Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 780
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 76(1): 138-147.e5, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31473102

RESUMO

Proteasomes are essential in all eukaryotic cells. However, their function and regulation remain considerably elusive, particularly those of less abundant variants. We demonstrate the human 20S proteasome recombinant assembly and confirmed the recombinant complex integrity biochemically and with a 2.6 Å resolution cryo-EM map. To assess its competence to form higher-order assemblies, we prepared and analyzed recombinant human 20S-PA200, a poorly characterized nuclear complex. Its 3.0 Å resolution cryo-EM structure reveals the PA200 unique architecture; the details of its intricate interactions with the proteasome, resulting in unparalleled proteasome α ring rearrangements; and the molecular basis for PA200 allosteric modulation of the proteasome active sites. Non-protein cryo-EM densities could be assigned to PA200-bound inositol phosphates, and we speculate regarding their functional role. Here we open extensive opportunities to study the fundamental properties of the diverse and distinct eukaryotic proteasome variants and to improve proteasome targeting under different therapeutic conditions.


Assuntos
Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Regulação Alostérica , Animais , Sítios de Ligação , Humanos , Fosfatos de Inositol/metabolismo , Modelos Moleculares , Proteínas Nucleares/genética , Proteínas Nucleares/ultraestrutura , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/ultraestrutura , Ligação Proteica , Conformação Proteica , Proteólise , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Células Sf9 , Spodoptera , Relação Estrutura-Atividade
2.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33753480

RESUMO

Through dominant mutations, aminoacyl-tRNA synthetases constitute the largest protein family linked to Charcot-Marie-Tooth disease (CMT). An example is CMT subtype 2N (CMT2N), caused by individual mutations spread out in AlaRS, including three in the aminoacylation domain, thereby suggesting a role for a tRNA-charging defect. However, here we found that two are aminoacylation defective but that the most widely distributed R329H is normal as a purified protein in vitro and in unfractionated patient cell samples. Remarkably, in contrast to wild-type (WT) AlaRS, all three mutant proteins gained the ability to interact with neuropilin 1 (Nrp1), the receptor previously linked to CMT pathogenesis in GlyRS. The aberrant AlaRS-Nrp1 interaction is further confirmed in patient samples carrying the R329H mutation. However, CMT2N mutations outside the aminoacylation domain do not induce the Nrp1 interaction. Detailed biochemical and biophysical investigations, including X-ray crystallography, small-angle X-ray scattering, hydrogen-deuterium exchange (HDX), switchSENSE hydrodynamic diameter determinations, and protease digestions reveal a mutation-induced structural loosening of the aminoacylation domain that correlates with the Nrp1 interaction. The b1b2 domains of Nrp1 are responsible for the interaction with R329H AlaRS. The results suggest Nrp1 is more broadly associated with CMT-associated members of the tRNA synthetase family. Moreover, we revealed a distinct structural loosening effect induced by a mutation in the editing domain and a lack of conformational impact with C-Ala domain mutations, indicating mutations in the same protein may cause neuropathy through different mechanisms. Our results show that, as with other CMT-associated tRNA synthetases, aminoacylation per se is not relevant to the pathology.


Assuntos
Alanina-tRNA Ligase/metabolismo , Doença de Charcot-Marie-Tooth/genética , Neuropilina-1/metabolismo , Alanina-tRNA Ligase/química , Alanina-tRNA Ligase/genética , Aminoacilação/genética , Células Cultivadas , Doença de Charcot-Marie-Tooth/sangue , Cristalografia por Raios X , Medição da Troca de Deutério , Humanos , Linfócitos , Mutação , Neuropilina-1/genética , Cultura Primária de Células , Ligação Proteica/genética , Domínios Proteicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Espalhamento a Baixo Ângulo
3.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33753488

RESUMO

Chloride ion-pumping rhodopsin (ClR) in some marine bacteria utilizes light energy to actively transport Cl- into cells. How the ClR initiates the transport is elusive. Here, we show the dynamics of ion transport observed with time-resolved serial femtosecond (fs) crystallography using the Linac Coherent Light Source. X-ray pulses captured structural changes in ClR upon flash illumination with a 550 nm fs-pumping laser. High-resolution structures for five time points (dark to 100 ps after flashing) reveal complex and coordinated dynamics comprising retinal isomerization, water molecule rearrangement, and conformational changes of various residues. Combining data from time-resolved spectroscopy experiments and molecular dynamics simulations, this study reveals that the chloride ion close to the Schiff base undergoes a dissociation-diffusion process upon light-triggered retinal isomerization.


Assuntos
Canais de Cloreto/metabolismo , Cloretos/metabolismo , Rodopsinas Microbianas/metabolismo , Cátions Monovalentes/metabolismo , Canais de Cloreto/isolamento & purificação , Canais de Cloreto/efeitos da radiação , Canais de Cloreto/ultraestrutura , Cristalografia/métodos , Radiação Eletromagnética , Lasers , Simulação de Dinâmica Molecular , Nocardioides , Conformação Proteica em alfa-Hélice/efeitos da radiação , Estrutura Terciária de Proteína/efeitos da radiação , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/efeitos da radiação , Proteínas Recombinantes/ultraestrutura , Retinaldeído/metabolismo , Retinaldeído/efeitos da radiação , Rodopsinas Microbianas/isolamento & purificação , Rodopsinas Microbianas/efeitos da radiação , Rodopsinas Microbianas/ultraestrutura , Água/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(50): 31832-31837, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33257579

RESUMO

TIA1, a protein critical for eukaryotic stress response and stress granule formation, is structurally characterized in full-length form. TIA1 contains three RNA recognition motifs (RRMs) and a C-terminal low-complexity domain, sometimes referred to as a "prion-related domain" or associated with amyloid formation. Under mild conditions, full-length (fl) mouse TIA1 spontaneously oligomerizes to form a metastable colloid-like suspension. RRM2 and RRM3, known to be critical for function, are folded similarly in excised domains and this oligomeric form of apo fl TIA1, based on NMR chemical shifts. By contrast, the termini were not detected by NMR and are unlikely to be amyloid-like. We were able to assign the NMR shifts with the aid of previously assigned solution-state shifts for the RRM2,3 isolated domains and homology modeling. We present a micellar model of fl TIA1 wherein RRM2 and RRM3 are colocalized, ordered, hydrated, and available for nucleotide binding. At the same time, the termini are disordered and phase separated, reminiscent of stress granule substructure or nanoscale liquid droplets.


Assuntos
Proteínas Intrinsicamente Desordenadas/ultraestrutura , Antígeno-1 Intracelular de Células T/ultraestrutura , Proteínas Intrinsicamente Desordenadas/metabolismo , Espectroscopia de Ressonância Magnética , Micelas , Microscopia Eletrônica , Modelos Moleculares , Dobramento de Proteína , Multimerização Proteica , Motivos de Ligação ao RNA , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Antígeno-1 Intracelular de Células T/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(44): 27676-27684, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33077592

RESUMO

Proteinaceous liquid-liquid phase separation (LLPS) occurs when a polypeptide coalesces into a dense phase to form a liquid droplet (i.e., condensate) in aqueous solution. In vivo, functional protein-based condensates are often referred to as membraneless organelles (MLOs), which have roles in cellular processes ranging from stress responses to regulation of gene expression. Late embryogenesis abundant (LEA) proteins containing seed maturation protein domains (SMP; PF04927) have been linked to storage tolerance of orthodox seeds. The mechanism by which anhydrobiotic longevity is improved is unknown. Interestingly, the brine shrimp Artemia franciscana is the only animal known to express such a protein (AfrLEA6) in its anhydrobiotic embryos. Ectopic expression of AfrLEA6 (AWM11684) in insect cells improves their desiccation tolerance and a fraction of the protein is sequestered into MLOs, while aqueous AfrLEA6 raises the viscosity of the cytoplasm. LLPS of AfrLEA6 is driven by the SMP domain, while the size of formed MLOs is regulated by a domain predicted to engage in protein binding. AfrLEA6 condensates formed in vitro selectively incorporate target proteins based on their surface charge, while cytoplasmic MLOs formed in AfrLEA6-transfected insect cells behave like stress granules. We suggest that AfrLEA6 promotes desiccation tolerance by engaging in two distinct molecular mechanisms: by raising cytoplasmic viscosity at even modest levels of water loss to promote cell integrity during drying and by forming condensates that may act as protective compartments for desiccation-sensitive proteins. Identifying and understanding the molecular mechanisms that govern anhydrobiosis will lead to significant advancements in preserving biological samples.


Assuntos
Adaptação Fisiológica , Proteínas de Artrópodes/metabolismo , Desidratação/fisiopatologia , Extremófilos/fisiologia , Organelas/metabolismo , Animais , Artemia , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/isolamento & purificação , Proteínas de Artrópodes/ultraestrutura , Linhagem Celular , Clonagem Molecular , Biologia Computacional , Citoplasma/metabolismo , Citoplasma/ultraestrutura , Dessecação , Drosophila melanogaster , Embrião não Mamífero , Desenvolvimento Embrionário , Extremófilos/citologia , Microscopia Eletrônica de Varredura , Organelas/ultraestrutura , Pressão Osmótica/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura
6.
Proc Natl Acad Sci U S A ; 117(27): 15620-15631, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32576689

RESUMO

Repulsive guidance molecules (RGMs) are cell surface proteins that regulate the development and homeostasis of many tissues and organs, including the nervous, skeletal, and immune systems. They control fundamental biological processes, such as migration and differentiation by direct interaction with the Neogenin (NEO1) receptor and function as coreceptors for the bone morphogenetic protein (BMP)/growth differentiation factor (GDF) family. We determined crystal structures of all three human RGM family members in complex with GDF5, as well as the ternary NEO1-RGMB-GDF5 assembly. Surprisingly, we show that all three RGMs inhibit GDF5 signaling, which is in stark contrast to RGM-mediated enhancement of signaling observed for other BMPs, like BMP2. Despite their opposite effect on GDF5 signaling, RGMs occupy the BMP type 1 receptor binding site similar to the observed interactions in RGM-BMP2 complexes. In the NEO1-RGMB-GDF5 complex, RGMB physically bridges NEO1 and GDF5, suggesting cross-talk between the GDF5 and NEO1 signaling pathways. Our crystal structures, combined with structure-guided mutagenesis of RGMs and BMP ligands, binding studies, and cellular assays suggest that RGMs inhibit GDF5 signaling by competing with GDF5 type 1 receptors. While our crystal structure analysis and in vitro binding data initially pointed towards a simple competition mechanism between RGMs and type 1 receptors as a possible basis for RGM-mediated GDF5 inhibition, further experiments utilizing BMP2-mimicking GDF5 variants clearly indicate a more complex mechanism that explains how RGMs can act as a functionality-changing switch for two structurally and biochemically similar signaling molecules.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas Ligadas por GPI/metabolismo , Fator 5 de Diferenciação de Crescimento/metabolismo , Proteína da Hemocromatose/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/ultraestrutura , Moléculas de Adesão Celular Neuronais/ultraestrutura , Cristalografia por Raios X , Proteínas Ligadas por GPI/ultraestrutura , Fator 5 de Diferenciação de Crescimento/ultraestrutura , Proteína da Hemocromatose/ultraestrutura , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Proteínas do Tecido Nervoso/ultraestrutura , Domínios Proteicos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Transdução de Sinais
7.
Proc Natl Acad Sci U S A ; 117(1): 317-327, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31852826

RESUMO

Host-pathogen interactions are central to understanding microbial pathogenesis. The staphylococcal pore-forming cytotoxins hijack important immune molecules but little is known about the underlying molecular mechanisms of cytotoxin-receptor interaction and host specificity. Here we report the structures of a staphylococcal pore-forming cytotoxin, leukocidin GH (LukGH), in complex with its receptor (the α-I domain of complement receptor 3, CD11b-I), both for the human and murine homologs. We observe 2 binding interfaces, on the LukG and the LukH protomers, and show that human CD11b-I induces LukGH oligomerization in solution. LukGH binds murine CD11b-I weakly and is inactive toward murine neutrophils. Using a LukGH variant engineered to bind mouse CD11b-I, we demonstrate that cytolytic activity does not only require binding but also receptor-dependent oligomerization. Our studies provide an unprecedented insight into bicomponent leukocidin-host receptor interaction, enabling the development of antitoxin approaches and improved animal models to explore these approaches.


Assuntos
Proteínas de Bactérias/metabolismo , Antígeno CD11b/metabolismo , Leucocidinas/metabolismo , Antígeno de Macrófago 1/metabolismo , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/ultraestrutura , Antígeno CD11b/imunologia , Antígeno CD11b/ultraestrutura , Linhagem Celular , Membrana Celular/metabolismo , Cristalografia por Raios X , Humanos , Leucocidinas/imunologia , Antígeno de Macrófago 1/imunologia , Antígeno de Macrófago 1/ultraestrutura , Camundongos , Modelos Moleculares , Neutrófilos/citologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Domínios Proteicos/imunologia , Multimerização Proteica/imunologia , Coelhos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Especificidade da Espécie , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade
8.
Prep Biochem Biotechnol ; 53(6): 672-682, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36244016

RESUMO

Lipopolysaccharide (LPS), as a stubborn contamination, should be monitored and kept in an acceptable level during the pharmaceutical production process. Recombinant hepatitis B surface antigen (r-HBsAg) is one of the recombinant biological products, which is probable to suffer from extrinsic endotoxin due to its long and complex production process. This research aims to assess the potential interaction between LPS and r-HBsAg by recruiting immunoaffinity chromatography (IAC) as a novel tool to quantify the interaction. Molecular modeling was performed on the HBsAg molecule to theoretically predict its potential binding and interaction sites. Then dynamic light scattering (DLS) analysis was implemented on HBsAg, LPS, and mixtures of them to reveal the interaction. The virus-like particle (VLP) structure of HBsAg and the ribbon-like structure of LPS were visualized by transmission electron microscopy (TEM). Finally, the interaction was quantified by applying various LPS/HBsAg ratios ranging from 1.67 to 120 EU/dose in the IAC. Consequently, the LPS/HBsAg ratios in the eluate were measured from 1.67 to a maximum of 92.5 EU/dose. The results indicated that 77 to 100% of total LPS interacted with HBsAg by an inverse relationship to the incubated LPS concentration. The findings implied that the introduced procedure is remarkably practical in the quantification of LPS interaction with a target recombinant protein.


Assuntos
Cromatografia de Afinidade , Antígenos de Superfície da Hepatite B , Lipopolissacarídeos , Proteínas Recombinantes , Lipopolissacarídeos/análise , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/ultraestrutura , Antígenos de Superfície da Hepatite B/química , Antígenos de Superfície da Hepatite B/genética , Antígenos de Superfície da Hepatite B/isolamento & purificação , Antígenos de Superfície da Hepatite B/ultraestrutura , Microscopia Eletrônica de Transmissão , Vacinas contra Hepatite B/química , Vacinas contra Hepatite B/genética , Vacinas contra Hepatite B/isolamento & purificação , Modelos Químicos , Sequência de Aminoácidos , Difusão Dinâmica da Luz , Cromatografia de Afinidade/métodos
9.
J Biol Chem ; 296: 100737, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33933456

RESUMO

Synucleinopathies are neurodegenerative diseases characterized by the presence of intracellular deposits containing the protein alpha-synuclein (aSYN) within patients' brains. It has been shown that aSYN can form structurally distinct fibrillar assemblies, also termed polymorphs. We previously showed that distinct aSYN polymorphs assembled in vitro, named fibrils, ribbons, and fibrils 91, differentially bind to and seed the aggregation of endogenous aSYN in neuronal cells, which suggests that distinct synucleinopathies may arise from aSYN polymorphs. In order to better understand the differential interactions of aSYN polymorphs with their partner proteins, we mapped aSYN polymorphs surfaces. We used limited proteolysis, hydrogen-deuterium exchange, and differential antibody accessibility to identify amino acids on their surfaces. We showed that the aSYN C-terminal region spanning residues 94 to 140 exhibited similarly high solvent accessibility in these three polymorphs. However, the N-terminal amino acid residues 1 to 38 of fibrils were exposed to the solvent, while only residues 1 to 18 within fibrils 91 were exposed, and no N-terminal residues within ribbons were solvent-exposed. It is likely that these differences in surface accessibility contribute to the differential binding of distinct aSYN polymorphs to partner proteins. We thus posit that the polypeptides exposed on the surface of distinct aSYN fibrillar polymorphs are comparable to fingerprints. Our findings have diagnostic and therapeutic potential, particularly in the prion-like propagation of fibrillar aSYN, as they can facilitate the design of ligands that specifically bind and distinguish between fibrillar polymorphs.


Assuntos
Sinucleinopatias/metabolismo , alfa-Sinucleína/metabolismo , Humanos , Conformação Proteica , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Solventes/metabolismo , Sinucleinopatias/patologia , alfa-Sinucleína/química , alfa-Sinucleína/ultraestrutura
10.
Proc Natl Acad Sci U S A ; 116(29): 14547-14556, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31249143

RESUMO

Light-sensitive G protein-coupled receptors (GPCRs)-rhodopsins-absorb photons to isomerize their covalently bound retinal, triggering conformational changes that result in downstream signaling cascades. Monostable rhodopsins release retinal upon isomerization as opposed to the retinal in bistable rhodopsins that "reisomerize" upon absorption of a second photon. Understanding the mechanistic differences between these light-sensitive GPCRs has been hindered by the scarcity of recombinant models of the latter. Here, we reveal the high-resolution crystal structure of a recombinant bistable rhodopsin, jumping spider rhodopsin-1, bound to the inverse agonist 9-cis retinal. We observe a water-mediated network around the ligand hinting toward the basis of their bistable nature. In contrast to bovine rhodopsin (monostable), the transmembrane bundle of jumping spider rhodopsin-1 as well that of the bistable squid rhodopsin adopts a more "activation-ready" conformation often observed in other nonphotosensitive class A GPCRs. These similarities suggest the role of jumping spider rhodopsin-1 as a potential model system in the study of the structure-function relationship of both photosensitive and nonphotosensitive class A GPCRs.


Assuntos
Proteínas de Artrópodes/ultraestrutura , Rodopsina/ultraestrutura , Transdução de Sinais/efeitos da radiação , Aranhas , Animais , Proteínas de Artrópodes/isolamento & purificação , Proteínas de Artrópodes/metabolismo , Cristalografia por Raios X , Células HEK293 , Humanos , Ligantes , Luz , Simulação de Dinâmica Molecular , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestrutura , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Rodopsina/isolamento & purificação , Rodopsina/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade
11.
Proc Natl Acad Sci U S A ; 116(26): 13131-13136, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31182573

RESUMO

Steviol glucosides, such as stevioside and rebaudioside A, are natural products roughly 200-fold sweeter than sugar and are used as natural, noncaloric sweeteners. Biosynthesis of rebaudioside A, and other related stevia glucosides, involves formation of the steviol diterpenoid followed by a series of glycosylations catalyzed by uridine diphosphate (UDP)-dependent glucosyltransferases. UGT76G1 from Stevia rebaudiana catalyzes the formation of the branched-chain glucoside that defines the stevia molecule and is critical for its high-intensity sweetness. Here, we report the 3D structure of the UDP-glucosyltransferase UGT76G1, including a complex of the protein with UDP and rebaudioside A bound in the active site. The X-ray crystal structure and biochemical analysis of site-directed mutants identifies a catalytic histidine and how the acceptor site of UGT76G1 achieves regioselectivity for branched-glucoside synthesis. The active site accommodates a two-glucosyl side chain and provides a site for addition of a third sugar molecule to the C3' position of the first C13 sugar group of stevioside. This structure provides insight on the glycosylation of other naturally occurring sweeteners, such as the mogrosides from monk fruit, and a possible template for engineering of steviol biosynthesis.


Assuntos
Diterpenos do Tipo Caurano/metabolismo , Glucosídeos/biossíntese , Glucosiltransferases/ultraestrutura , Proteínas de Plantas/ultraestrutura , Stevia/enzimologia , Vias Biossintéticas/genética , Coenzimas/metabolismo , Cristalografia por Raios X , Diterpenos do Tipo Caurano/química , Ensaios Enzimáticos , Glucosídeos/química , Glucosiltransferases/genética , Glucosiltransferases/isolamento & purificação , Glucosiltransferases/metabolismo , Engenharia Metabólica/métodos , Mutagênese Sítio-Dirigida , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Edulcorantes/química , Edulcorantes/metabolismo , Difosfato de Uridina/metabolismo
12.
Biochemistry ; 60(47): 3644-3658, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34730940

RESUMO

The aggregation of α-synuclein (αSN) and increased oxidative stress leading to lipid peroxidation are pathological characteristics of Parkinson's disease (PD). Here, we report that aggregation of αSN in the presence of lipid peroxidation products 4-hydroxy-2-nonenal (HNE) and 4-oxo-2-nonenal (ONE) increases the stability and the yield of αSN oligomers (αSO). Further, we show that ONE is more efficient than HNE at inducing αSO. In addition, we demonstrate that the two αSO differ in both size and shape. ONE-αSO are smaller in size than HNE-αSO, except when they are formed at a high molar excess of aldehyde. In both monomeric and oligomeric αSN, His50 is the main target of HNE modification, and HNE-induced oligomerization is severely retarded in the mutant His50Ala αSN. In contrast, ONE-induced aggregation of His50Ala αSN occurs readily, demonstrating the different pathways for inducing αSN aggregation by HNE and ONE. Our results show different morphologies of the HNE-treated and ONE-treated αSO and different roles of His50 in their modification of αSN, but we also observe structural similarities between these αSO and the non-treated αSO, e.g., flexible C-terminus, a folded core composed of the N-terminal and NAC region. Furthermore, HNE-αSO show a similar deuterium uptake as a previously characterized oligomer formed by non-treated αSO, suggesting that the backbone conformational dynamics of their folded cores resemble one another.


Assuntos
Aldeídos/metabolismo , Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo , Aldeídos/química , Linhagem Celular Tumoral , Humanos , Peroxidação de Lipídeos , Ressonância Magnética Nuclear Biomolecular , Agregados Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Espalhamento a Baixo Ângulo , Difração de Raios X , alfa-Sinucleína/química , alfa-Sinucleína/isolamento & purificação , alfa-Sinucleína/ultraestrutura
13.
Biochem Biophys Res Commun ; 534: 292-296, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33272573

RESUMO

Formation of Aß oligomers and fibrils plays a central role in the pathogenesis of Alzheimer's disease. There are two major forms of Aß in the brain: Aß42 and Aß40. Aß42 is the major component of the amyloid plaques, but the overall abundance of Aß40 is several times that of Aß42. In vitro experiments show that Aß42 and Aß40 affect each other's aggregation. In mouse models of Alzheimer's disease, overexpression of Aß40 has been shown to reduce the plaque pathology, suggesting that Aß42 and Aß40 also interact in vivo. Here we address the question of whether Aß42 and Aß40 interact with each other in the formation of oligomers using electron paramagnetic resonance (EPR) spectroscopy. When the Aß42 oligomers were formed using only spin-labeled Aß42, the dipolar interaction between spin labels that are within 20 Å range broadened the EPR spectrum and reduced its amplitude. Oligomers formed with a mixture of spin-labeled Aß42 and wild-type Aß42 gave an EPR spectrum with higher amplitude due to weakened spin-spin interactions, suggesting molecular mixing of labeled and wild-type Aß42. When spin-labeled Aß42 and wild-type Aß40 were mixed to form oligomers, the resulting EPR spectrum also showed reduced amplitude, suggesting that wild-type Aß40 can also form oligomers with spin-labeled Aß42. Therefore, our results suggest that Aß42 and Aß40 form mixed oligomers with direct molecular interactions. Our results point to the importance of investigating Aß42-Aß40 interactions in the brain for a complete understanding of Alzheimer's pathogenesis and therapeutic interventions.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/ultraestrutura , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Espectroscopia de Ressonância de Spin Eletrônica , Humanos , Técnicas In Vitro , Camundongos , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/ultraestrutura , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Placa Amiloide/ultraestrutura , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/ultraestrutura , Marcadores de Spin
14.
Mol Pharm ; 18(2): 726-734, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32530637

RESUMO

Two well-defined synthetic polyphosphazene immunoadjuvants, PCPP and PCEP, were studied for their ability to potentiate the immune response to the hepatitis C virus (HCV) E2 glycoprotein antigen in vivo. We report that PCEP induced significantly higher serum neutralization and HCV-specific IgG titers in mice compared to other adjuvants used in the study: PCPP, Alum, and Addavax. PCEP also shifted the response toward the desirable balanced Th1/Th2 immunity, as evaluated by the antibody isotype ratio (IgG2a/IgG1). The in vivo results were analyzed in the context of antigen-adjuvant molecular interactions in the system and in vitro immunostimulatory activity of formulations. Asymmetric flow field flow fractionation (AF4) and dynamic light scattering (DLS) analysis showed that both PCPP and PCEP spontaneously self-assemble with the E2 glycoprotein with the formation of multimeric water-soluble complexes, which demonstrates the role of polyphosphazene macromolecules as vaccine delivery vehicles. Intrinsic in vitro immunostimulatory activity of polyphosphazene adjuvants, which was assessed using a mouse macrophage cell line, revealed comparable activities of both polymers and did not provide an explanation of their in vivo performance. However, PCEP complexes with E2 displayed greater stability against agglomeration and improved in vitro immunostimulatory activity compared to those of PCPP, which is in line with superior in vivo performance of PCEP. The results emphasize the importance of often neglected antigen-polyphosphazene self-assembly mechanisms in formulations, which can provide important insights on their in vivo behavior and facilitate the establishment of a structure-activity relationship for this important class of immunoadjuvants.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos da Hepatite C/administração & dosagem , Hepatite C/prevenção & controle , Proteínas do Envelope Viral/administração & dosagem , Vacinas contra Hepatite Viral/administração & dosagem , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Portadores de Fármacos/química , Composição de Medicamentos/métodos , Feminino , Hepacivirus/imunologia , Hepatite C/imunologia , Hepatite C/virologia , Antígenos da Hepatite C/imunologia , Antígenos da Hepatite C/ultraestrutura , Humanos , Imunogenicidade da Vacina , Camundongos , Modelos Animais , Compostos Organofosforados/administração & dosagem , Compostos Organofosforados/imunologia , Polímeros/administração & dosagem , Polímeros/química , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/ultraestrutura , Relação Estrutura-Atividade , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/isolamento & purificação , Proteínas do Envelope Viral/ultraestrutura , Vacinas contra Hepatite Viral/imunologia
15.
Arterioscler Thromb Vasc Biol ; 40(3): 624-637, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31969014

RESUMO

OBJECTIVE: The αIIbß3 antagonist antiplatelet drug abciximab is the chimeric antigen-binding fragment comprising the variable regions of murine monoclonal antibody 7E3 and the constant domains of human IgG1 and light chain κ. Previous mutagenesis studies suggested that abciximab binds to the ß3 C177-C184 specificity-determining loop (SDL) and Trp129 on the adjacent ß1-α1 helix. These studies could not, however, assess whether 7E3 or abciximab prevents fibrinogen binding by steric interference, disruption of either the αIIbß3-binding pocket for fibrinogen or the ß3 SDL (which is not part of the binding pocket but affects fibrinogen binding), or some combination of these effects. To address this gap, we used cryo-electron microscopy to determine the structure of the αIIbß3-abciximab complex at 2.8 Å resolution. Approach and Results: The interacting surface of abciximab is comprised of residues from all 3 complementarity-determining regions of both the light and heavy chains, with high representation of aromatic residues. Binding is primarily to the ß3 SDL and neighboring residues, the ß1-α1 helix, and ß3 residues Ser211, Val212 and Met335. Unexpectedly, the structure also indicated several interactions with αIIb. As judged by the cryo-electron microscopy model, molecular-dynamics simulations, and mutagenesis, the binding of abciximab does not appear to rely on the interaction with the αIIb residues and does not result in disruption of the fibrinogen-binding pocket; it does, however, compress and reduce the flexibility of the SDL. CONCLUSIONS: We deduce that abciximab prevents ligand binding by steric interference, with a potential contribution via displacement of the SDL and limitation of the flexibility of the SDL residues.


Assuntos
Abciximab/ultraestrutura , Microscopia Crioeletrônica , Integrina alfa2/ultraestrutura , Integrina beta3/ultraestrutura , Inibidores da Agregação Plaquetária , Abciximab/metabolismo , Sítios de Ligação , Ligação Competitiva , Células HEK293 , Humanos , Integrina alfa2/genética , Integrina alfa2/metabolismo , Integrina beta3/genética , Integrina beta3/metabolismo , Ligantes , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Mutação , Inibidores da Agregação Plaquetária/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/ultraestrutura , Relação Estrutura-Atividade
16.
Biol Pharm Bull ; 44(9): 1202-1209, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34471048

RESUMO

Peroxisome proliferator-activated receptor (PPAR)α, a member of the nuclear receptor family, is a transcription factor that regulates the expression of genes related to lipid metabolism in a ligand-dependent manner, and has attracted attention as a target for hypolipidemic drugs. We have been developing phenylpropaonic acid derivatives as PPARα-targeted drug candidates for the treatment of metabolic diseases. Recently, we have developed the "ligand-exchange soaking method," which crystallizes the recombinant PPARα ligand-binding domain (LBD) as a complex with intrinsic fatty acids derived from an expression host Escherichia (E.) coli and thereafter replaces them with other higher-affinity ligands by soaking. Here we applied this method for preparation of cocrystals of PPARα LBD with its ligands that have not been obtained with the conventional cocrystallization method. We revealed the high-resolution structures of the cocrystals of PPARα LBD and the three synthetic phenylpropaonic acid derivatives: TIPP-703, APHM19, and YN4pai, the latter two of which are the first observations. The overall structures of cocrystals obtained from the two methods are identical and illustrate the close interaction between these ligands and the surrounding amino acid residues of PPARα LBD. This ligand-exchange soaking method could be applicable to high throughput preparations of co-crystals with another subtype PPARδ LBD for high resolution X-ray crystallography, because it also crystallizes in complex with intrinsic fatty acid(s) while not in the apo-form.


Assuntos
PPAR alfa/ultraestrutura , Fenilpropionatos/metabolismo , Domínios Proteicos , Humanos , Ligantes , PPAR alfa/isolamento & purificação , PPAR alfa/metabolismo , Fenilpropionatos/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Difração de Raios X
17.
Biol Pharm Bull ; 44(9): 1196-1201, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34471047

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARγ) is a nuclear receptor and the molecular target of thiazolidinedione-class antidiabetic drugs. It has been reported that the loss of function R288H mutation in the human PPARγ ligand-binding domain (LBD) may be associated with the onset of colon cancer. A previous in vitro study showed that this mutation dampens 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2, a natural PPARγ agonist)-dependent transcriptional activation; however, it is poorly understood why the function of the R288H mutant is impaired and what role this arginine (Arg) residue plays. In this study, we found that the apo-form of R288H PPARγ mutant displays several altered conformational arrangements of the amino acid side chains in LBD: 1) the loss of a salt bridge between Arg288 and Glu295 leads to increased helix 3 movement; 2) closer proximity of Gln286 and His449 via a hydrogen bond, and closer proximity of Cys285 and Phe363 via hydrophobic interaction, stabilize the helix 3-helix 11 interaction; and 3) there is steric hindrance between Cys285/Gln286/Ser289/His449 and the flexible ligands 15d-PGJ2, 6-oxotetracosahexaenoic acid (6-oxoTHA), and 17-oxodocosahexaenoic acid (17-oxoDHA). These results suggest why Arg288 plays an important role in ligand binding and why the R288H mutation is disadvantageous for flexible ligand binding.


Assuntos
PPAR gama/genética , Substituição de Aminoácidos , Animais , Arginina/genética , Células COS , Chlorocebus aethiops , Cristalografia por Raios X , Histidina/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Mutação com Perda de Função , PPAR gama/isolamento & purificação , PPAR gama/metabolismo , PPAR gama/ultraestrutura , Domínios Proteicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Relação Estrutura-Atividade
18.
Biol Pharm Bull ; 44(9): 1210-1219, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34471049

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptor-type transcription factors that consist of three subtypes (α, γ, and ß/δ) with distinct functions and PPAR dual/pan agonists are expected to be the next generation of drugs for metabolic diseases. Saroglitazar is the first clinically approved PPARα/γ dual agonist for treatment of diabetic dyslipidemia and is currently in clinical trials to treat non-alcoholic fatty liver disease (NAFLD); however, the structural information of its interaction with PPARα/γ remains unknown. We recently revealed the high-resolution co-crystal structure of saroglitazar and the PPARα-ligand binding domain (LBD) through X-ray crystallography, and in this study, we report the structure of saroglitazar and the PPARγ-LBD. Saroglitazar was located at the center of "Y"-shaped PPARγ-ligand-binding pocket (LBP), just as it was in the respective region of PPARα-LBP. Its carboxylic acid was attached to four amino acids (Ser289/His323/His449/Thr473), which contributes to the stabilization of Activating Function-2 helix 12, and its phenylpyrrole moiety was rotated 121.8 degrees in PPARγ-LBD from that in PPARα-LBD to interact with Phe264. PPARδ-LBD has the consensus four amino acids (Thr253/His287/His413/Tyr437) towards the carboxylic acids of its ligands, but it seems to lack sufficient space to accept saroglitazar because of the steric hindrance between the Trp228 or Arg248 residue of PPARδ-LBD and its methylthiophenyl moiety. Accordingly, in a coactivator recruitment assay, saroglitazar activated PPARα-LBD and PPARγ-LBD but not PPARδ-LBD, whereas glycine substitution of either Trp228, Arg248, or both of PPARδ-LBD conferred saroglitazar concentration-dependent activation. Our findings may be valuable in the molecular design of PPARα/γ dual or PPARα/γ/δ pan agonists.


Assuntos
Hipolipemiantes/farmacologia , PPAR alfa/ultraestrutura , PPAR gama/ultraestrutura , Fenilpropionatos/farmacologia , Pirróis/farmacologia , Sítios de Ligação , Cristalografia por Raios X , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dislipidemias/tratamento farmacológico , Dislipidemias/etiologia , Humanos , Hipolipemiantes/química , Hipolipemiantes/uso terapêutico , Metabolismo dos Lipídeos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , PPAR alfa/agonistas , PPAR alfa/isolamento & purificação , PPAR alfa/metabolismo , PPAR gama/agonistas , PPAR gama/isolamento & purificação , PPAR gama/metabolismo , Fenilpropionatos/química , Fenilpropionatos/uso terapêutico , Domínios Proteicos , Pirróis/química , Pirróis/uso terapêutico , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura
19.
Molecules ; 26(20)2021 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-34684779

RESUMO

Caveolin-1 (CAV1), a membrane protein that is necessary for the formation and maintenance of caveolae, is a promising drug target for the therapy of various diseases, such as cancer, diabetes, and liver fibrosis. The biology and pathology of caveolae have been widely investigated; however, very little information about the structural features of full-length CAV1 is available, as well as its biophysical role in reshaping the cellular membrane. Here, we established a method, with high reliability and reproducibility, for the expression and purification of CAV1. Amyloid-like properties of CAV1 and its C-terminal peptide CAV1(168-178) suggest a structural basis for the short linear CAV1 assemblies that have been recently observed in caveolin polyhedral cages in Escherichia coli (E. coli). Reconstitution of CAV1 into artificial lipid membranes induces a caveolae-like membrane curvature. Structural characterization of CAV1 in the membrane by solid-state nuclear magnetic resonance (ssNMR) indicate that it is largely α-helical, with very little ß-sheet content. Its scaffolding domain adopts a α-helical structure as identified by chemical shift analysis of threonine (Thr). Taken together, an in vitro model was developed for the CAV1 structural study, which will further provide meaningful evidences for the design and screening of bioactive compounds targeting CAV1.


Assuntos
Caveolina 1/química , Sequência de Aminoácidos , Caveolina 1/genética , Caveolina 1/ultraestrutura , Humanos , Espectroscopia de Ressonância Magnética , Lipídeos de Membrana/química , Membranas Artificiais , Microscopia Eletrônica de Transmissão , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/ultraestrutura
20.
Mol Pharmacol ; 97(4): 287-294, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32086254

RESUMO

Because of ß-lactamase-mediated resistance, ß-lactam antibiotics were long considered ineffective drugs for tuberculosis (TB) treatment. However, some ß-lactams, including meropenem and faropenem, are being re-evaluated in patients infected with TB. Penicillin-binding protein (PBP) 3, or ftsI, is an essential transpeptidase in Mycobacterium tuberculosis (Mtb) required for cell division, and thus it is an important drug target. Structures of apo MtbPBP3 and of complexes with five ß-lactams, including meropenem and faropenem, reveal how they cause inactivation via formation of hydrolytically stable acyl-enzyme complexes. The structures reveal unique features of the antibiotic interactions, both in terms of differences in their binding to MtbPBP3 and in comparison with structures of other PBPs and serine ß-lactamases, including the tautomerization status of the carbapenem-derived acyl-enzyme complexes. The results suggest that rather than hoping PBP inhibitors developed for other infections will work against TB, work should focus on developing PBP inhibitors specialized for treating TB. SIGNIFICANCE STATEMENT: The structures of Mycobacterium tuberculosis penicillin-binding protein 3, an essential protein in M. tuberculosis, in complex with a number of widely used ß-lactam antibiotics (e.g., meropenem, aztreonam, and amoxicillin) were solved. These data provide new insights for next-generation rational approaches to design tuberculosis (TB)-specific ß-lactam or nonlactam antibiotics. This manuscript is a seminal article in the field of anti-TB drug discovery and suitable for the broad readership.


Assuntos
Antibacterianos/química , Mycobacterium tuberculosis/fisiologia , Proteínas de Ligação às Penicilinas/ultraestrutura , Resistência beta-Lactâmica , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Cristalografia por Raios X , Desenho de Fármacos , Meropeném/química , Meropeném/farmacologia , Testes de Sensibilidade Microbiana , Modelos Moleculares , Mycobacterium tuberculosis/efeitos dos fármacos , Proteínas de Ligação às Penicilinas/antagonistas & inibidores , Proteínas de Ligação às Penicilinas/metabolismo , Ligação Proteica , Domínios Proteicos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia , beta-Lactamas/química , beta-Lactamas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA