of intensities in dark-field electron microscope images obtained in the tilted-beam mode of a transmission electron microscope. MPL values for fibrils formed by residues 218-289 of the HET-s fungal prion protein, for 2-fold- and 3-fold-symmetric fibrils formed by the 40-residue beta-amyloid peptide, and for fibrils formed by the yeast prion protein Sup35NM are in good agreement with previous results from scanning transmission electron microscopy. Results for fibrils formed by the yeast prion protein Rnq1, for which the MPL value has not been previously reported, support an in-register parallel beta-sheet structure, with one Rnq1 molecule per 0.47-nm beta-sheet repeat spacing. Since tilted-beam dark-field images can be obtained on many transmission electron microscopes, this work should facilitate MPL determination by a large number of research groups engaged in studies of amyloid fibrils and similar supramolecular assemblies.


Assuntos
Peptídeos beta-Amiloides/ultraestrutura , Microscopia Eletrônica de Transmissão/métodos , Fragmentos de Peptídeos/ultraestrutura , Peptídeos beta-Amiloides/química , Proteínas Fúngicas/química , Proteínas Fúngicas/ultraestrutura , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Fluorescência Verde/ultraestrutura , Humanos , Peso Molecular , Fragmentos de Peptídeos/química , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/ultraestrutura , Príons/química , Príons/ultraestrutura , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/ultraestrutura
18.
Proc Natl Acad Sci U S A ; 106(52): 22193-8, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20018767

RESUMO

Stable, single alpha-helix (SAH) domains are widely distributed in the proteome, including in myosins, but their functions are unknown. To test whether SAH domains can act as levers, we replaced four of the six calmodulin-binding IQ motifs in the levers of mouse myosin 5a (Myo5) with the putative SAH domain of Dictyostelium myosin MyoM of similar length. The SAH domain was inserted between the IQ motifs and the coiled coil in a Myo5 HMM construct in which the levers were truncated from six to two IQ motifs (Myo5-2IQ). Electron microscopy of this chimera (Myo5-2IQ-SAH) showed the SAH domain was straight and 17 nm long as predicted, restoring the truncated lever to the length of wild-type (Myo5-6IQ). The powerstroke (of 21.5 nm) measured in the optical trap was slightly less than that for Myo5-6IQ but much greater than for Myo5-2IQ. Myo5-2IQ-SAH moved processively along actin at physiological ATP concentrations with similar stride and run lengths to Myo5-6IQ in in-vitro single molecule assays. In comparison, Myo5-2IQ is not processive under these conditions. Solution biochemical experiments indicated that the rear head did not mechanically gate the rate of ADP release from the lead head, unlike Myo5-6IQ. These data show that the SAH domain can form part of a functional lever in myosins, although its mechanical stiffness might be lower. More generally, we conclude that SAH domains can act as stiff structural extensions in aqueous solution and this structural role may be important in other proteins.


Assuntos
Miosinas/química , Actinas/metabolismo , Difosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Recombinante/genética , Técnicas In Vitro , Camundongos , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Cadeias Pesadas de Miosina/ultraestrutura , Miosina Tipo V/química , Miosina Tipo V/genética , Miosina Tipo V/metabolismo , Miosina Tipo V/ultraestrutura , Miosinas/genética , Miosinas/metabolismo , Miosinas/ultraestrutura , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/ultraestrutura , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura
19.
Yao Xue Xue Bao ; 47(1): 116-23, 2012 Jan.
Artigo em Zh | MEDLINE | ID: mdl-22493816

RESUMO

This study is to report the preparation of complexes of Ad5 and anionic liposomes (AL-Ad5), the amplification of adenoviruses with enhanced green fluorescent protein (eGFP) reporter gene performed by HEK 293 cells, the adenoviral vectors purified by cesium chloride gradient centrifugation, and the titer of adenovirus determined by cytopathic effect (CPE) method, hexon capsid immunoassay and quantitative-PCR (Q-PCR), separately. The prescription and experiment conditions were optimized by central composite design (CCD). The complexes of Ad5 and AL-Ad5 were formulated by the calcium-induced phase change method. The morpholopy, particle size and zeta potential were detected by dynamic light scattering (DLS) and transmission electron microscopy (TEM), respectively. Additionally, the bicolourable fluoresce-labeled complexes (F(labeled)-AL-Ad5) were prepared and their intracellular location in MDCK cells was detected by confocal laser scanning microscopy (CLSM). The results indicate that the complexes of AL-Ad5 exhibited a uniform distribution with a particle size of 211 +/- 10 nm and a zeta potential of -41.2 +/- 2.2 mV. The result of CLSM demonstrates that the intracellular location of red fluoresce-labeled adenovirus was consistent with that of green fluoresce-labeled liposomes suggesting that the naked adenovirus was well encapsulated by the anionic liposomes in complexes of AL-Ad5.


Assuntos
Adenoviridae/ultraestrutura , Composição de Medicamentos/métodos , Lipossomos/ultraestrutura , Adenoviridae/genética , Animais , Ânions , Efeito Citopatogênico Viral , Cães , Vetores Genéticos , Proteínas de Fluorescência Verde/química , Células HEK293 , Humanos , Lipossomos/química , Lipossomos/farmacocinética , Células Madin Darby de Rim Canino , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Tamanho da Partícula , Reação em Cadeia da Polimerase/métodos , Proteínas Recombinantes de Fusão/ultraestrutura
20.
Arch Biochem Biophys ; 506(1): 99-108, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21094125

RESUMO

Amebin [formerly termed as ApABP-FI; Sobczak et al. (2007) Biochem. Cell Biol. 85] is encoded in Amoeba proteus by two transcripts, 2672-nt and 1125-nt. A product of the shorter transcript (termed as C-amebin), comprising C-terminal 375 amino-acid-residue fragment of amebin, has been expressed and purified as the recombinant GST-fusion protein. GST-C-amebin bound both to monomeric and filamentous actin. The binding was Ca(2+)-independent and promoted filament bundling, as revealed with the transmission electron microscopy. GST-C-amebin significantly decreased MgATPase activity of rabbit skeletal muscle acto-S1. Removal with endoproteinase ArgC of a positively charged C-terminal region of GST-amebin containing KLASMWEQ sequence abolished actin-binding and bundling as well as the ATPase-inhibitory effect of C-amebin, indicating that this protein region was involved in the interaction with actin. Microinjection of amoebae with antibody against C-terminus of amebin significantly affected amoebae morphology, disturbed cell polarization and transport of cytoplasmic granules as well as blocked migration. These data indicate that amebin may be one of key regulators of the actin-cytoskeleton dynamics and actin-dependent motility in A. proteus.


Assuntos
Citoesqueleto de Actina/metabolismo , Amoeba/química , Amoeba/fisiologia , Miosinas/antagonistas & inibidores , Proteínas de Protozoários/química , Proteínas de Protozoários/fisiologia , Citoesqueleto de Actina/química , Citoesqueleto de Actina/ultraestrutura , Sequência de Aminoácidos , Amoeba/genética , Animais , Técnicas In Vitro , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Movimento/fisiologia , Complexos Multiproteicos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Fragmentos de Peptídeos/ultraestrutura , Ligação Proteica , Proteínas de Protozoários/genética , Proteínas de Protozoários/ultraestrutura , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA