Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Structure ; 30(9): 1254-1268.e7, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35870450

RESUMO

The mitochondrial Lon protease (LonP1) regulates mitochondrial health by removing redundant proteins from the mitochondrial matrix. We determined LonP1 in eight nucleotide-dependent conformational states by cryoelectron microscopy (cryo-EM). The flexible assembly of N-terminal domains had 3-fold symmetry, and its orientation depended on the conformational state. We show that a conserved structural motif around T803 with a high similarity to the trypsin catalytic triad is essential for proteolysis. We show that LonP1 is not regulated by redox potential, despite the presence of two conserved cysteines at disulfide-bonding distance in its unfoldase core. Our data indicate how sequential ATP hydrolysis controls substrate protein translocation in a 6-fold binding change mechanism. Substrate protein translocation, rather than ATP hydrolysis, is a rate-limiting step, suggesting that LonP1 is a Brownian ratchet with ATP hydrolysis preventing translocation reversal. 3-fold rocking motions of the flexible N-domain assembly may assist thermal unfolding of the substrate protein.


Assuntos
Protease La , Proteases Dependentes de ATP/genética , Proteases Dependentes de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Microscopia Crioeletrônica , Humanos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Protease La/genética , Protease La/metabolismo
2.
J Am Chem Soc ; 142(47): 19956-19968, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33170675

RESUMO

The fabrication of dynamic, transformable biomaterials that respond to environmental cues represents a significant step forward in the development of synthetic materials that rival their highly functional, natural counterparts. Here, we describe the design and synthesis of crystalline supramolecular architectures from charge-complementary heteromeric pairs of collagen-mimetic peptides (CMPs). Under appropriate conditions, CMP pairs spontaneously assemble into either 1D ultraporous (pore diameter >100 nm) tubes or 2D bilayer nanosheets due to the structural asymmetry that arises from heteromeric self-association. Crystalline collagen tubes represent a heretofore unobserved morphology of this common biomaterial. In-depth structural characterization from a suite of biophysical methods, including TEM, AFM, high-resolution cryo-EM, and SAXS/WAXS measurements, reveals that the sheet and tube assemblies possess a similar underlying lattice structure. The experimental evidence suggests that the tubular structures are a consequence of the self-scrolling of incipient 2D layers of collagen triple helices and that the scrolling direction determines the formation of two distinct structural isoforms. Furthermore, we show that nanosheets and tubes can spontaneously interconvert through manipulation of the assembly pH and systematic adjustment of the CMP sequence. Altogether, we establish initial guidelines for the construction of dynamically responsive 1D and 2D assemblies that undergo a structurally programmed morphological transition.


Assuntos
Colágeno/química , Nanoestruturas/química , Peptídeos/química , Sequência de Aminoácidos , Microscopia Crioeletrônica , Concentração de Íons de Hidrogênio , Microscopia de Força Atômica , Nanotubos/química , Porosidade
3.
Biochemistry ; 53(40): 6302-8, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25198136

RESUMO

Protein/peptide oligomerization, cross-ß strand fibrillation, and amyloid deposition play a critical role in many diseases, but despite extensive biophysical characterization, the structural and dynamic details of oligomerization and fibrillation of amyloidic peptides/proteins remain to be fully clarified. Here, we simultaneously monitored the atomic, molecular, and mesoscopic states of aggregating Alzheimer's amyloid ß (Aß) peptides over time, using a slow aggregation protocol and a fast aggregation protocol, and determined the cytotoxicity of the intermediate states. We show that in the early stage of fast fibrillation (the lag phase) the Aß peptides coalesced into apparently unstructured globules (15-200 nm in diameter), which slowly grew larger. Then a sharp transition occurred, characterized by the first appearance of single fibrillar structures of approximately ≥100 nm. These fibrils emerged from the globules. Simultaneously, an increase was observed for the cross-ß strand conformation that is characteristic of the fibrils that constitute mature amyloid. The number and size of single fibrils rapidly increased. Eventually, the fibrils coalesced into mature amyloid. Samples from the early lag phase of slow fibrillation conditions were especially toxic to cells, and this toxicity sharply decreased when fibrils formed and matured into amyloid. Our results suggest that the formation of fibrils may protect cells by reducing the toxic structures that appear in the early lag phase of fibrillation.


Assuntos
Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/química , Amiloide/fisiologia , Peptídeos beta-Amiloides/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Cinética , Microscopia de Força Atômica , Nanoestruturas , Fragmentos de Peptídeos/fisiologia , Agregação Patológica de Proteínas
4.
Biomacromolecules ; 15(6): 1985-91, 2014 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-24750033

RESUMO

Polyamines promote the formation of the Aß peptide amyloid fibers that are a hallmark of Alzheimer's disease. Here we show that polyamines interact with nonaggregated Aß peptides, thereby reducing the peptide's hydrophobic surface. We characterized the associated conformational change through NMR titrations and molecular dynamics simulations. We found that even low concentrations of spermine, sperimidine, and putrescine fully protected SH-SY5Y (a neuronal cell model) against the most toxic conformational species of Aß, even at an Aß oligomer concentration that would otherwise kill half of the cells or even more. These observations lead us to conclude that polyamines interfere with the more toxic prefibrillar conformations and might protect cells by promoting the structural transition of Aß toward its less toxic fibrillar state that we reported previously. Since polyamines are present in brain fluid at the concentrations where we observed all these effects, their activity needs to be taken into account in understanding the molecular processes related to the development of Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Poliaminas/metabolismo , Agregação Patológica de Proteínas/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Humanos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/toxicidade , Poliaminas/química , Agregação Patológica de Proteínas/patologia , Estrutura Secundária de Proteína
5.
ACS Chem Neurosci ; 4(3): 454-62, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23509981

RESUMO

The cellular polyamines spermine, spermidine, and their metabolic precursor putrescine, have long been associated with cell-growth, tumor-related gene regulations, and Alzheimer's disease. Here, we show by in vitro spectroscopy and AFM imaging, that these molecules promote aggregation of amyloid-beta (Aß) peptides into fibrils and modulate the aggregation pathways. NMR measurements showed that the three polyamines share a similar binding mode to monomeric Aß(1-40) peptide. Kinetic ThT studies showed that already very low polyamine concentrations promote amyloid formation: addition of 10 µM spermine (normal intracellular concentration is ~1 mM) significantly decreased the lag and transition times of the aggregation process. Spermidine and putrescine additions yielded similar but weaker effects. CD measurements demonstrated that the three polyamines induce different aggregation pathways, involving different forms of induced secondary structure. This is supported by AFM images showing that the three polyamines induce Aß(1-40) aggregates with different morphologies. The results reinforce the notion that designing suitable ligands which modulate the aggregation of Aß peptides toward minimally toxic pathways may be a possible therapeutic strategy for Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Fragmentos de Peptídeos/metabolismo , Putrescina/metabolismo , Transdução de Sinais/fisiologia , Espermidina/metabolismo , Espermina/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Humanos , Fragmentos de Peptídeos/química , Poliaminas/química , Poliaminas/metabolismo , Putrescina/química , Espermidina/química , Espermina/química
6.
Cell Motil Cytoskeleton ; 66(10): 824-38, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19396870

RESUMO

Mammalian microtubule plus-end tracking proteins (+TIPs) specifically associate with the ends of growing microtubules. +TIPs are involved in many cellular processes, including mitosis, cell migration and neurite extension. Navigators are mammalian homologues of the C. elegans unc-53 protein, an ATPase that has been linked to the migration and outgrowth of muscles, axons and excretory canals. Here we show that all three mammalian Navigators are +TIPs, consistent with a previous study on Navigator 1 (NAV1) (Martinez-Lopez et al., Mol Cell Neurosci 2005;28:599-612). Overexpression of GFP-tagged Navigators causes displacement of CAP_GLY-motif containing +TIPs, such as CLIP-170, from microtubule ends, suggesting that the Navigator-binding sites on microtubule ends overlap with those of the CAP_GLY-motif proteins. In interphase cells, mammalian Navigators also prominently localize to centrosomes, a localization that does not depend on an intact microtubule network. Fluorescence recovery after photobleaching (FRAP) experiments indicate that NAV1 associates with intracellular structures other than microtubules or centrosomes. Expression of GFP-tagged Navigators induces the formation of neurite-like extensions in non-neuronal cells, showing that Navigators can dominantly alter cytoskeletal behavior. For NAV1 this function depends on its ATPase activity; it is not achieved by a classical type of MT bundling and stabilization. Combined our data suggest that Navigators are +TIPs that can reorganize the cytoskeleton to guide cell shape changes. Our data are consistent with a role for Navigators in neurite outgrowth.


Assuntos
Citoesqueleto/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/fisiologia , Neuritos/ultraestrutura , Adenosina Trifosfatases/metabolismo , Animais , Sítios de Ligação , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/ultraestrutura , Células HeLa , Humanos , Camundongos , Proteínas de Neoplasias/fisiologia , Proteínas de Neurofilamentos/fisiologia , Estrutura Terciária de Proteína
7.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 8): 1464-6, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15272178

RESUMO

Interest in protease inhibitors has been renewed because of their potent activity in preventing carcinogenesis in a wide variety of in vivo and in vitro model systems. Potato tubers contain a wide range of such protease inhibitors. In cv. Elkana potato tubers, protease inhibitors represent about 50% of the total amount of soluble protein. Potato serine protease inhibitor (PSPI), one of the isoforms of the most abundant group of protease inhibitors, is a dimeric double-headed Kunitz-type inhibitor. No high-resolution structural information on this type of inhibitor has so far been obtained, as all currently known structures are of the monomeric single-headed or monomeric double-headed types. Crystals were grown in 0.1 M HEPES pH 7.5, 10% PEG 8000 and 8% ethylene glycol complemented with 9 mM 1-s-octyl-beta-D-thioglucoside or 0.1 M glycine. Data were collected from a single crystal under cryoconditions to 1.8 A resolution. The protein crystallized in space group P2(1), with unit-cell parameters a = 54.82, b = 93.92, c = 55.44 A, beta = 100.7 degrees; the scaling Rsym is 0.044 for 45,456 unique reflections.


Assuntos
Peptídeos/química , Proteínas de Plantas/química , Solanum tuberosum/química , Cristalização , Cristalografia por Raios X
8.
Exp Cell Res ; 289(1): 36-46, 2003 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-12941602

RESUMO

Apoptin, a protein derived from chicken anemia virus, induces apoptosis in human transformed or tumor cells but not in normal cells. When produced in bacteria as a recombinant fusion with maltose-binding protein (MBP-Apoptin), Apoptin forms a distinct, stable multimeric complex that is remarkably homogeneous and uniform. Here, using cytoplasmic microinjection, we showed that recombinant MBP-Apoptin multimers retained the characteristics of the ectopically expressed wild-type Apoptin; namely, the complexes translocated to the nucleus of tumor cells and induced apoptosis, whereas they remained in the cytoplasm of normal, primary cells and exerted no apparent toxic effect. In normal cells, MBP-Apoptin formed increasingly large, organelle-sized globular bodies with time postinjection and eventually lost the ability to be detected by immunofluorescence analysis. Costaining with an acidotrophic marker indicated that these globular structures did not correspond to lysosomes. Immunoprecipitation studies showed that MBP-Apoptin remained fully antibody-accessible regardless of buffer stringency when microinjected into tumor cells. In contrast, MBP-Apoptin in normal cells was only recoverable under stringent lysis conditions, whereas under milder conditions they became fully shielded with time on two epitopes spanning the entire protein. Further biochemical analysis showed that the long-term fate of Apoptin protein aggregates in normal cells was their eventual elimination. Our results provide the first example of a tumor-specific apoptosis-inducing aggregate that is essentially sequestered by factors or conditions present in the cytoplasm of healthy, nontransformed cells. This characteristic should reveal more about the cellular interactions of this viral protein as well as further enhance its safety as a potential tumor-specific therapeutic agent.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Proteínas do Capsídeo/uso terapêutico , Neoplasias/tratamento farmacológico , Antineoplásicos/imunologia , Apoptose/fisiologia , Proteínas do Capsídeo/imunologia , Proteínas de Transporte/farmacologia , Proteínas de Transporte/uso terapêutico , Epitopos/imunologia , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Substâncias Macromoleculares , Proteínas Ligantes de Maltose , Neoplasias/imunologia , Neoplasias/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Células Tumorais Cultivadas
9.
Eur J Biochem ; 270(17): 3619-27, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12919326

RESUMO

Recombinant, bacterially expressed apoptin protein induces apoptosis in human tumour cell lines but not in normal cells, mimicking the behaviour of ectopically expressed apoptin. Recombinant apoptin is isolated exclusively as a highly stable multimeric complex of 30-40 monomers, with little, if any, alpha-helical and beta-sheet structure. Despite its apparent disorder, multimeric apoptin is biologically active. Here, we present evidence that most of the apoptin moieties within the complex may well share a similar conformation. Furthermore, the multimer has extensive and uniform hydrophobic patches and conformationally stable domains. Only a small fraction of apoptin subunits can exchange between multimers under physiologically relevant conditions. These results prompt a model in which the apoptin multimer has a highly stable core of nonexchangeable subunits to which exchangeable subunits are attached through hydrophobic interactions. In combination with previous findings, our results lead us to propose that the stable core of apoptin is the biologically relevant structure.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Substituição de Aminoácidos , Naftalenossulfonato de Anilina/metabolismo , Sítios de Ligação , Proteínas do Capsídeo/genética , Neoplasias do Colo/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Transferência Ressonante de Energia de Fluorescência , Humanos , Interações Hidrofóbicas e Hidrofílicas , Peso Molecular , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Titulometria , Células Tumorais Cultivadas
10.
Nucleic Acids Res ; 31(16): 4805-13, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12907722

RESUMO

The chicken anaemia virus-derived protein apoptin is a tumour-specific cell-killing agent. It is biologically active as a highly stable, multimeric complex, consisting of 30-40 monomers. In tumour cells, but negligibly in normal cells, apoptin is imported into the nucleus prior to the induction of apoptosis. Immunoelectron microscopic data we report here indicate that apoptin predominantly co-localises with heterochromatin and nucleoli within tumour cells. Apoptin's preference for these DNA-dense nuclear bodies may be explained by our finding that apoptin cooperatively forms distinct superstructures with DNA in vitro. These superstructures do not grow beyond a diameter of approximately 200 nm, containing up to 20 multimeric apoptin complexes and approximately 3 kb of DNA. Furthermore, we show a single apoptin multimer to have eight independent, non-specific DNA-binding sites which preferentially bind strand ends, but which can also collaborate to bind longer stretches of DNA. Apoptin's high affinity for naked, undecorated double- and single-stranded DNA and for DNA fibre ends suggests that it may also capture such DNA in superstructures in vivo. Since these forms of DNA are predominantly found in transcriptionally active, replicating and damaged DNA, apoptin could be triggering apoptosis by interfering with DNA transcription and synthesis.


Assuntos
Proteínas do Capsídeo/metabolismo , DNA de Neoplasias/metabolismo , Nucleoproteínas/metabolismo , Sítios de Ligação , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral/ultraestrutura , Nucléolo Celular/metabolismo , DNA/metabolismo , Dimerização , Heterocromatina/metabolismo , Humanos , Cinética , Proteínas Ligantes de Maltose , Microscopia de Força Atômica , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Plasmídeos/genética , Ligação Proteica , Transfecção
11.
Nat Struct Biol ; 10(6): 468-74, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12730684

RESUMO

The Na,K-ATPase hydrolyzes ATP to drive the coupled extrusion and uptake of Na+ and K+ ions across the plasma membrane. Here, we report two high-resolution NMR structures of the 213-residue nucleotide-binding domain of rat alpha1 Na,K-ATPase, determined in the absence and the presence of ATP. The nucleotide binds in the anti conformation and shows a relative paucity of interactions with the protein, reflecting the low-affinity ATP-binding state. Binding of ATP induces substantial conformational changes in the binding pocket and in residues located in the hinge region connecting the N- and P-domains. Structural comparison with the Ca-ATPase stabilized by the inhibitor thapsigargin, E2(TG), and the model of the H-ATPase in the E1 form suggests that the observed changes may trigger the series of events necessary for the release of the K+ ions and/or disengagement of the A-domain, leading to the eventual transfer of the gamma-phosphate group to the invariant Asp369.


Assuntos
Trifosfato de Adenosina/metabolismo , Nucleotídeos/metabolismo , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/metabolismo , Complexo 2 de Proteínas Adaptadoras/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Conformação Proteica , Ratos , Homologia de Sequência de Aminoácidos , ATPase Trocadora de Sódio-Potássio/genética , Tapsigargina/química , Tapsigargina/metabolismo
12.
J Biol Chem ; 278(32): 29463-70, 2003 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12773530

RESUMO

C1-inhibitor (C1-Inh) is a serine protease inhibitor (serpin) with a unique, non-conserved N-terminal domain of unknown function. Genetic deficiency of C1-Inh causes hereditary angioedema. A novel type of mutation (Delta 3) in exon 3 of the C1-Inh gene, resulting in deletion of Asp62-Thr116 in this unique domain, was encountered in a hereditary angioedema pedigree. Because the domain is supposedly not essential for inhibitory activity, the unexpected loss-of-function of this deletion mutant was further investigated. The Delta 3 mutant and three additional mutants starting at Pro76, Gly98, and Ser115, lacking increasing parts of the N-terminal domain, were produced recombinantly. C1-Inh76 and C1-Inh98 retained normal conformation and interaction kinetics with target proteases. In contrast, C1-Inh115 and Delta 3, which both lack the connection between the serpin and the non-serpin domain via two disulfide bridges, were completely non-functional because of a complex-like and multimeric conformation, as demonstrated by several criteria. The Delta 3 mutant also circulated in multimeric form in plasma from affected family members. The C1-Inh mutant reported here is unique in that deletion of an entire amino acid stretch from a domain not shared by other serpins leads to a loss-of-function. The deletion in the unique N-terminal domain results in a "multimerization phenotype" of C1-Inh, because of diminished stability of the central beta-sheet. This phenotype, as well as the location of the disulfide bridges between the serpin and the non-serpin domain of C1-Inh, suggests that the function of the N-terminal region may be similar to one of the effects of heparin in antithrombin III, maintenance of the metastable serpin conformation.


Assuntos
Mutação , Serpinas/química , Sequência de Aminoácidos , Sequência de Bases , Western Blotting , Centrifugação com Gradiente de Concentração , Proteínas Inativadoras do Complemento 1 , Proteína Inibidora do Complemento C1 , Cristalografia por Raios X , Cisteína/química , DNA/metabolismo , Dissulfetos/química , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Éxons , Deleção de Genes , Glicina/química , Heparina/química , Temperatura Alta , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Fenótipo , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Prolina/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Serina/química , Sacarose/farmacologia , Temperatura
13.
J Biol Chem ; 278(30): 27729-36, 2003 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12754198

RESUMO

The chicken anemia virus-derived protein Apoptin induces apoptosis specifically in human tumor and transformed cells and not in normal, untransformed cells. The cell killing activity correlates with a predominantly nuclear localization of Apoptin in tumor cells, whereas in normal cells, it is detected mainly in cytoplasmic structures. To explore the role of nuclear localization for Apoptin-induced cell death in tumor cells, we employed a mutagenesis strategy. First, we demonstrated that the C terminus of Apoptin contains a bipartite-type nuclear localization signal. Strikingly, further investigation showed that Apoptin contains two different domains that induce apoptosis independently, and for both domains, we found a strong correlation between localization and killing activity. Using inhibitors, we ruled out the involvement of de novo gene transcription and translation and further showed that Apoptin itself does not have any significant transcriptional repression activity, suggesting that Apoptin exerts its effects in the nucleus by some other method. To determine whether nuclear localization is sufficient to enable Apoptin to kill normal, untransformed cells, we expressed full-length Apoptin fused to a heterologous nuclear localization signal in these cells. However, despite its nuclear localization, no apoptosis was induced, which suggests that nuclear localization per se is not sufficient for Apoptin to become active. These studies increase our understanding of the molecular pathway of Apoptin and may also shed light on the mechanism of cellular transformation.


Assuntos
Apoptose , Proteínas do Capsídeo , Capsídeo/metabolismo , Núcleo Celular/metabolismo , Transformação Celular Neoplásica , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Capsídeo/química , Linhagem Celular , Linhagem Celular Transformada , Núcleo Celular/ultraestrutura , Citoplasma/metabolismo , Fibroblastos/metabolismo , Genes Reporter , Humanos , Luciferases/metabolismo , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação , Sinais de Localização Nuclear , Plasmídeos/metabolismo , Biossíntese de Proteínas , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas
14.
J Mol Biol ; 326(3): 823-33, 2003 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-12581643

RESUMO

Antithrombin is a member of the serpin family of protease inhibitors and the major inhibitor of the blood coagulation cascade. It is unique amongst the serpins in that it circulates in a conformation that is inactive against its target proteases. Activation of antithrombin is brought about by a conformational change initiated upon binding heparin or heparan sulphate. Two isoforms exist in the circulation, alpha-antithrombin and beta-antithrombin, which differ in the amount of glycosylation present on the polypeptide chain; beta-antithrombin lacks the carbohydrate present at Asn135 in alpha-antithrombin. Of the two forms, beta-antithrombin has the higher affinity for heparin and thus functions as the major inhibitor in vivo even though it is the less abundant form. The reason for the differences in heparin affinity between the alpha and beta-forms have been shown to be due to the additional carbohydrate changing the rate of the conformational change. Here, we describe the most accurate structures of alpha-antithrombin and alpha-antithrombin+heparin pentasaccharide reported to date (2.6A and 2.9A resolution, respectively, both re-refinements using old data), and the structure of beta-antithrombin (2.6A resolution). The new structures have a remarkable degree of ordered carbohydrate and include parts of the antithrombin chain not modeled before. The structures have allowed a detailed comparison of the conformational differences between the three. They show that the structural basis of the lower affinity for heparin of alpha-antithrombin over beta-antithrombin is due to the conformational change that occurs upon heparin binding being sterically hindered by the presence of the additional bulky carbohydrate at Asn135.


Assuntos
Antitrombinas/química , Antitrombinas/metabolismo , Heparina/metabolismo , Cristalografia por Raios X , Glicosilação , Humanos , Modelos Moleculares , Conformação Proteica
15.
J Biol Chem ; 278(11): 9042-51, 2003 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-12496278

RESUMO

The chicken anemia virus-derived Apoptin protein induces tumor-specific apoptosis. Here, we show that recombinant Apoptin protein spontaneously forms non-covalent globular aggregates comprising 30 to 40 subunits in vitro. This multimerization is robust and virtually irreversible, and the globular aggregates are also stable in cell extracts, suggesting that they remain intact within the cell. Furthermore, studies of Apoptin expressed in living cells confirm that Apoptin indeed exists in large complexes in vivo. We map the structural motifs responsible for multimerization in vitro and aggregation in vivo to the N-terminal half of the protein. Moreover, we show that covalently fixing the Apoptin monomers within the recombinant protein multimer by internal cross-linking does not affect the biological activity of Apoptin, as these fixed aggregates exhibit similar tumor-specific localization and apoptosis-inducing properties as non-cross-linked Apoptin. Taken together, our results imply that recombinant Apoptin protein is a multimer when inducing apoptosis, and we propose that this multimeric state is an essential feature of its ability to do so. Finally, we determine that Apoptin adopts little, if any, regular secondary structure within the aggregates. This surprising result would classify Apoptin as the first protein for which, rather than the formation of a well defined tertiary and quaternary structure, semi-random aggregation is sufficient for activity.


Assuntos
Apoptose , Proteínas do Capsídeo , Capsídeo/fisiologia , Motivos de Aminoácidos , Western Blotting , Calibragem , Capsídeo/metabolismo , Proteínas de Transporte/metabolismo , Vírus da Anemia da Galinha/metabolismo , Cromatografia , Dicroísmo Circular , Clonagem Molecular , Reagentes de Ligações Cruzadas/farmacologia , Detergentes/farmacologia , Dimerização , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Vetores Genéticos , Luz , Proteínas Ligantes de Maltose , Microscopia de Força Atômica , Microscopia Eletrônica , Microscopia de Fluorescência , Fases de Leitura Aberta , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espalhamento de Radiação , Espectrometria de Fluorescência , Transfecção , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA