Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
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
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.
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
4.
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
5.
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
6.
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
7.
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
8.
Acta Biomater ; 9(6): 6806-13, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23485856

RESUMO

Chimeric growth factors may represent a powerful alternative to their natural counterparts for the functionalization of tissue-engineered scaffolds and applications in regenerative medicine. Their rational design should provide a simple, readily scalable production strategy while improving retention at the site of action. In that endeavor, we here report the synthesis of a chimeric protein corresponding to human vascular endothelial growth factor 165 being N-terminally fused to an E5 peptide tag (E5-VEGF). E5-VEGF was successfully expressed as a homodimer in mammalian cells. Following affinity purification, in vitro surface plasmon resonance biosensing and cell survival assays confirmed diffusible E5-VEGF ability to bind to its receptor ectodomains, while observed morphological phenotypes confirmed its anti-apoptotic features. Additional surface plasmon resonance assays highlighted that E5-VEGF could be specifically captured with high stability when interacting with covalently immobilized K5 peptide (a synthetic peptide designed to bind to the E5 moiety of chimeric hVEGF). This immobilization strategy was applied to glass substrates and chimeric hVEGF was shown to be maintained in a functionally active state following capture. Altogether, our data demonstrated that stable hVEGF capture can be performed via coiled-coil interactions without impacting hVEGF bioactivity, thus opening up the way to future applications in the field of tissue engineering and regenerative medicine.


Assuntos
Proteínas de Fluorescência Verde/química , Engenharia de Proteínas/métodos , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/ultraestrutura , Proteínas Recombinantes de Fusão/síntese química , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/ultraestrutura , Sítios de Ligação , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/ultraestrutura , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Fator A de Crescimento do Endotélio Vascular/genética
9.
FEBS Lett ; 586(21): 3799-804, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23010594

RESUMO

Aminoacylase 3 (AA3) mediates deacetylation of N-acetyl aromatic amino acids and mercapturic acids. Deacetylation of mercapturic acids of exo- and endobiotics are likely involved in their toxicity. AA3 is predominantly expressed in kidney, and to a lesser extent in liver, brain, and blood. AA3 has been recently reported to interact with the hepatitis C virus core protein (HCVCP) in the yeast two-hybrid system. Here we demonstrate that AA3 directly binds to HCVCP (K(d) ~10 µM) that may by implicated in HCV pathogenesis. AA3 also revealed a weak endopeptidase activity towards the N-terminus of HCVCP.


Assuntos
Amidoidrolases/química , Hepacivirus/química , Proteínas do Core Viral/química , Amidoidrolases/genética , Amidoidrolases/ultraestrutura , Sequência de Aminoácidos , Animais , Escherichia coli/genética , Humanos , Cinética , Camundongos , Microscopia Eletrônica , Dados de Sequência Molecular , Ligação Proteica , Proteólise , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Homologia de Sequência de Aminoácidos , Soluções , Ressonância de Plasmônio de Superfície , Técnicas do Sistema de Duplo-Híbrido , Proteínas do Core Viral/genética , Proteínas do Core Viral/ultraestrutura
10.
Yao Xue Xue Bao ; 47(1): 116-23, 2012 Jan.
Artigo em Chinês | 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
11.
PLoS One ; 7(3): e31253, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22438863

RESUMO

Nine neurodegenerative disorders, called polyglutamine (polyQ) diseases, are characterized by the formation of intranuclear amyloid-like aggregates by nine proteins containing a polyQ tract above a threshold length. These insoluble aggregates and/or some of their soluble precursors are thought to play a role in the pathogenesis. The mechanism by which polyQ expansions trigger the aggregation of the relevant proteins remains, however, unclear. In this work, polyQ tracts of different lengths were inserted into a solvent-exposed loop of the ß-lactamase BlaP and the effects of these insertions on the properties of BlaP were investigated by a range of biophysical techniques. The insertion of up to 79 glutamines does not modify the structure of BlaP; it does, however, significantly destabilize the enzyme. The extent of destabilization is largely independent of the polyQ length, allowing us to study independently the effects intrinsic to the polyQ length and those related to the structural integrity of BlaP on the aggregating properties of the chimeras. Only chimeras with 55Q and 79Q readily form amyloid-like fibrils; therefore, similarly to the proteins associated with diseases, there is a threshold number of glutamines above which the chimeras aggregate into amyloid-like fibrils. Most importantly, the chimera containing 79Q forms amyloid-like fibrils at the same rate whether BlaP is folded or not, whereas the 55Q chimera aggregates into amyloid-like fibrils only if BlaP is unfolded. The threshold value for amyloid-like fibril formation depends, therefore, on the structural integrity of the ß-lactamase moiety and thus on the steric and/or conformational constraints applied to the polyQ tract. These constraints have, however, no significant effect on the propensity of the 79Q tract to trigger fibril formation. These results suggest that the influence of the protein context on the aggregating properties of polyQ disease-associated proteins could be negligible when the latter contain particularly long polyQ tracts.


Assuntos
Amiloide/genética , Amiloide/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Sequência de Aminoácidos , Amiloide/química , Amiloide/ultraestrutura , Bacillus/enzimologia , Bacillus/genética , Cristalografia por Raios X , Estabilidade Enzimática , Humanos , Técnicas In Vitro , Cinética , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Dados de Sequência Molecular , Doenças Neurodegenerativas/etiologia , Peptídeos/química , Conformação Proteica , Desnaturação Proteica , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Termodinâmica , Expansão das Repetições de Trinucleotídeos , beta-Lactamases/química , beta-Lactamases/genética , beta-Lactamases/metabolismo , beta-Lactamases/ultraestrutura
12.
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
13.
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
14.
FEBS Lett ; 584(13): 2786-90, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20471980

RESUMO

Human respiratory syncytial virus (HRSV) is the leading cause of lower respiratory tract disease in infants. The HRSV small hydrophobic (SH) protein plays an important role in HRSV pathogenesis, although its mode of action is unclear. Analysis of the ability of SH protein to induce membrane permeability and form homo-oligomers suggests it acts as a viroporin. For the first time, we directly observed functional SH protein using electron microscopy, which revealed SH forms multimeric ring-like objects with a prominent central stained region. Based on current and existing functional data, we propose this region represents the channel that mediates membrane permeability.


Assuntos
Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Vírus Sincicial Respiratório Humano/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Proteínas Oncogênicas de Retroviridae/ultraestrutura , Western Blotting , Cromatografia Líquida de Alta Pressão , Eletroforese em Gel de Poliacrilamida , Lipossomos/química , Microscopia Eletrônica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/genética
15.
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
16.
Methods Cell Biol ; 91: 41-61, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20409779

RESUMO

Despite more than 40 years of investigation since the discovery of dynein [Gibbons, I. R. and Rowe, A. J. (1965). Science149, 424-426] our understanding of how this microtubule-based motor generates force and movement remains frustratingly incomplete at the atomic level. Electron microscopy (EM) has played a major role in establishing dynein's complex architecture and its nucleotide-dependent conformational changes. In this chapter we review recent structural studies and describe in detail negative stain EM and computational single-particle image processing techniques that have been used to investigate dynein. We describe studies of both Chlamydomonas flagellar inner arm dynein-c and recombinant cytoplasmic dynein from Dictyostelium. We also detail methods for locating green fluorescent protein (GFP) and blue fluorescent protein (BFP) tags inserted at specific locations within the dynein motor, which can be used to map subdomains and conformational changes.


Assuntos
Dineínas/ultraestrutura , Processamento de Imagem Assistida por Computador/métodos , Microscopia Eletrônica/métodos , Microscopia Crioeletrônica/instrumentação , Microscopia Crioeletrônica/métodos , Dineínas/genética , Microscopia Eletrônica/instrumentação , Modelos Moleculares , Conformação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura
17.
Arch Virol ; 153(11): 2027-39, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18979257

RESUMO

Asymmetrical-flow field flow fractionation with multiple-angle light scattering (AFFFF-MALS) was, for the first time, used to characterize the size of murine polyomavirus virus-like particles (MPV VLPs) packaged with either insect cell genomic DNA or non-viral protein. Encapsidation of both genomic DNA and non-viral protein were found to cause a contraction in VLP radii of gyration by approximately 1 nm. Non-viral protein packaged into VLPs consisted of a series of glutathione-S-transferase, His and S tags attached to the N-terminal end of the MPV structural protein VP2 (M(r) = 67108). Transmission electron microscopy analysis of MPV VLPs packaging non-viral protein suggested that VLPs grew in diameter by approximately 5 nm, highlighting the differences between this invasive technique and the relatively non-invasive AFFFF-MALS technique. Encapsulation of non-viral protein into MPV VLPs was found to prevent co-encapsidation of genomic DNA. Further investigation into why this occurred led to the discovery that encapsulation of non-viral protein alters the nuclear localization of MPV VLPs during in vivo assembly. VLPs were relocated away from the ring zone and the nuclear membrane towards the centre of the nucleus amongst the virogenic stroma. The change in nuclear localization away from the site where VLP assembly usually occurs is a likely reason why encapsidation of genomic DNA did not take place.


Assuntos
Genoma de Inseto , Polyomavirus/química , Polyomavirus/fisiologia , Montagem de Vírus , Animais , Linhagem Celular , DNA Viral/genética , Glutationa Transferase/química , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Insetos , Microscopia Eletrônica de Transmissão , Polyomavirus/genética , Polyomavirus/ultraestrutura , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/isolamento & purificação , Proteínas Virais/metabolismo , Proteínas Virais/ultraestrutura , Vírion/química , Vírion/genética , Vírion/fisiologia , Vírion/ultraestrutura
18.
J Struct Biol ; 160(3): 287-94, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17959389

RESUMO

Mitochondrial F(1)F(o)-ATP synthase is a molecular motor that couples the energy generated by oxidative metabolism to the synthesis of ATP. Direct visualization of the rotary action of the bacterial ATP synthase has been well characterized. However, direct observation of rotation of the mitochondrial enzyme has not been reported yet. Here, we describe two methods to reconstitute mitochondrial F(1)F(o)-ATP synthase into lipid bilayers suitable for structure analysis by electron and atomic force microscopy (AFM). Proteoliposomes densely packed with bovine heart mitochondria F(1)F(o)-ATP synthase were obtained upon detergent removal from ternary mixtures (lipid, detergent and protein). Two-dimensional crystals of recombinant hexahistidine-tagged yeast F(1)F(o)-ATP synthase were grown using the supported monolayer technique. Because the hexahistidine-tag is located at the F(1) catalytic subcomplex, ATP synthases were oriented unidirectionally in such two-dimensional crystals, exposing F(1) to the lipid monolayer and the F(o) membrane region to the bulk solution. This configuration opens a new avenue for the determination of the c-ring stoichiometry of unknown hexahistidine-tagged ATP synthases and the organization of the membrane intrinsic subunits within F(o) by electron microscopy and AFM.


Assuntos
Proteolipídeos/química , ATPases Translocadoras de Prótons/química , Proteínas de Saccharomyces cerevisiae/química , Animais , Bovinos , Cristalização , Histidina/química , Processamento de Imagem Assistida por Computador , Bicamadas Lipídicas , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Mitocôndrias/enzimologia , Mitocôndrias Cardíacas/enzimologia , Conformação Proteica , Subunidades Proteicas , Proteolipídeos/ultraestrutura , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/isolamento & purificação , ATPases Translocadoras de Prótons/ultraestrutura , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/isolamento & purificação , Proteínas de Saccharomyces cerevisiae/ultraestrutura
19.
Biochemistry ; 46(38): 11013-22, 2007 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-17718511

RESUMO

FtsZ polymerizes to form a dynamic ring structure called the Z-ring at the midcell of bacteria. EzrA, a membrane protein, has been shown to prevent the formation of aberrant Z-rings in the low GC Gram-positive bacteria by inhibiting FtsZ assembly. In this study, we show that Bacillus subtilis (B. subtilis) EzrA inhibited the assembly and bundling of B. subtilis FtsZ. It increased the critical concentration of FtsZ assembly and depolymerized the preformed FtsZ polymers in vitro. We obtained evidence suggesting that B. subtilis EzrA forms complex with B. subtilis FtsZ in vitro. EzrA was found to bind to FtsZ at a single site with a dissociation constant of 4.3 +/- 0.6 microM. EzrA-FtsZ interaction has a significant electrostatic contribution as apparent from the effect of salt on their binding interactions. To elucidate the site of interaction between EzrA and FtsZ, we deleted 16 amino acid residues from the extreme C-terminal tail of B. subtilis FtsZ, which are conserved in FtsZ orthologues. EzrA did not inhibit the assembly of C-terminal truncated B. subtilis FtsZ. It also did not bind to the C-terminal truncated FtsZ detectably, suggesting that EzrA interacts with FtsZ through its conserved C-terminal tail residues. Further, a 17-residue synthetic peptide (365-382) of the C-terminal tail of FtsZ (CTP17) was used to probe the interaction of EzrA with the C-terminal tail of FtsZ. CTP17 bound to EzrA, inhibited the binding of EzrA to FtsZ, and surmounted the inhibitory effects of EzrA on the assembly of FtsZ in vitro. The data together showed that EzrA binds to the C-terminal tail of FtsZ. FtsA, a positive regulator of FtsZ assembly, is also known to interact with the C-terminal tail of FtsZ. The results indicated an interesting possibility that the assembly dynamics of FtsZ in the Z-ring is regulated by the competition between positive and negative regulators sharing the same binding site on FtsZ.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sequência de Aminoácidos , Bacillus subtilis/metabolismo , Bacillus subtilis/ultraestrutura , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/ultraestrutura , Sítios de Ligação , Ligação Competitiva , Divisão Celular/fisiologia , Citocinese/efeitos dos fármacos , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/ultraestrutura , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Regulação Bacteriana da Expressão Gênica , Guanosina Trifosfato/metabolismo , Hidrólise , Cinética , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Mutação , Peptídeos/síntese química , Peptídeos/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/ultraestrutura , Deleção de Sequência
20.
FEBS Lett ; 581(14): 2702-8, 2007 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-17531226

RESUMO

The Coxsackie and adenovirus receptor (CAR) is a cell adhesion molecule that is highly expressed in the developing brain. CAR is enriched in growth cone particles (GCP) after subcellular fractionation. In GCP, we identified actin as an interaction partner of the cytoplasmic domain of CAR. In vivo, actin and CAR co-immunoprecipitate and co-localize. In vitro, the binding is direct, with a K(d) of approximately 2.6 microM, and leads to actin bundling. We previously demonstrated that CAR interacts with microtubules. These data suggest a role for CAR in processes requiring dynamic reorganization of the cytoskeleton such as neurite outgrowth and cell migration.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Receptores Virais/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Cones de Crescimento/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Imunoprecipitação , Cinética , Camundongos , Microscopia Eletrônica , Microscopia de Fluorescência , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/metabolismo , Ligação Proteica , Receptores Virais/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA