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











Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 4(1): 849, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34239038

RESUMO

Huntington disease (HD) is a neurodegenerative trinucleotide repeat disorder caused by an expanded poly-glutamine (polyQ) tract in the mutant huntingtin (mHTT) protein. The formation and topology of filamentous mHTT inclusions in the brain (hallmarks of HD implicated in neurotoxicity) remain elusive. Using cryo-electron tomography and subtomogram averaging, here we show that mHTT exon 1 and polyQ-only aggregates in vitro are structurally heterogenous and filamentous, similar to prior observations with other methods. Yet, we find filaments in both types of aggregates under ~2 nm in width, thinner than previously reported, and regions forming large sheets. In addition, our data show a prevalent subpopulation of filaments exhibiting a lumpy slab morphology in both aggregates, supportive of the polyQ core model. This provides a basis for future cryoET studies of various aggregated mHTT and polyQ constructs to improve their structure-based modeling as well as their identification in cells without fusion tags.


Assuntos
Tomografia com Microscopia Eletrônica/métodos , Éxons/genética , Proteína Huntingtina/genética , Mutação , Peptídeos/genética , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/ultraestrutura , Doença de Huntington/genética , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/ultraestrutura , Agregados Proteicos , Agregação Patológica de Proteínas , Conformação Proteica
2.
J Biol Chem ; 296: 100333, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33508321

RESUMO

ß2-Microglobulin (ß2m) is the causative protein of dialysis-related amyloidosis. Its unfolding mainly proceeds along the pathway of NC →UC ⇄ UT, whereas refolding follows the UT → IT (→NT) →NC pathway, in which N, I, and U are the native, intermediate, and unfolded states, respectively, with the Pro32 peptidyl-prolyl bond in cis or trans conformation as indicated by the subscript. It is noted that the IT state is a putative amyloidogenic precursor state. Several aggregation-prone variants of ß2m have been reported to date. One of these variants is D76N ß2m, which is a naturally occurring amyloidogenic mutant. To elucidate the molecular mechanisms contributing to the enhanced amyloidogenicity of the mutant, we investigated the equilibrium and kinetic transitions of pressure-induced folding/unfolding equilibria in the wild type and D76N mutant by monitoring intrinsic tryptophan and 1-anilino-8-naphthalene sulfonate fluorescence. An analysis of kinetic data revealed that the different folding/unfolding behaviors of the wild type and D76N mutant were due to differences in the activation energy between the unfolded and the intermediate states as well as stability of the native state, leading to more rapid accumulation of IT state for D76N in the refolding process. In addition, the IT state was found to assume more hydrophobic nature. These changes induced the enhanced amyloidogenicity of the D76N mutant and the distinct pathogenic symptoms of patients. Our results suggest that the stabilization of the native state will be an effective approach for suppressing amyloid fibril formation of this mutant.


Assuntos
Amiloidose/genética , Proteínas Mutantes/química , Agregados Proteicos/genética , Microglobulina beta-2/química , Amiloidose/metabolismo , Amiloidose/patologia , Naftalenossulfonato de Anilina/química , Humanos , Cinética , Proteínas Mutantes/genética , Proteínas Mutantes/ultraestrutura , Pressão , Dobramento de Proteína , Diálise Renal/efeitos adversos , Transdução de Sinais/genética , Triptofano/química , Microglobulina beta-2/genética , Microglobulina beta-2/ultraestrutura
3.
PLoS One ; 14(1): e0210963, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30645640

RESUMO

The formation of hemostatic plugs at sites of vascular injury crucially involves the multimeric glycoprotein von Willebrand factor (VWF). VWF multimers are linear chains of N-terminally linked dimers. The latter are formed from monomers via formation of the C-terminal disulfide bonds Cys2771-Cys2773', Cys2773-Cys2771', and Cys2811-Cys2811'. Mutations in VWF that impair multimerization can lead to subtype 2A of the bleeding disorder von Willebrand Disease (VWD). Commonly, the multimer size distribution of VWF is assessed by electrophoretic multimer analysis. Here, we present atomic force microscopy (AFM) imaging as a method to determine the size distribution of VWF variants by direct visualization at the single-molecule level. We first validated our approach by investigating recombinant wildtype VWF and a previously studied mutant (p.Cys1099Tyr) that impairs N-terminal multimerization. We obtained excellent quantitative agreement with results from earlier studies and with electrophoretic multimer analysis. We then imaged specific mutants that are known to exhibit disturbed C-terminal dimerization. For the mutants p.Cys2771Arg and p.Cys2773Arg, we found the majority of monomers (87 ± 5% and 73 ± 4%, respectively) not to be C-terminally dimerized. While these results confirm that Cys2771 and Cys2773 are crucial for dimerization, they additionally provide quantitative information on the mutants' different abilities to form alternative C-terminal disulfides for residual dimerization. We further mutated Cys2811 to Ala and found that only 23 ± 3% of monomers are not C-terminally dimerized, indicating that Cys2811 is structurally less important for dimerization. Furthermore, for mutants p.Cys2771Arg, p.Cys2773Arg, and p.Cys2811Ala we found 'even-numbered' non-native multimers, i.e. multimers with monomers attached on both termini; a multimer species that cannot be distinguished from native multimers by conventional multimer analysis. Summarizing, we demonstrate that AFM imaging can provide unique insights into VWF processing defects at the single-molecule level that cannot be gained from established methods of multimer analysis.


Assuntos
Microscopia de Força Atômica/métodos , Imagem Individual de Molécula/métodos , Fator de von Willebrand/química , Fator de von Willebrand/ultraestrutura , Substituição de Aminoácidos , Cisteína/química , Dimerização , Células HEK293 , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/ultraestrutura , Mutação de Sentido Incorreto , Tamanho da Partícula , Multimerização Proteica/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/ultraestrutura , Doenças de von Willebrand/sangue , Doenças de von Willebrand/genética , Fator de von Willebrand/genética
4.
Nature ; 563(7731): 426-430, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30405239

RESUMO

ABCG2 is a transporter protein of the ATP-binding-cassette (ABC) family that is expressed in the plasma membrane in cells of various tissues and tissue barriers, including the blood-brain, blood-testis and maternal-fetal barriers1-4. Powered by ATP, it translocates endogenous substrates, affects the pharmacokinetics of many drugs and protects against a wide array of xenobiotics, including anti-cancer drugs5-12. Previous studies have revealed the architecture of ABCG2 and the structural basis of its inhibition by small molecules and antibodies13,14. However, the mechanisms of substrate recognition and ATP-driven transport are unknown. Here we present high-resolution cryo-electron microscopy (cryo-EM) structures of human ABCG2 in a substrate-bound pre-translocation state and an ATP-bound post-translocation state. For both structures, we used a mutant containing a glutamine replacing the catalytic glutamate (ABCG2EQ), which resulted in reduced ATPase and transport rates and facilitated conformational trapping for structural studies. In the substrate-bound state, a single molecule of estrone-3-sulfate (E1S) is bound in a central, hydrophobic and cytoplasm-facing cavity about halfway across the membrane. Only one molecule of E1S can bind in the observed binding mode. In the ATP-bound state, the substrate-binding cavity has collapsed while an external cavity has opened to the extracellular side of the membrane. The ATP-induced conformational changes include rigid-body shifts of the transmembrane domains, pivoting of the nucleotide-binding domains (NBDs), and a change in the relative orientation of the NBD subdomains. Mutagenesis and in vitro characterization of transport and ATPase activities demonstrate the roles of specific residues in substrate recognition, including a leucine residue that forms a 'plug' between the two cavities. Our results show how ABCG2 harnesses the energy of ATP binding to extrude E1S and other substrates, and suggest that the size and binding affinity of compounds are important for distinguishing substrates from inhibitors.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/ultraestrutura , Trifosfato de Adenosina/metabolismo , Microscopia Crioeletrônica , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/ultraestrutura , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/química , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Sítios de Ligação , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutação , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Conformação Proteica , Especificidade por Substrato
5.
Nature ; 557(7703): 62-67, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29695864

RESUMO

Gasdermins mediate inflammatory cell death after cleavage by caspases or other, unknown enzymes. The cleaved N-terminal fragments bind to acidic membrane lipids to form pores, but the mechanism of pore formation remains unresolved. Here we present the cryo-electron microscopy structures of the 27-fold and 28-fold single-ring pores formed by the N-terminal fragment of mouse GSDMA3 (GSDMA3-NT) at 3.8 and 4.2 Å resolutions, and of a double-ring pore at 4.6 Å resolution. In the 27-fold pore, a 108-stranded anti-parallel ß-barrel is formed by two ß-hairpins from each subunit capped by a globular domain. We identify a positively charged helix that interacts with the acidic lipid cardiolipin. GSDMA3-NT undergoes radical conformational changes upon membrane insertion to form long, membrane-spanning ß-strands. We also observe an unexpected additional symmetric ring of GSDMA3-NT subunits that does not insert into the membrane in the double-ring pore, which may represent a pre-pore state of GSDMA3-NT. These structures provide a basis that explains the activities of several mutant gasdermins, including defective mutants that are associated with cancer.


Assuntos
Microscopia Crioeletrônica , Proteínas/química , Proteínas/ultraestrutura , Animais , Membrana Celular/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipídeos de Membrana/metabolismo , Camundongos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/ultraestrutura , Mutação , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestrutura , Neoplasias/genética , Perforina/química , Perforina/metabolismo , Proteínas de Ligação a Fosfato , Conformação Proteica , Dobramento de Proteína , Multimerização Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Relação Estrutura-Atividade
6.
Nature ; 553(7689): 521-525, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29342139

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) causes Kaposi's sarcoma, a cancer that commonly affects patients with AIDS and which is endemic in sub-Saharan Africa. The KSHV capsid is highly pressurized by its double-stranded DNA genome, as are the capsids of the eight other human herpesviruses. Capsid assembly and genome packaging of herpesviruses are prone to interruption and can therefore be targeted for the structure-guided development of antiviral agents. However, herpesvirus capsids-comprising nearly 3,000 proteins and over 1,300 Å in diameter-present a formidable challenge to atomic structure determination and functional mapping of molecular interactions. Here we report a 4.2 Å resolution structure of the KSHV capsid, determined by electron-counting cryo-electron microscopy, and its atomic model, which contains 46 unique conformers of the major capsid protein (MCP), the smallest capsid protein (SCP) and the triplex proteins Tri1 and Tri2. Our structure and mutagenesis results reveal a groove in the upper domain of the MCP that contains hydrophobic residues that interact with the SCP, which in turn crosslinks with neighbouring MCPs in the same hexon to stabilize the capsid. Multiple levels of MCP-MCP interaction-including six sets of stacked hairpins lining the hexon channel, disulfide bonds across channel and buttress domains in neighbouring MCPs, and an interaction network forged by the N-lasso domain and secured by the dimerization domain-define a robust capsid that is resistant to the pressure exerted by the enclosed genome. The triplexes, each composed of two Tri2 molecules and a Tri1 molecule, anchor to the capsid floor via a Tri1 N-anchor to plug holes in the MCP network and rivet the capsid floor. These essential roles of the MCP N-lasso and Tri1 N-anchor are verified by serial-truncation mutageneses. Our proof-of-concept demonstration of the use of polypeptides that mimic the smallest capsid protein to inhibit KSHV lytic replication highlights the potential for exploiting the interaction hotspots revealed in our atomic structure to develop antiviral agents.


Assuntos
Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica , Herpesvirus Humano 8/crescimento & desenvolvimento , Herpesvirus Humano 8/ultraestrutura , Mutagênese , Replicação Viral , Capsídeo/química , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/ultraestrutura , Dissulfetos/metabolismo , Desenho de Fármacos , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Mutação , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , Estabilidade Proteica , Replicação Viral/genética
7.
Nat Commun ; 8(1): 781, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974687

RESUMO

Nipah virus is an emergent paramyxovirus that causes deadly encephalitis and respiratory infections in humans. Two glycoproteins coordinate the infection of host cells, an attachment protein (G), which binds to cell surface receptors, and a fusion (F) protein, which carries out the process of virus-cell membrane fusion. The G protein binds to ephrin B2/3 receptors, inducing G conformational changes that trigger F protein refolding. Using an optical approach based on second harmonic generation, we show that monomeric and dimeric receptors activate distinct conformational changes in G. The monomeric receptor-induced changes are not detected by conformation-sensitive monoclonal antibodies or through electron microscopy analysis of G:ephrinB2 complexes. However, hydrogen/deuterium exchange experiments confirm the second harmonic generation observations and reveal allosteric changes in the G receptor binding and F-activating stalk domains, providing insights into the pathway of receptor-activated virus entry.Nipah virus causes encephalitis in humans. Here the authors use a multidisciplinary approach to study the binding of the viral attachment protein G to its host receptor ephrinB2 and show that monomeric and dimeric receptors activate distinct conformational changes in G and discuss implications for receptor-activated virus entry.


Assuntos
Efrina-B2/metabolismo , Vírus Nipah/metabolismo , Proteínas do Envelope Viral/metabolismo , Regulação Alostérica , Anticorpos Monoclonais/metabolismo , Medição da Troca de Deutério , Células HEK293 , Humanos , Espectrometria de Massas , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Coloração Negativa , Ligação Proteica , Multimerização Proteica
8.
J Am Chem Soc ; 139(38): 13466-13475, 2017 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-28863262

RESUMO

Self-assembly of plasma membrane-associated Ras GTPases has major implications to the regulation of cell signaling. However, the structural basis of homo-oligomerization and the fractional distribution of oligomeric states remained undetermined. We have addressed these issues by deciphering the distribution of dimers and higher-order oligomers of K-Ras4B, the most frequently mutated Ras isoform in human cancers. We focused on the constitutively active G12V K-Ras and two of its variants, K101E and K101C/E107C, which respectively destabilize and stabilize oligomers. Using raster image correlation spectroscopy and number and brightness analysis combined with fluorescence recovery after photobleaching, fluorescence correlation spectroscopy and electron microscopy in live cells, we show that G12V K-Ras exists as a mixture of monomers, dimers and larger oligomers, while the K101E mutant is predominantly monomeric and K101C/E107C is dominated by oligomers. This observation demonstrates the ability of K-Ras to exist in multiple oligomeric states whose population can be altered by interfacial mutations. Using molecular modeling and simulations we further show that K-Ras uses two partially overlapping interfaces to form compositionally and topologically diverse oligomers. Our results thus provide the first detailed insight into the multiplicity, structure, and membrane organization of K-Ras homomers.


Assuntos
Membrana Celular/metabolismo , Multimerização Proteica , Proteínas ras/química , Proteínas ras/metabolismo , Animais , Hominidae , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Proteínas ras/genética , Proteínas ras/ultraestrutura
9.
Sci Rep ; 6: 33631, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27632940

RESUMO

Lon is an essential, multitasking AAA(+) protease regulating many cellular processes in species across all kingdoms of life. Altered expression levels of the human mitochondrial Lon protease (hLon) are linked to serious diseases including myopathies, paraplegia, and cancer. Here, we present the first 3D structure of full-length hLon using cryo-electron microscopy. hLon has a unique three-dimensional structure, in which the proteolytic and ATP-binding domains (AP-domain) form a hexameric chamber, while the N-terminal domain is arranged as a trimer of dimers. These two domains are linked by a narrow trimeric channel composed likely of coiled-coil helices. In the presence of AMP-PNP, the AP-domain has a closed-ring conformation and its N-terminal entry gate appears closed, but in ADP binding, it switches to a lock-washer conformation and its N-terminal gate opens, which is accompanied by a rearrangement of the N-terminal domain. We have also found that both the enzymatic activities and the 3D structure of a hLon mutant lacking the first 156 amino acids are severely disturbed, showing that hLon's N-terminal domains are crucial for the overall structure of the hLon, maintaining a conformation allowing its proper functioning.


Assuntos
Mitocôndrias/enzimologia , Protease La/química , Trifosfato de Adenosina/metabolismo , Adenilil Imidodifosfato/metabolismo , Bacillus subtilis/enzimologia , Humanos , Processamento de Imagem Assistida por Computador , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Protease La/ultraestrutura , Domínios Proteicos , Proteólise
10.
J Virol ; 90(19): 8542-51, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27440903

RESUMO

UNLABELLED: The adeno-associated viruses (AAV) are promising therapeutic gene delivery vectors and better understanding of their capsid assembly and genome packaging mechanism is needed for improved vector production. Empty AAV capsids assemble in the nucleus prior to genome packaging by virally encoded Rep proteins. To elucidate the capsid determinants of this process, structural differences between wild-type (wt) AAV2 and a packaging deficient variant, AAV2-R432A, were examined using cryo-electron microscopy and three-dimensional image reconstruction both at an ∼5.0-Å resolution (medium) and also at 3.8- and 3.7-Å resolutions (high), respectively. The high resolution structures showed that removal of the arginine side chain in AAV2-R432A eliminated hydrogen bonding interactions, resulting in altered intramolecular and intermolecular interactions propagated from under the 3-fold axis toward the 5-fold channel. Consistent with these observations, differential scanning calorimetry showed an ∼10°C decrease in thermal stability for AAV2-R432A compared to wt-AAV2. In addition, the medium resolution structures revealed differences in the juxtaposition of the less ordered, N-terminal region of their capsid proteins, VP1/2/3. A structural rearrangement in AAV2-R432A repositioned the ßA strand region under the icosahedral 2-fold axis rather than antiparallel to the ßB strand, eliminating many intramolecular interactions. Thus, a single amino acid substitution can significantly alter the AAV capsid integrity to the extent of reducing its stability and possibly rendering it unable to tolerate the stress of genome packaging. Furthermore, the data show that the 2-, 3-, and 5-fold regions of the capsid contributed to producing the packaging defect and highlight a tight connection between the entire capsid in maintaining packaging efficiency. IMPORTANCE: The mechanism of AAV genome packaging is still poorly understood, particularly with respect to the capsid determinants of the required capsid-Rep interaction. Understanding this mechanism may aid in the improvement of AAV packaging efficiency, which is currently ∼1:10 (10%) genome packaged to empty capsid in vector preparations. This report identifies regions of the AAV capsid that play roles in genome packaging and that may be important for Rep recognition. It also demonstrates the need to maintain capsid stability for the success of this process. This information is important for efforts to improve AAV genome packaging and will also inform the engineering of AAV capsid variants for improved tropism, specific tissue targeting, and host antibody escape by defining amino acids that cannot be altered without detriment to infectious vector production.


Assuntos
Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/ultraestrutura , Dependovirus/fisiologia , Dependovirus/ultraestrutura , Montagem de Vírus , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Imageamento Tridimensional , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Ligação Proteica , Mapeamento de Interação de Proteínas , Vírion/química , Vírion/efeitos da radiação
11.
J Biol Chem ; 290(28): 17451-61, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25995452

RESUMO

Huntington disease, a neurodegenerative disorder characterized by functional deficits and loss of striatal neurons, is linked to an expanded and unstable CAG trinucleotide repeat in the huntingtin gene (HTT). This DNA sequence translates to a polyglutamine repeat in the protein product, leading to mutant huntingtin (mHTT) protein aggregation. The aggregation of mHTT is inhibited in vitro and in vivo by the TCP-1 ring complex (TRiC) chaperonin. Recently, a novel complex comprised of a single type of TRiC subunit has been reported to inhibit mHTT aggregation. Specifically, the purified CCT5 homo-oligomer complex, when compared with TRiC, has a similar structure, ATP use, and substrate refolding activity, and, importantly, it also inhibits mHTT aggregation. Using an aggregation suppression assay and cryoelectron tomography coupled with a novel computational classification method, we uncover the interactions between the synthetic CCT5 complex (∼ 1 MDa) and aggregates of mutant huntingtin exon 1 containing 46 glutamines (mHTTQ46-Ex1). We find that, in a similar fashion to TRiC, synthetic CCT5 complex caps mHTT fibrils at their tips and encapsulates mHTT oligomers, providing a structural description of the inhibition of mHTTQ46-Ex1 by CCT5 complex and a shared mechanism of mHTT inhibition between TRiC chaperonin and the CCT5 complex: cap and contain.


Assuntos
Chaperonina com TCP-1/química , Proteínas Mutantes/química , Proteínas do Tecido Nervoso/química , Chaperonina com TCP-1/genética , Chaperonina com TCP-1/ultraestrutura , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/ultraestrutura , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/ultraestrutura , Agregados Proteicos , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/ultraestrutura
12.
Biochemistry ; 54(3): 666-76, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25531836

RESUMO

The process of amyloid formation by the normally soluble hormone islet amyloid polypeptide (IAPP) contributes to ß-cell death in type 2 diabetes and in islet transplants. There are no clinically approved inhibitors of islet amyloidosis, and the mode of action of existing inhibitors is not well-understood. Resveratrol, a natural polyphenol, has been reported to inhibit amyloid formation by IAPP and by the Alzheimer's disease Aß peptide. The mechanism of action of this compound is not known, nor is its mode of interaction with IAPP. In this study, we use a series of IAPP variants to examine possible interactions between resveratrol and IAPP. Fluorescence assays, transmission electron microscopy, and mass spectrometry demonstrate that resveratrol is much less effective as an inhibitor of IAPP amyloid formation than the polyphenol (-)-epigallocatechin 3-gallate (EGCG) and, unlike EGCG, does not significantly disaggregate preformed IAPP amyloid fibrils. Resveratrol is also shown to interfere with thioflavin-T assays. His-18 mutants, a truncation mutant, mutants of each of the aromatic residues, and mutants of Arg-11 of IAPP were examined. Mutation of His to Gln or Leu weakens the ability of resveratrol to inhibit amyloid formation by IAPP, as do mutations of Arg-11, Phe-15, or Tyr-37 to Leu, and truncation to form the variant Ac 8-37-IAPP, which removes the first seven residues to eliminate Lys-1 and the N-terminal amino group. In contrast, replacement of Phe-23 with Leu has a smaller effect. The data highlight Phe-15, His-18, and Tyr-37 as being important for IAPP-resveratrol interactions and are consistent with a potential role of the N-terminus and Arg-11 in polypeptide-resveratrol interactions.


Assuntos
Aminoácidos Aromáticos/metabolismo , Amiloide/metabolismo , Histidina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Estilbenos/farmacologia , Sequência de Aminoácidos , Amiloide/ultraestrutura , Arginina/metabolismo , Benzotiazóis , Catequina/análogos & derivados , Catequina/farmacologia , Análise Mutacional de DNA , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/ultraestrutura , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Resveratrol , Espectrometria de Massas por Ionização por Electrospray , Relação Estrutura-Atividade , Tiazóis/metabolismo
13.
Biochim Biophys Acta ; 1843(9): 2089-99, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24909779

RESUMO

EPM1 is a rare progressive myoclonus epilepsy accompanied by apoptosis in the cerebellum of patients. Mutations in the gene of stefin B (cystatin B) are responsible for the primary defect underlying EPM1. Taking stefin B aggregates as a model we asked what comes first, protein aggregation or oxidative stress, and how these two processes correlate with cell death. We studied the aggregation in cells of the stefin B wild type, G4R mutant, and R68X fragment before (Ceru et al., 2010, Biol. Cell). The present study was performed on two more missense mutants of human stefin B, G50E and Q71P, and they similarly showed numerous aggregates upon overexpression. Mutant- and oligomer-dependent increase in oxidative stress and cell death in cells bearing aggregates was shown. On the other hand, there was no correlation between the size and number of the aggregates and cell death. We suggest that differences in toxicity of the aggregates depend on whether they are in oligomeric/protofibrillar or fibrillar form. This in turn likely depends on the mutant's 3D structure where unfolded proteins show lower toxicity. Imaging by transmission electron microscopy showed that the aggregates in cells are of different types: bigger perinuclear, surrounded by membranes and sometimes showing vesicle-like invaginations, or smaller, punctual and dispersed throughout the cytoplasm. All EPM1 mutants studied were inactive as cysteine proteases inhibitors and in this way contribute to loss of stefin B functions. Relevance to EPM1 disease by gain in toxic function is discussed.


Assuntos
Cistatina B/química , Cistatina B/genética , Proteínas Mutantes/química , Estresse Oxidativo , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/patologia , Amiloide/metabolismo , Animais , Anexina A5/metabolismo , Benzotiazóis , Células CHO , Contagem de Células , Morte Celular , Sobrevivência Celular , Cricetinae , Cricetulus , Cistatina B/ultraestrutura , Células HEK293 , Humanos , Cinética , Proteínas Mutantes/ultraestrutura , Propídio/metabolismo , Estrutura Quaternária de Proteína , Espectrometria de Fluorescência , Tiazóis/metabolismo , Transfecção
14.
FEBS J ; 279(2): 234-50, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22074440

RESUMO

Replicative helicases are major motor proteins essential for chromosomal DNA replication in prokaryotes. Usually hexameric in solution, their DNA binding property must have different roles at stages ranging from the loading onto a branched structure at initiation from the origin to the highly processive translocation during elongation. Here, we have analysed the DNA binding activity of Helicobacter pylori (Hp) replicative helicase, DnaB. The results indicate that while the C-terminal region is important for its DNA binding activity, the N-terminus appears to dampen the protein's affinity for DNA. The masking activity of the N-terminus does not require ATP or hexamerization of HpDnaB and can be overcome by deleting the N-terminus. It can also be neutralized by engaging the N-terminus in an interaction with a partner like the C-terminus of DnaG primase. The inhibitory effect of the N-terminus on DNA binding activity is consistent with the 3D homology model of HpDnaB. Electron microscopy of the HpDnaB-ssDNA complex showed that HpDnaB preferentially bound at the ends of linear ssDNA and translocated along the DNA in the presence of ATP. These results provide an insight into the stimulatory and inhibitory effects of different regions of HpDnaB on DNA binding activities that may be central to the loading and translocation functions of DnaB helicases.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , DnaB Helicases/química , DnaB Helicases/metabolismo , Helicobacter pylori/enzimologia , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/ultraestrutura , Sítios de Ligação , Transporte Biológico , Dicroísmo Circular , DNA Primase/química , DNA Primase/metabolismo , Replicação do DNA , DNA de Cadeia Simples/química , DNA de Cadeia Simples/metabolismo , DNA de Cadeia Simples/ultraestrutura , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/ultraestrutura , DnaB Helicases/genética , DnaB Helicases/ultraestrutura , Cinética , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Conformação Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Homologia de Sequência de Aminoácidos
15.
J Bone Miner Res ; 26(8): 1926-38, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21472776

RESUMO

Familial expansile osteolysis and related disorders are caused by heterozygous tandem duplication mutations in the signal peptide region of the gene encoding receptor activator of NF-κB (RANK), a receptor critical for osteoclast formation and function. Previous studies have shown that overexpression of these mutant proteins causes constitutive activation of NF-κB signaling in vitro, and it has been assumed that this accounts for the focal osteolytic lesions that are seen in vivo. We show here that constitutive activation of NF-κB occurred in HEK293 cells overexpressing wild-type or mutant RANK but not in stably transfected cell lines expressing low levels of each RANK gene. Importantly, only cells expressing wild-type RANK demonstrated ligand-dependent activation of NF-κB. When overexpressed, mutant RANK did not localize to the plasma membrane but localized to extensive areas of organized smooth endoplasmic reticulum, whereas, as expected, wild-type RANK was detected at the plasma membrane and in the Golgi apparatus. This intracellular accumulation of the mutant proteins is probably the result of lack of signal peptide cleavage because, using two in vitro translation systems, we demonstrate that the mutations in RANK prevent cleavage of the signal peptide. In conclusion, signal peptide mutations lead to accumulation of RANK in the endoplasmic reticulum and prevent direct activation by RANK ligand. These results strongly suggest that the increased osteoclast formation/activity caused by these mutations cannot be explained by studying the homozygous phenotype alone but requires further detailed investigation of the heterozygous expression of the mutant RANK proteins.


Assuntos
Mutação/genética , NF-kappa B/metabolismo , Sinais Direcionadores de Proteínas/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Sequência de Bases , Linhagem Celular , DNA Nucleotidiltransferases/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Dados de Sequência Molecular , Peso Molecular , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Osteoclastos/metabolismo , Osteoclastos/ultraestrutura , Transporte Proteico , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/ultraestrutura , Reprodutibilidade dos Testes , Frações Subcelulares/metabolismo , Transfecção
16.
Virology ; 410(2): 345-52, 2011 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-21185050

RESUMO

The protruding (P) domain of norovirus VP1 is responsible for immune recognition and host receptor interaction. Our previous studies have demonstrated that a modification of the ends of the P domain affects the conformation and/or function of the P protein. An expression of the P domain with or without the hinge, or with an additional cysteine at either ends of the P protein resulted in P dimers and/or P particles. Here we report a new type of subviral particle, the small P particles, through a further modification, either an addition of the flag tag or a change of the arginine cluster, at the C-terminus of the cysteine-containing P domain. Gel filtration and cryo-EM studies showed that the small P particles are tetrahedrons formed by 6 P dimers or 12 P monomers that is half-size of the P particles. Fitting of the crystal structure of the P domain into the cryo-EM density map of the particle indicated similar conformations of the P dimers as those in P particles. The small P particles bind human HBGAs and are antigenically reactive similar to their parental VLPs and P particles. These data suggest that the C-terminus of the P domain is an important factor in the formation of the P particles. Further elucidation of the mechanism of these modifications in the P particle formation would be important in structure biology and morphogenesis of noroviruses. The small P particles may also be a useful alternative in study of norovirus-host interaction and vaccine development for noroviruses.


Assuntos
Norovirus/genética , Multimerização Proteica , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Cromatografia em Gel , Microscopia Crioeletrônica , Interações Hospedeiro-Patógeno , Humanos , Substâncias Macromoleculares/ultraestrutura , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Norovirus/ultraestrutura , Ligação Proteica , Proteínas Estruturais Virais/ultraestrutura , Vacinas Virais/imunologia , Virossomos/ultraestrutura , Ligação Viral
17.
Cell Motil Cytoskeleton ; 66(11): 986-99, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19530175

RESUMO

Short polypeptides from intermediate filament (IF) proteins containing one of the two IF-consensus motifs interfere severely with filament assembly in vitro. We now have systematically investigated a series of larger fragments of the muscle-specific IF protein desmin representing entire functional domains such as coil1 or coil 2. "Half molecules" comprising the amino-terminal portion of desmin, such as DesDeltaC240 and the "tagged" derivative Des(ESA)DeltaC244, assembled into large, roundish aggregates already at low ionic strength, DesDeltaC250 formed extended, relatively uniform filaments, whereas DesDeltaC265 and DesDeltaC300 were soluble under these conditions. Surprisingly, all mutant desmin fragments assembled very rapidly into long thick filaments or spacious aggregates when the ionic strength was raised to standard assembly conditions. In contrast, when these desmin mutants were assembled in the presence of wild-type (WT) desmin, their assembly properties were completely changed: The elongation of the two shorter desmin fragments was completely inhibited by WT desmin, whereas DesDeltaC250, DesDeltaC265 and DesDeltaC300 coassembled with desmin into filaments, but these mixed filaments were distinctly disturbed and exhibited a very different phenotype for each mutant. After transfection into fibroblasts and cardiomyocytes, the truncated mutant Des (ESA)DeltaC244 localized largely to the cytoplasm, as revealed by a tag-specific monoclonal antibody, and also partially colocalized there with the collapsed endogenous vimentin and desmin systems indicating its interference with IF-organizing processes. In contrast, in cells without an authentic cytoplasmic IF system such as line SW13, Des(ESA)DeltaC242 entered the nucleus and was deposited in small dot-like structures in chromatin-free spaces without any noticeable effect on nuclear morphology.


Assuntos
Desmina/química , Desmina/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Domínios e Motivos de Interação entre Proteínas/fisiologia , Células 3T3 , Carcinoma Adrenocortical/metabolismo , Carcinoma Adrenocortical/ultraestrutura , Animais , Linhagem Celular Transformada , Desmina/ultraestrutura , Átrios do Coração/metabolismo , Átrios do Coração/ultraestrutura , Humanos , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Multimerização Proteica , Relação Estrutura-Atividade , Vimentina/metabolismo , Vimentina/ultraestrutura
18.
Nature ; 457(7230): 694-8, 2009 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19194444

RESUMO

For a retrovirus such as HIV to be infectious, a properly formed capsid is needed; however, unusually among viruses, retrovirus capsids are highly variable in structure. According to the fullerene conjecture, they are composed of hexamers and pentamers of capsid protein (CA), with the shape of a capsid varying according to how the twelve pentamers are distributed and its size depending on the number of hexamers. Hexamers have been studied in planar and tubular arrays, but the predicted pentamers have not been observed. Here we report cryo-electron microscopic analyses of two in-vitro-assembled capsids of Rous sarcoma virus. Both are icosahedrally symmetric: one is composed of 12 pentamers, and the other of 12 pentamers and 20 hexamers. Fitting of atomic models of the two CA domains into the reconstructions shows three distinct inter-subunit interactions. These observations substantiate the fullerene conjecture, show how pentamers are accommodated at vertices, support the inference that nucleation is a crucial morphologic determinant, and imply that electrostatic interactions govern the differential assembly of pentamers and hexamers.


Assuntos
Capsídeo/metabolismo , Capsídeo/ultraestrutura , Vírus do Sarcoma de Rous/química , Vírus do Sarcoma de Rous/ultraestrutura , Montagem de Vírus , Capsídeo/química , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , HIV/química , HIV/genética , HIV/ultraestrutura , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Mutação , Polimorfismo Genético , Multimerização Proteica , Estrutura Quaternária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Vírus do Sarcoma de Rous/genética , Eletricidade Estática
19.
J Mol Biol ; 385(4): 1257-65, 2009 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-19041877

RESUMO

Amyloid plaques, formed from amyloid beta (Abeta) peptides (mainly Abeta40 or Abeta42), are one of the most important pathological characteristics of Alzheimer's disease. Here, a single D-form proline substitution in the 40-amino-acid Abeta40 peptide can totally change the aggregation behavior of this peptide. The residue immediately preceding each glycine in Abeta40 (S8, V24, I32, and V36) was individually replaced by D-form proline ((D)Pro). The resulting (D)P-G sequence (the (D)Pro residue and the following Gly residue) was designed as a "structural clip" to force the formation of a bend in the peptide, as this sequence has been reported to be a strong promoter of beta-hairpin formation. The mutant peptide with Val24-to-(D)Pro substitution, named V24P, formed a new amyloid-like beta-aggregate at high peptide concentration. The aggregate has most of the characteristics of amyloid fibrils, except fibril morphology. Moreover, the mutant peptide V24P, when mixed with Abeta40, can attenuate the cytotoxicity of Abeta40.


Assuntos
Peptídeos beta-Amiloides/química , Substituição de Aminoácidos , Peptídeos beta-Amiloides/farmacologia , Peptídeos beta-Amiloides/ultraestrutura , Animais , Benzotiazóis , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Vermelho Congo , Camundongos , Modelos Biológicos , Proteínas Mutantes/química , Proteínas Mutantes/farmacologia , Proteínas Mutantes/ultraestrutura , Mutação/genética , Peptídeos/química , Peptídeos/farmacologia , Prolina/genética , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Tiazóis/metabolismo , Fatores de Tempo , Valina/genética
20.
J Mol Biol ; 365(3): 812-24, 2007 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-17097677

RESUMO

A single multi-domain viral protein, termed Gag, is sufficient for assembly of retrovirus-like particles in mammalian cells. We have purified the human immunodeficiency virus type 1 (HIV-1) Gag protein (lacking myristate at its N terminus and the p6 domain at its C terminus) from bacteria. This protein is capable of assembly into virus-like particles in a defined in vitro system. We have reported that it is in monomer-dimer equilibrium in solution, and have described a mutant Gag protein that remains monomeric at high concentrations in solution. We report that the mutant protein retains several properties of wild-type Gag. This mutant enabled us to analyze solutions of monomeric protein. Hydrodynamic studies on the mutant protein showed that it is highly asymmetric, with a frictional ratio of 1.66. Small-angle neutron scattering (SANS) experiments confirmed its asymmetry and yielded an R(g) value of 34 A. Atomic-level structures of individual domains within Gag have previously been determined, but these domains are connected in Gag by flexible linkers. We constructed a series of models of the mutant Gag protein based on these domain structures, and tested each model computationally for its agreement with the experimental hydrodynamic and SANS data. The only models consistent with the data were those in which Gag was folded over, with its N-terminal matrix domain near its C-terminal nucleocapsid domain in three-dimensional space. Since Gag is a rod-shaped molecule in the assembled immature virion, these findings imply that Gag undergoes a major conformational change upon virus assembly.


Assuntos
Produtos do Gene gag/química , HIV-1/química , Dicroísmo Circular , Produtos do Gene gag/análise , Produtos do Gene gag/ultraestrutura , Humanos , Modelos Moleculares , Proteínas Mutantes/ultraestrutura , Mutação/genética , Difração de Nêutrons , Conformação Proteica , Espalhamento a Baixo Ângulo , Soluções
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA