Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
1.
Nat Struct Mol Biol ; 28(11): 936-944, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34759376

RESUMO

The ß1-adrenergic receptor (ß1-AR) can activate two families of G proteins. When coupled to Gs, ß1-AR increases cardiac output, and coupling to Gi leads to decreased responsiveness in myocardial infarction. By comparative structural analysis of turkey ß1-AR complexed with either Gi or Gs, we investigate how a single G-protein-coupled receptor simultaneously signals through two G proteins. We find that, although the critical receptor-interacting C-terminal α5-helices on Gαi and Gαs interact similarly with ß1-AR, the overall interacting modes between ß1-AR and G proteins vary substantially. Functional studies reveal the importance of the differing interactions and provide evidence that the activation efficacy of G proteins by ß1-AR is determined by the entire three-dimensional interaction surface, including intracellular loops 2 and 4 (ICL2 and ICL4).


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Estrutura Terciária de Proteína/fisiologia , Receptores Adrenérgicos beta 1/metabolismo , Animais , Débito Cardíaco/genética , Débito Cardíaco/fisiologia , Linhagem Celular , Microscopia Crioeletrônica , AMP Cíclico/metabolismo , Ativação Enzimática/fisiologia , Células HEK293 , Cardiopatias/patologia , Humanos , Hipertensão/patologia , Isoproterenol/química , Estrutura Secundária de Proteína/fisiologia , Células Sf9 , Transdução de Sinais/fisiologia
2.
Mol Cells ; 44(10): 758-769, 2021 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-34711692

RESUMO

Calcium homeostasis modulator 1 (CALHM1) is a membrane protein with four transmembrane helices that form an octameric ion channel with voltage-dependent activation. There are four conserved cysteine (Cys) residues in the extracellular domain that form two intramolecular disulfide bonds. We investigated the roles of C42-C127 and C44-C161 in human CALHM1 channel biogenesis and the ionic current (ICALHM1). Replacing Cys with Ser or Ala abolished the membrane trafficking as well as ICALHM1. Immunoblotting analysis revealed dithiothreitol-sensitive multimeric CALHM1, which was markedly reduced in C44S and C161S, but preserved in C42S and C127S. The mixed expression of C42S and wild-type did not show a dominant-negative effect. While the heteromeric assembly of CALHM1 and CALHM3 formed active ion channels, the co-expression of C42S and CALHM3 did not produce functional channels. Despite the critical structural role of the extracellular cysteine residues, a treatment with the membrane-impermeable reducing agent tris(2-carboxyethyl) phosphine (TCEP, 2 mM) did not affect ICALHM1 for up to 30 min. Interestingly, incubation with TCEP (2 mM) for 2-6 h reduced both ICALHM1 and the surface expression of CALHM1 in a time-dependent manner. We propose that the intramolecular disulfide bonds are essential for folding, oligomerization, trafficking and maintenance of CALHM1 in the plasma membrane, but dispensable for the voltage-dependent activation once expressed on the plasma membrane.


Assuntos
Canais de Cálcio/metabolismo , Dissulfetos/metabolismo , Homeostase/fisiologia , Glicoproteínas de Membrana/metabolismo , Estrutura Secundária de Proteína/fisiologia , Humanos
3.
Int J Mol Sci ; 22(10)2021 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-34069753

RESUMO

Hydrocarbon stapling is a useful tool for stabilizing the secondary structure of peptides. Among several methods, hydrocarbon stapling at i,i + 1 positions was not extensively studied, and their secondary structures are not clarified. In this study, we investigate i,i + 1 hydrocarbon stapling between cis-4-allyloxy-l-proline and various olefin-tethered amino acids. Depending on the ring size of the stapled side chains and structure of the olefin-tethered amino acids, E- or Z-selectivities were observed during the ring-closing metathesis reaction (E/Z was up to 8.5:1 for 17-14-membered rings and up to 1:20 for 13-membered rings). We performed X-ray crystallographic analysis of hydrocarbon stapled peptide at i,i + 1 positions. The X-ray crystallographic structure suggested that the i,i + 1 staple stabilizes the peptide secondary structure to the right-handed α-helix. These findings are especially important for short oligopeptides because the employed stapling method uses two minimal amino acid residues adjacent to each other.


Assuntos
Hidrocarbonetos/química , Peptídeos/química , Estabilidade Proteica/efeitos dos fármacos , Alcenos/química , Sequência de Aminoácidos/genética , Aminoácidos/química , Dicroísmo Circular/métodos , Cristalografia por Raios X/métodos , Oligopeptídeos/química , Prolina/química , Conformação Proteica em alfa-Hélice/fisiologia , Estrutura Secundária de Proteína/fisiologia , Raios X
4.
Biochemistry ; 60(6): 431-439, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33533248

RESUMO

The field of de novo protein design has met with considerable success over the past few decades. Heme, a cofactor, has often been introduced to impart a diverse array of functions to a protein, ranging from electron transport to respiration. In nature, heme is found to occur predominantly in α-helical structures over ß-sheets, which has resulted in significant designs of heme proteins utilizing coiled-coil helices. By contrast, there are only a few known ß-sheet proteins that bind heme and designs of ß-sheets frequently result in amyloid-like aggregates. This review reflects on our success in designing a series of multistranded ß-sheet heme binding peptides that are well folded in both aqueous and membrane-like environments. Initially, we designed a ß-hairpin peptide that self-assembles to bind heme and performs peroxidase activity in membrane. The ß-hairpin was optimized further to accommodate a heme binding pocket within multistranded ß-sheets for catalysis and electron transfer in membranes. Furthermore, we de novo designed and characterized ß-sheet peptides and miniproteins that are soluble in an aqueous environment capable of binding single and multiple hemes with high affinity and stability. Collectively, these studies highlight the substantial progress made toward the design of functional ß-sheets.


Assuntos
Hemeproteínas/química , Conformação Proteica em Folha beta/fisiologia , Engenharia de Proteínas/métodos , Sequência de Aminoácidos , Dicroísmo Circular , Heme/química , Heme/metabolismo , Hemeproteínas/metabolismo , Oxirredução , Peptídeos/química , Dobramento de Proteína , Estrutura Secundária de Proteína/fisiologia
5.
Biochemistry ; 59(47): 4488-4498, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33190490

RESUMO

ZntA from Escherichia coli confers resistance to toxic concentrations of Pb2+, Zn2+, and Cd2+. It is a member of the P1B-ATPase transporter superfamily, which includes the human Cu+-transporting proteins ATP7A and ATP7B. P1B-type ATPases typically have a hydrophilic N-terminal metal-binding domain and eight transmembrane helices. A splice variant of ATP7B was reported, which has 100-fold higher night-specific expression in the pineal gland; it lacks the entire N-terminal domain and the first four transmembrane helices. Here, we report our findings with Δ231-ZntA, a similar truncation we created in ZntA. Δ231-ZntA has no in vivo and greatly reduced in vitro activity. It binds one metal ion per dimer at the transmembrane site, with a 15-19000-fold higher binding affinity, indicating highly significant changes in the dimer structure of Δ231-ZntA relative to that of ZntA. Cd2+ has the highest affinity for Δ231-ZntA, in contrast to ZntA, which has the highest affinity for Pb2+. Site-specific mutagenesis of the metal-binding residues, 392Cys, 394Cys, and 714Asp, showed that there is considerable flexibility at the metal-binding site, with any two of these three residues able to bind Zn2+ and Pb2+ unlike in ZntA. However, Cd2+ binds to only 392Cys and 714Asp, with 394Cys not involved in Cd2+ binding. Three-dimensional homology models show that there is a dramatic difference between the ZntA and Δ231-ZntA dimer structures, which help to explain these observations. Therefore, the first four transmembrane helices in ZntA and P1B-type ATPases play an important role in maintaining the correct dimer structure.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Catálise/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/metabolismo , Cinética , Metais/farmacologia , Modelos Moleculares , Mutagênese Sítio-Dirigida , Organismos Geneticamente Modificados , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas/genética , Estrutura Secundária de Proteína/fisiologia , Relação Estrutura-Atividade
6.
Nat Chem ; 12(11): 1081-1088, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32839601

RESUMO

Peptides that contain ß-amino acids display stable secondary structures, such as helices and sheets, and are often referred to as foldamers. Cyclic ß2,3-amino acids (cßAAs), such as 2-aminocyclohexanecarboxylic acid (2-ACHC), are strong helix/turn inducers due to their restricted conformations. Here we report the ribosomal synthesis of foldamer peptides that contain multiple, up to ten, consecutive cßAAs via genetic code reprogramming. We also report the de novo discovery of macrocyclic cßAA-containing peptides capable of binding to a protein target. As a demonstration, potent binders with low-to-subnanomolar KD values were identified for human factor XIIa (hFXIIa) and interferon-gamma receptor 1, from a library of their 1012 members. One of the anti-hFXIIa macrocyclic peptides that exhibited a high inhibitory activity and serum stability was co-crystallized with hFXIIa. The X-ray structure revealed that it adopts an antiparallel ß-sheet structure induced by a (1S,2S)-2-ACHC residue via the formation of two γ-turns. This work demonstrates the potential of this platform to explore the previously inaccessible sequence space of cßAA-containing peptides.


Assuntos
Aminoácidos/química , Peptídeos/química , Estrutura Secundária de Proteína/fisiologia , Aminas , Sequência de Aminoácidos , Cristalografia por Raios X , Ligação de Hidrogênio , Conformação Proteica , Dobramento de Proteína
7.
J Struct Biol ; 212(1): 107605, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32805410

RESUMO

BCP1 is a protein enriched in the nucleus that is required for Mss4 nuclear export and identified as the chaperone of ribosomal protein Rpl23 in Saccharomyces cerevisiae. According to sequence homology, BCP1 is related to the mammalian BRCA2-interacting protein BCCIP and belongs to the BCIP protein family (PF13862) in the Pfam database. However, the BCIP family has no discernible similarity to proteins with known structure. Here, we report the crystal structure of BCP1, presenting an α/ß fold in which the central antiparallel ß-sheet is flanked by helices. Protein structural classification revealed that BCP1 has similarity to the GNAT superfamily but no conserved substrate-binding residues. Further modeling and protein-protein docking work provide a plausible model to explain the interaction between BCP1 and Rpl23. Our structural analysis presents the first structure of BCIP family and provides a foundation for understanding the molecular basis of BCP1 as a chaperone of Rpl23 for ribosome biosynthesis.


Assuntos
Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Sítios de Ligação/fisiologia , Cristalografia por Raios X/métodos , Conformação Proteica em Folha beta/fisiologia , Estrutura Secundária de Proteína/fisiologia , Proteínas Ribossômicas/química , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo
8.
Sci Rep ; 10(1): 5120, 2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32198463

RESUMO

More than 20 unique diseases such as diabetes, Alzheimer's disease, Parkinson's disease are caused by the abnormal aggregations of pathogenic proteins such as amylin, ß-amyloid (Aß), and α-synuclein. All pathogenic proteins differ from each other in biological function, primary sequences, and morphologies; however, the proteins are toxic when aggregated. Here, we investigated the cellular toxicity of pathogenic or non-pathogenic protein aggregates. In this study, six proteins were selected and they were incubated at acid pH and high temperature. The aggregation kinetic and cellular toxicity of protein species with time were characterized. Three non-pathogenic proteins, bovine serum albumin (BSA), catalase, and pepsin at pH 2 and 65 °C were stable in protein structure and non-toxic at a lower concentration of 1 mg/mL. They formed aggregates at a higher concentration of 20 mg/mL with time and they induced the toxicity in short incubation time points, 10 min and 20 min only and they became non-toxic after 30 min. Other three pathogenic proteins, lysozyme, superoxide dismutase (SOD), and insulin, also produced the aggregates with time and they caused cytotoxicity at both 1 mg/mL and 20 mg/mL after 10 min. TEM images and DSC analysis demonstrated that fibrils or aggregates at 1 mg/mL induced cellular toxicity due to low thermal stability. In DSC data, fibrils or aggregates of pathogenic proteins had low thermal transition compared to fresh samples. The results provide useful information to understand the aggregation and cellular toxicity of pathogenic and non-pathogenic proteins.


Assuntos
Catalase/metabolismo , Insulina/metabolismo , Muramidase/metabolismo , Pepsina A/metabolismo , Agregados Proteicos/fisiologia , Agregação Patológica de Proteínas/patologia , Soroalbumina Bovina/metabolismo , Superóxido Dismutase/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Linhagem Celular , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Modelos Moleculares , Doença de Parkinson/genética , Doença de Parkinson/patologia , Estrutura Secundária de Proteína/fisiologia , alfa-Sinucleína/metabolismo
9.
Biochemistry ; 59(4): 364-378, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31895552

RESUMO

In aqueous solutions, the 214-residue low-complexity domain of the FUS protein (FUS-LC) is known to undergo liquid-liquid phase separation and also to self-assemble into amyloid-like fibrils. In previous work based on solid state nuclear magnetic resonance (ssNMR) methods, a structural model for the FUS-LC fibril core was developed, showing that residues 39-95 form the fibril core. Unlike fibrils formed by amyloid-ß peptides, α-synuclein, and other amyloid-forming proteins, the FUS-LC core is largely devoid of purely hydrophobic amino acid side chains. Instead, the core-forming segment contains numerous hydroxyl-bearing residues, including 18 serines, six threonines, and eight tyrosines, suggesting that the FUS-LC fibril structure may be stabilized in part by inter-residue hydrogen bonds among side chain hydroxyl groups. Here we describe ssNMR measurements, performed on 2H,15N,13C-labeled FUS-LC fibrils, that provide new information about the interactions of hydroxyl-bearing residues with one another and with water. The ssNMR data support the involvement of specific serine, threonine, and tyrosine residues in hydrogen-bonding interactions. The data also reveal differences in hydrogen exchange rates with water for different side chain hydroxyl groups, providing information about solvent exposure and penetration of water into the FUS-LC fibril core.


Assuntos
Proteína FUS de Ligação a RNA/química , Proteína FUS de Ligação a RNA/ultraestrutura , Sequência de Aminoácidos/genética , Amiloide/química , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas/metabolismo , Humanos , Hidrogênio/metabolismo , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Espectroscopia de Ressonância Magnética/métodos , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular/métodos , Conformação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína/fisiologia , Proteína FUS de Ligação a RNA/genética
11.
Biochim Biophys Acta Biomembr ; 1861(3): 677-684, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30615859

RESUMO

Abundant attention has focused on synaptotagmin's C2 domains, but less is known about the structure and function of its other regions. Here, we synthesized the N-acetylated, C-end amidated and Cys-palmitated peptide (VLTCCFCICK KCLFKKKNKK K) which includes the fatty acylated cysteine residues in the membrane-affiliated domain of synaptotagmin-1. Fourier-transform infrared spectrometry indicated that this peptide's conformation is influenced by environmental polarity. In artificial bilayer membranes, this peptide exhibited abundant ß-structure. Electron microscopy revealed that this peptide also promoted the stacking of liposome membranes. Together these results suggest that the fatty acylated region of synaptotagmin-1 is likely to adopt ß-structure in biological membranes. This preference for ß-structure versus α-helix has functional implications for the role of synaptotagmin-1 in synaptic vesicle exocytosis.


Assuntos
Ácidos Graxos/química , Ácidos Graxos/metabolismo , Sinaptotagmina I/química , Sinaptotagmina I/fisiologia , Acilação , Exocitose/fisiologia , Humanos , Lipossomos/química , Lipossomos/metabolismo , Espectrometria de Massas , Fusão de Membrana , Domínios Proteicos , Estrutura Secundária de Proteína/fisiologia , Espectroscopia de Infravermelho com Transformada de Fourier , Relação Estrutura-Atividade , Transmissão Sináptica , Sinaptotagmina I/metabolismo
12.
Biochim Biophys Acta Biomembr ; 1861(3): 565-572, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30550881

RESUMO

A short sequence on the gp41 envelope protein of HIV-1 is integral to infection by the virus. Without this sequence, termed the fusion peptide (FP), the virus is far less effective at fusing with the cellular membrane. One of the interesting features of the isolated FP is that it transitions between an α-helical conformation and a ß-sheet conformation in lipid bilayer membranes as a function of lipid composition and concentration, and the transition correlates with fusion. To better understand how the conformations of the FP impact lipid bilayer membranes, a variant of the FP that does not strongly promote fusion, termed gp41rk, was studied. Circular dichroism spectroscopy, dynamic light scattering, small-angle neutron scattering (SANS) and neutron spin echo spectroscopy (NSE) were used to relate the conformation of gp41rk to the structure and mechanical properties of lipid bilayer membrane vesicles composed of a 7:3 molar ratio mixture of 1,2-dimyristoyl-sn-glycero-3-phosphocholine and 1,2-dimyristoyl-sn-glycero-3-phospho-(1'-rac-glycerol). At a peptide-to-lipid ratio (P/L) of 1/200, it adopts an α-helical conformation, while gp41rk is a ß-sheet at a P/L of 1/50 in the unilamellar vesicles. SANS reveals that the lipid bilayer membrane becomes thicker when gp41rk adopts a ß-sheet conformation, which indicates that the high-concentration state of the peptide increases the order of the lipid acyl chains. At the same time, NSE demonstrates that the bilayer becomes more rigid, demonstrating that the ß-sheet conformation, which correlates with fusion for the native FP sequence, stiffens the bilayer. The results have implications for the function of the FP.


Assuntos
Fenômenos Biomecânicos/fisiologia , Proteína gp41 do Envelope de HIV/química , Bicamadas Lipídicas/química , Fusão de Membrana/efeitos dos fármacos , Sequência de Aminoácidos , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Dicroísmo Circular , Difusão Dinâmica da Luz , Proteína gp41 do Envelope de HIV/farmacologia , HIV-1/fisiologia , Humanos , Bicamadas Lipídicas/metabolismo , Espectroscopia de Ressonância Magnética , Fluidez de Membrana/efeitos dos fármacos , Fusão de Membrana/genética , Modelos Moleculares , Peptídeos/química , Peptídeos/farmacologia , Estrutura Secundária de Proteína/fisiologia , Relação Estrutura-Atividade
13.
ACS Chem Neurosci ; 10(1): 563-572, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30346704

RESUMO

The abnormal aggregation of amyloid ß-protein (Aß) is considered central in the pathogenesis of Alzheimer's disease. We focused on membrane-mediated amyloidogenesis and found that amyloid fibrils formed on monosialoganglioside GM1 clusters were more toxic than those formed in aqueous solution. In this study, we investigated the structure of the toxic fibrils by Aß-(1-40) in detail in comparison with less-toxic fibrils formed in aqueous solution. The less-toxic fibrils contain in-resister parallel ß-sheets, whereas the structure of the toxic fibrils is unknown. Atomic force microscopy revealed that the toxic fibrils had a flat, tape-like morphology composed of a single ß-sheet layer. Isotope-edited infrared spectroscopy indicated that almost the entire sequence of Aß is included in the ß-sheet. Chemical cross-linking experiments using Cys-substituted Aßs suggested that the fibrils mainly contained both in-resister parallel and two-residue-shifted antiparallel ß-sheet structures. Solid-state NMR experiments also supported this conclusion. Thus, the toxic fibrils were found to possess a novel unique structure.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas/metabolismo , Gangliosídeo G(M1)/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/química , Amiloide/metabolismo , Peptídeos beta-Amiloides/química , Amiloidose/metabolismo , Gangliosídeo G(M1)/química , Humanos , Estrutura Secundária de Proteína/fisiologia
14.
ACS Chem Neurosci ; 9(6): 1469-1476, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29601177

RESUMO

α-Synuclein (aS) forms toxic intermediates ranging from small oligomers and protofibrils to large amyloid fibrils. Understanding the time course of aS fibril formation and the role played by its regions is critical for therapeutic intervention. Here, we used pulsed hydrogen-deuterium exchange and mass spectrometry (HDX-MS) for the first time to probe kinetic intermediates of the full aS aggregation in vitro, achieving kinetic snapshots containing spatially resolved protein information about critical stages. Monitoring the resultant mass shifts shows distinct binomial abundances for two main exchange profiles: one that represents a fast-exchanging, solvent-accessible species and another with a more protected nature. We show using a series of proteolytic peptides from the full protein that self-association is most pronounced in the non-amyloid-ß-component region and less so for either terminus. The N-terminus, however, shows a minor protected population at mid- and late times, whereas the C-terminus shows predominantly unimodal HDX, indicating that these regions are devoid of any large conformational rearrangements. Focusing on the hydrophobic core, we confirmed and modeled the different isotopic distributions and calculated their relative fractions to discern their individual contributions. The data fitting reports respective t1/2 values, which are nearly identical and do not depend on location. We followed the aggregation by complementary transmission electron microscopy to observe the morphology of aggregates and circular dichroism to assess changes in secondary structure. Our results provide a detailed picture of aS aggregation in vitro and demonstrate that HDX-MS offers unique spatially resolved, coexisting kinetic intermediates in solution. This new platform is suitable for testing promising inhibitors of aS aggregation.


Assuntos
Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Doença de Parkinson/metabolismo , Peptídeos/metabolismo , alfa-Sinucleína/metabolismo , Amiloide/química , Amiloide/metabolismo , Medição da Troca de Deutério/métodos , Humanos , Hidrogênio/química , Hidrogênio/metabolismo , Espectrometria de Massas/métodos , Conformação Proteica , Estrutura Secundária de Proteína/fisiologia , alfa-Sinucleína/química
15.
Biochim Biophys Acta Biomembr ; 1859(2): 135-145, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27814978

RESUMO

Apolipoprotein B (apoB) is a large amphipathic protein that is the structural scaffold for the formation of several classes of lipoproteins involved in lipid transport throughout the body. The goal of the present study was to identify specific domains in the apoB sequence that contribute to its lipid binding properties. A sequence analysis algorithm was developed to identify stretches of hydrophobic amino acids devoid of charged amino acids, which are referred to as hydrophobic cluster domains (HCDs). This analysis identified 78 HCDs in apoB with hydrophobic stretches ranging from 6 to 26 residues. Each HCD was analyzed in silico for secondary structure and lipid binding properties, and a subset was synthesized for experimental evaluation. One HCD peptide, B38, showed high affinity binding to both isolated HDL and LDL, and could exchange between lipoproteins. All-atom molecular dynamics simulations indicate that B38 inserts 3.7Å below the phosphate plane of the bilayer. B38 forms an unusual α-helix with a broad hydrophobic face and polar serine and threonine residues on the opposite face. Based on this structure, we hypothesized that B38 could efflux cholesterol from cells. B38 showed a 12-fold greater activity than the 5A peptide, a bihelical Class A amphipathic helix (EC50 of 0.2658 vs. 3.188µM; p<0.0001), in promoting cholesterol efflux from ABCA1 expressing BHK-1 cells. In conclusion, we have identified novel domains within apoB that contribute to its lipid biding properties. Additionally, we have discovered a unique amphipathic helix design for efficient ABCA1-specific cholesterol efflux.


Assuntos
Apolipoproteínas B/química , Apolipoproteínas B/metabolismo , Lipídeos/química , Estrutura Secundária de Proteína/fisiologia , Transportador 1 de Cassete de Ligação de ATP/química , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Sítios de Ligação/fisiologia , Células Cultivadas , HDL-Colesterol/química , HDL-Colesterol/metabolismo , LDL-Colesterol/química , LDL-Colesterol/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica/fisiologia
16.
Sci Rep ; 5: 12129, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26159651

RESUMO

Environmental and occupational inhalants may induce a large number of pulmonary diseases, with asbestos exposure being the most risky. The mechanisms are clearly related to chemical composition and physical and surface properties of materials. A combination of X-ray fluorescence (µXRF) and Fourier Transform InfraRed (µFTIR) microscopy was used to chemically characterize and compare asbestos bodies versus environmental particulates (anthracosis) in lung tissues from asbestos exposed and control patients. µXRF analyses revealed heterogeneously aggregated particles in the anthracotic structures, containing mainly Si, K, Al and Fe. Both asbestos and particulates alter lung iron homeostasis, with a more marked effect in asbestos exposure. µFTIR analyses revealed abundant proteins on asbestos bodies but not on anthracotic particles. Most importantly, the analyses demonstrated that the asbestos coating proteins contain high levels of ß-sheet structures. The occurrence of conformational changes in the proteic component of the asbestos coating provides new insights into long-term asbestos effects.


Assuntos
Amianto/efeitos adversos , Asbestose/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dobramento de Proteína , Estrutura Secundária de Proteína/fisiologia
17.
PLoS Pathog ; 10(12): e1004534, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25502789

RESUMO

Apolipoprotein B (ApoB) and ApoE have been shown to participate in the particle formation and the tissue tropism of hepatitis C virus (HCV), but their precise roles remain uncertain. Here we show that amphipathic α-helices in the apolipoproteins participate in the HCV particle formation by using zinc finger nucleases-mediated apolipoprotein B (ApoB) and/or ApoE gene knockout Huh7 cells. Although Huh7 cells deficient in either ApoB or ApoE gene exhibited slight reduction of particles formation, knockout of both ApoB and ApoE genes in Huh7 (DKO) cells severely impaired the formation of infectious HCV particles, suggesting that ApoB and ApoE have redundant roles in the formation of infectious HCV particles. cDNA microarray analyses revealed that ApoB and ApoE are dominantly expressed in Huh7 cells, in contrast to the high level expression of all of the exchangeable apolipoproteins, including ApoA1, ApoA2, ApoC1, ApoC2 and ApoC3 in human liver tissues. The exogenous expression of not only ApoE, but also other exchangeable apolipoproteins rescued the infectious particle formation of HCV in DKO cells. In addition, expression of these apolipoproteins facilitated the formation of infectious particles of genotype 1b and 3a chimeric viruses. Furthermore, expression of amphipathic α-helices in the exchangeable apolipoproteins facilitated the particle formation in DKO cells through an interaction with viral particles. These results suggest that amphipathic α-helices in the exchangeable apolipoproteins play crucial roles in the infectious particle formation of HCV and provide clues to the understanding of life cycle of HCV and the development of novel anti-HCV therapeutics targeting for viral assembly.


Assuntos
Apolipoproteínas B/química , Apolipoproteínas B/fisiologia , Apolipoproteínas E/química , Apolipoproteínas E/fisiologia , Hepacivirus/patogenicidade , Estrutura Secundária de Proteína/fisiologia , Vírion/patogenicidade , Apolipoproteínas A/fisiologia , Apolipoproteínas B/genética , Apolipoproteínas C/fisiologia , Apolipoproteínas E/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Hepacivirus/fisiologia , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , RNA Interferente Pequeno/farmacologia , Vírion/fisiologia , Replicação Viral/fisiologia
18.
Sci Rep ; 4: 5224, 2014 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-24909411

RESUMO

The megakaryoblastic leukemia 1 (MKL1) protein functions as a transcriptional coactivator of the serum response factor. MKL1 has three RPEL motifs (RPEL1, RPEL2, and RPEL3) in its N-terminal region. MKL1 binds to monomeric G-actin through RPEL motifs, and the dissociation of MKL1 from G-actin promotes the translocation of MKL1 to the nucleus. Although structural data are available for RPEL motifs of MKL1 in complex with G-actin, the structural characteristics of RPEL motifs in the free state have been poorly defined. Here we characterized the structures of free RPEL motifs using NMR and CD spectroscopy. NMR and CD measurements showed that free RPEL motifs are largely unstructured in solution. However, NMR analysis identified transient α-helices in the regions where helices α1 and α2 are induced upon binding to G-actin. Proline mutagenesis showed that the transient α-helices are locally formed without helix-helix interactions. The helix content is higher in the order of RPEL1, RPEL2, and RPEL3. The amount of preformed structure may correlate with the binding affinity between the intrinsically disordered protein and its target molecule.


Assuntos
Estrutura Secundária de Proteína/fisiologia , Fator de Resposta Sérica/química , Transativadores/química , Actinas/química , Actinas/metabolismo , Motivos de Aminoácidos/fisiologia , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Ligação Proteica/fisiologia , Fator de Resposta Sérica/metabolismo , Transativadores/metabolismo
19.
J Mol Biol ; 425(18): 3389-402, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-23810904

RESUMO

Gram-negative bacteria possess an outer membrane envelope consisting of an outer leaflet of lipopolysaccharides, also called endotoxins, which protect the pathogen from antimicrobial peptides and have multifaceted roles in virulence. Lipopolysaccharide consists of a glycan moiety attached to lipid A, embedded in the outer membrane. Modification of the lipid A headgroups by phosphoethanolamine (PEA) or 4-amino-arabinose residues increases resistance to the cationic cyclic polypeptide antibiotic, polymyxin. Lipid A PEA transferases are members of the YhjW/YjdB/YijP superfamily and usually consist of a transmembrane domain anchoring the enzyme to the periplasmic face of the cytoplasmic membrane attached to a soluble catalytic domain. The crystal structure of the soluble domain of the protein of the lipid A PEA transferase from Neisseria meningitidis has been determined crystallographically and refined to 1.4Å resolution. The structure reveals a core hydrolase fold similar to that of alkaline phosphatase. Loop regions in the structure differ, presumably to enable interaction with the membrane-localized substrates and to provide substrate specificity. A phosphorylated form of the putative nucleophile, Thr280, is observed. Metal ions present in the active site are coordinated to Thr280 and to residues conserved among the family of transferases. The structure reveals the protein components needed for the transferase chemistry; however, substrate-binding regions are not evident and are likely to reside in the transmembrane domain of the protein.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Etanolaminofosfotransferase/química , Neisseria meningitidis/enzimologia , Polimixinas/farmacologia , Sítios de Ligação , Etanolaminofosfotransferase/genética , Etanolaminofosfotransferase/metabolismo , Etanolaminas/metabolismo , Lipídeo A/metabolismo , Lipopolissacarídeos/metabolismo , Modelos Biológicos , Modelos Moleculares , Neisseria meningitidis/efeitos dos fármacos , Neisseria meningitidis/genética , Domínios e Motivos de Interação entre Proteínas/genética , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína/fisiologia
20.
Acta Pharmacol Sin ; 34(8): 1084-92, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23685951

RESUMO

AIM: Vasodilator-stimulated phosphoprotein (VASP) expression is upregulated in human cancers and correlates with more invasive advanced tumor stages. The aim of this study was to elucidate the mechanisms by which matrine, an alkaloid derived from Sophora species plants, acted on the VASP protein in human gastric cancer cells in vitro. METHODS: VASP was expressed and purified. Intrinsic fluorescence spectroscopy was used to study the binding of matrine to VASP. CD spectroscopy was used to examine the changes in the VASP protein secondary structure. Human gastric carcinoma cell line BGC823 was tested. Scratch wound and cell adhesion assays were used to detect the cell migration and adhesion, respectively. Real-time PCR and Western blotting assays were used to measure mRNA and protein expression of VASP. RESULTS: In the fluorescence assay, the dissociation constant for binding of matrine to VASP protein was 0.86 mmol/L, thus the direct binding between the two molecules was weak. However, matrine (50 µg/mL) caused obvious change in the secondary structure of VASP protein shown in CD spectrum. Treatments of BGC823 cells with matrine (50 µg/mL) significantly inhibited the cell migration and adhesion. The alkaloid changed the subcellular distribution of VASP and formation of actin stress fibers in BGC823 cells. The alkaloid caused small but statistically significant decreases in VASP protein expression and phosphorylation, but had no significant effect on VASP mRNA expression. CONCLUSION: Matrine modulates the structure, subcellular distribution, expression and phosphorylation of VASP in human gastric cancer cells, thus inhibiting the cancer cell adhesion and migration.


Assuntos
Alcaloides/farmacologia , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/fisiologia , Inibição de Migração Celular/efeitos dos fármacos , Inibição de Migração Celular/fisiologia , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/fisiologia , Fosfoproteínas/química , Fosfoproteínas/fisiologia , Quinolizinas/farmacologia , Neoplasias Gástricas/metabolismo , Alcaloides/uso terapêutico , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Moléculas de Adesão Celular/antagonistas & inibidores , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Humanos , Proteínas dos Microfilamentos/antagonistas & inibidores , Fosfoproteínas/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína/efeitos dos fármacos , Estrutura Secundária de Proteína/fisiologia , Quinolizinas/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Matrinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA