Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33903234

RESUMO

Amyloid fibril formation of α-synuclein (αS) is associated with multiple neurodegenerative diseases, including Parkinson's disease (PD). Growing evidence suggests that progression of PD is linked to cell-to-cell propagation of αS fibrils, which leads to seeding of endogenous intrinsically disordered monomer via templated elongation and secondary nucleation. A molecular understanding of the seeding mechanism and driving interactions is crucial to inhibit progression of amyloid formation. Here, using relaxation-based solution NMR experiments designed to probe large complexes, we probe weak interactions of intrinsically disordered acetylated-αS (Ac-αS) monomers with seeding-competent Ac-αS fibrils and seeding-incompetent off-pathway oligomers to identify Ac-αS monomer residues at the binding interface. Under conditions that favor fibril elongation, we determine that the first 11 N-terminal residues on the monomer form a common binding site for both fibrils and off-pathway oligomers. Additionally, the presence of off-pathway oligomers within a fibril seeding environment suppresses seeded amyloid formation, as observed through thioflavin-T fluorescence experiments. This highlights that off-pathway αS oligomers can act as an auto-inhibitor against αS fibril elongation. Based on these data taken together with previous results, we propose a model in which Ac-αS monomer recruitment to the fibril is driven by interactions between the intrinsically disordered monomer N terminus and the intrinsically disordered flanking regions (IDR) on the fibril surface. We suggest that this monomer recruitment may play a role in the elongation of amyloid fibrils and highlight the potential of the IDRs of the fibril as important therapeutic targets against seeded amyloid formation.


Assuntos
Amiloide/ultraestrutura , Proteínas Intrinsicamente Desordenadas/ultraestrutura , Doença de Parkinson/genética , alfa-Sinucleína/ultraestrutura , Amiloide/química , Amiloide/genética , Benzotiazóis/química , Benzotiazóis/metabolismo , Sítios de Ligação/genética , Humanos , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Ressonância Magnética Nuclear Biomolecular , Doença de Parkinson/patologia , alfa-Sinucleína/química , alfa-Sinucleína/genética
2.
Proteins ; 90(5): 1044-1053, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34375467

RESUMO

Since the identification of the SARS-CoV-2 virus as the causative agent of the current COVID-19 pandemic, considerable effort has been spent characterizing the interaction between the Spike protein receptor-binding domain (RBD) and the human angiotensin converting enzyme 2 (ACE2) receptor. This has provided a detailed picture of the end point structure of the RBD-ACE2 binding event, but what remains to be elucidated is the conformation and dynamics of the RBD prior to its interaction with ACE2. In this work, we utilize molecular dynamics simulations to probe the flexibility and conformational ensemble of the unbound state of the receptor-binding domain from SARS-CoV-2 and SARS-CoV. We have found that the unbound RBD has a localized region of dynamic flexibility in Loop 3 and that mutations identified during the COVID-19 pandemic in Loop 3 do not affect this flexibility. We use a loop-modeling protocol to generate and simulate novel conformations of the CoV2-RBD Loop 3 region that sample conformational space beyond the ACE2 bound crystal structure. This has allowed for the identification of interesting substates of the unbound RBD that are lower energy than the ACE2-bound conformation, and that block key residues along the ACE2 binding interface. These novel unbound substates may represent new targets for therapeutic design.


Assuntos
COVID-19 , Glicoproteína da Espícula de Coronavírus , Enzima de Conversão de Angiotensina 2 , Sítios de Ligação , Humanos , Simulação de Dinâmica Molecular , Pandemias , Ligação Proteica , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química
3.
Proteins ; 90(5): 1054-1080, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34580920

RESUMO

Understanding the molecular evolution of the SARS-CoV-2 virus as it continues to spread in communities around the globe is important for mitigation and future pandemic preparedness. Three-dimensional structures of SARS-CoV-2 proteins and those of other coronavirusess archived in the Protein Data Bank were used to analyze viral proteome evolution during the first 6 months of the COVID-19 pandemic. Analyses of spatial locations, chemical properties, and structural and energetic impacts of the observed amino acid changes in >48 000 viral isolates revealed how each one of 29 viral proteins have undergone amino acid changes. Catalytic residues in active sites and binding residues in protein-protein interfaces showed modest, but significant, numbers of substitutions, highlighting the mutational robustness of the viral proteome. Energetics calculations showed that the impact of substitutions on the thermodynamic stability of the proteome follows a universal bi-Gaussian distribution. Detailed results are presented for potential drug discovery targets and the four structural proteins that comprise the virion, highlighting substitutions with the potential to impact protein structure, enzyme activity, and protein-protein and protein-nucleic acid interfaces. Characterizing the evolution of the virus in three dimensions provides testable insights into viral protein function and should aid in structure-based drug discovery efforts as well as the prospective identification of amino acid substitutions with potential for drug resistance.


Assuntos
COVID-19 , Pandemias , Aminoácidos , Humanos , Estudos Prospectivos , Proteoma , SARS-CoV-2 , Proteínas Virais/genética , Proteínas Virais/metabolismo
4.
Proc Natl Acad Sci U S A ; 114(49): 12946-12951, 2017 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29158386

RESUMO

The influenza M2 protein not only forms a proton channel but also mediates membrane scission in a cholesterol-dependent manner to cause virus budding and release. The atomic interaction of cholesterol with M2, as with most eukaryotic membrane proteins, has long been elusive. We have now determined the cholesterol-binding site of the M2 protein in phospholipid bilayers using solid-state NMR spectroscopy. Chain-fluorinated cholesterol was used to measure cholesterol proximity to M2 while sterol-deuterated cholesterol was used to measure bound-cholesterol orientation in lipid bilayers. Carbon-fluorine distance measurements show that at a cholesterol concentration of 17 mol%, two cholesterol molecules bind each M2 tetramer. Cholesterol binds the C-terminal transmembrane (TM) residues, near an amphipathic helix, without requiring a cholesterol recognition sequence motif. Deuterium NMR spectra indicate that bound cholesterol is approximately parallel to the bilayer normal, with the rough face of the sterol rings apposed to methyl-rich TM residues. The distance- and orientation-restrained cholesterol-binding site structure shows that cholesterol is stabilized by hydrophobic interactions with the TM helix and polar and aromatic interactions with neighboring amphipathic helices. At the 1:2 binding stoichiometry, lipid 31P spectra show an isotropic peak indicative of high membrane curvature. This M2-cholesterol complex structure, together with previously observed M2 localization at phase boundaries, suggests that cholesterol mediates M2 clustering to the neck of the budding virus to cause the necessary curvature for membrane scission. The solid-state NMR approach developed here is generally applicable for elucidating the structural basis of cholesterol's effects on membrane protein function.


Assuntos
Colesterol/química , Bicamadas Lipídicas/química , Proteínas da Matriz Viral/química , Sítios de Ligação , Vírus da Influenza A/ultraestrutura , Simulação de Acoplamento Molecular , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica em alfa-Hélice , Domínios Proteicos
5.
Proteomics ; 18(21-22): e1800109, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30142698

RESUMO

Several intrinsically disordered proteins have been implicated in the process of amyloid fibril formation in neurodegenerative disease, and developing approaches to inhibit the aggregation of these intrinsically disordered proteins is critical for establishing effective therapies against disease progression. The aggregation pathway of the intrinsically disordered protein alpha-synuclein, which is implicated in several neurodegenerative diseases known as synucleinopathies, has been extensively characterized. Less attention has been leveraged on beta-synuclein, a homologous intrinsically disordered protein that co-localizes with alpha-synuclein and is known to delay alpha-synuclein fibril formation. In this review, we focus on beta-synuclein and the molecular-level interactions between alpha-synuclein and beta-synuclein that underlie the delay of fibril formation. We highlight studies that begin to define alpha-synuclein and beta-synuclein interactions at the monomer, oligomer, and surface levels, and suggest that beta-synuclein plays a role in regulation of inhibition at many different stages of alpha-synuclein aggregation.


Assuntos
Proteínas Intrinsicamente Desordenadas/metabolismo , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , Animais , Ligação Proteica
6.
J Biol Chem ; 292(43): 17876-17884, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-28893910

RESUMO

The influenza A and B viruses are the primary cause of seasonal flu epidemics. Common to both viruses is the M2 protein, a homotetrameric transmembrane proton channel that acidifies the virion after endocytosis. Although influenza A M2 (AM2) and B M2 (BM2) are functional analogs, they have little sequence homology, except for a conserved HXXXW motif, which is responsible for proton selectivity and channel gating. Importantly, BM2 contains a second titratable histidine, His-27, in the tetrameric transmembrane domain that forms a reverse WXXXH motif with the gating tryptophan. To understand how His-27 affects the proton conduction property of BM2, we have used solid-state NMR to characterize the pH-dependent structure and dynamics of His-27. In cholesterol-containing lipid membranes mimicking the virus envelope, 15N NMR spectra show that the His-27 tetrad protonates with higher pKa values than His-19, indicating that the solvent-accessible His-27 facilitates proton conduction of the channel by increasing the proton dissociation rates of His-19. AM2 is inhibited by the amantadine class of antiviral drugs, whereas BM2 has no known inhibitors. We measured the N-terminal interhelical separation of the BM2 channel using fluorinated Phe-5. The interhelical 19F-19F distances show a bimodal distribution of a short distance of 7 Å and a long distance of 15-20 Å, indicating that the phenylene rings do not block small-molecule entry into the channel pore. These results give insights into the lack of amantadine inhibition of BM2 and reveal structural diversities in this family of viral proton channels.


Assuntos
Vírus da Influenza B/química , Canais Iônicos/química , Membranas Artificiais , Proteínas da Matriz Viral/química , Motivos de Aminoácidos , Vírus da Influenza B/genética , Vírus da Influenza B/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Ressonância Magnética Nuclear Biomolecular , Domínios Proteicos , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo
7.
J Am Chem Soc ; 138(26): 8143-55, 2016 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-27286559

RESUMO

Together with the influenza A virus, influenza B virus causes seasonal flu epidemics. The M2 protein of influenza B (BM2) forms a tetrameric proton-conducting channel that is important for the virus lifecycle. BM2 shares little sequence homology with AM2, except for a conserved HxxxW motif in the transmembrane (TM) domain. Unlike AM2, no antiviral drugs have been developed to block the BM2 channel. To elucidate the proton-conduction mechanism of BM2 and to facilitate the development of BM2 inhibitors, we have employed solid-state NMR spectroscopy to investigate the conformation, dynamics, and hydration of the BM2 TM domain in lipid bilayers. BM2 adopts an α-helical conformation in lipid membranes. At physiological temperature and low pH, the proton-selective residue, His19, shows relatively narrow (15)N chemical exchange peaks for the imidazole nitrogens, indicating fast proton shuttling that interconverts cationic and neutral histidines. Importantly, pH-dependent (15)N chemical shifts indicate that His19 retains the neutral population to much lower pH than His37 in AM2, indicating larger acid-dissociation constants or lower pKa's. We attribute these dynamical and equilibrium differences to the presence of a second titratable histidine, His27, which may increase the proton-dissociation rate of His19. Two-dimensional (1)H-(13)C correlation spectra probing water (1)H polarization transfer to the peptide indicates that the BM2 channel becomes much more hydrated at low pH than at high pH, particularly at Ser12, indicating that the pore-facing serine residues in BM2 mediate proton relay to the proton-selective histidine.


Assuntos
Membrana Celular/metabolismo , Vírus da Influenza B , Ressonância Magnética Nuclear Biomolecular , Prótons , Proteínas da Matriz Viral/química , Água/metabolismo , Temperatura Baixa , Histidina/metabolismo , Concentração de Íons de Hidrogênio , Modelos Moleculares , Conformação Proteica , Proteínas da Matriz Viral/metabolismo
8.
J Biomol NMR ; 61(2): 97-107, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25510834

RESUMO

The measurement of long-range distances remains a challenge in solid-state NMR structure determination of biological macromolecules. In 2D and 3D correlation spectra of uniformly (13)C-labeled biomolecules, inter-residue, inter-segmental, and intermolecular (13)C-(13)C cross peaks that provide important long-range distance constraints for three-dimensional structures often overlap with short-range cross peaks that only reflect the covalent structure of the molecule. It is therefore desirable to develop new approaches to obtain spectra containing only long-range cross peaks. Here we show that a relaxation-compensated modification of the commonly used 2D (1)H-driven spin diffusion (PDSD) experiment allows the clean detection of such long-range cross peaks. By adding a z-filter to keep the total z-period of the experiment constant, we compensate for (13)C T1 relaxation. As a result, the difference spectrum between a long- and a scaled short-mixing time spectrum show only long-range correlation signals. We show that one- and two-bond cross peaks equalize within a few tens of milliseconds. Within ~200 ms, the intensity equilibrates within an amino acid residue and a monosaccharide to a value that reflects the number of spins in the local network. With T1 relaxation compensation, at longer mixing times, inter-residue and inter-segmental cross peaks increase in intensity whereas intra-segmental cross-peak intensities remain unchanged relative to each other and can all be subtracted out. Without relaxation compensation, the difference 2D spectra exhibit both negative and positive intensities due to heterogeneous T1 relaxation in most biomolecules, which can cause peak cancellation. We demonstrate this relaxation-compensated difference PDSD approach on amino acids, monosaccharides, a crystalline model peptide, a membrane-bound peptide and a plant cell wall sample. The resulting difference spectra yield clean multi-bond, inter-residue and intermolecular correlation peaks, which are often difficult to resolve in the parent 2D spectra.


Assuntos
Espectroscopia de Ressonância Magnética Nuclear de Carbono-13/métodos , Parede Celular/química , Ressonância Magnética Nuclear Biomolecular/métodos , Proteínas de Plantas/química , Polissacarídeos/química , Arabidopsis , Isótopos de Carbono/química , Glucose/química , Glutamina/química , Histidina/química , N-Formilmetionina Leucil-Fenilalanina/química , Proteínas de Plantas/análise , Polissacarídeos/análise
9.
Solid State Nucl Magn Reson ; 72: 118-26, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26440131

RESUMO

The four aromatic amino acids in proteins, namely histidine, phenylalanine, tyrosine, and tryptophan, have strongly overlapping (13)C chemical shift ranges between 100 and 160ppm, and have so far been largely neglected in solid-state NMR determination of protein structures. Yet aromatic residues play important roles in biology through π-π and cation-π interactions. To better resolve and assign aromatic residues' (13)C signals in magic-angle-spinning (MAS) solid-state NMR spectra, we introduce two spectral editing techniques. The first method uses gated (1)H decoupling in a proton-driven spin-diffusion (PDSD) experiment to remove all protonated (13)C signals and retain only non-protonated carbon signals in the aromatic region of the (13)C spectra. The second technique uses chemical shift filters and (1)H-(13)C dipolar dephasing to selectively detect the Cα, Cß and CO cross peaks of aromatic residues while suppressing the signals of all aliphatic residues. We demonstrate these two techniques on amino acids, a model peptide, and the microcrystalline protein GB1, and show that they significantly simplify the 2D NMR spectra and both reveal and permit the ready assignment of the aromatic residues' signals.


Assuntos
Espectroscopia de Ressonância Magnética/métodos , Proteínas/química , Fenômenos Magnéticos , Modelos Moleculares , Isótopos de Nitrogênio/química , Conformação Proteica
10.
Biophys J ; 104(8): 1698-708, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23601317

RESUMO

The M2 protein of the influenza virus conducts protons into the virion under external acidic pH. The proton selectivity of the tetrameric channel is controlled by a single histidine (His(37)), whereas channel gating is accomplished by a single tryptophan (Trp(41)) in the transmembrane domain of the protein. Aromatic interaction between these two functional residues has been previously observed in Raman spectra, but atomic-resolution evidence for this interaction remains scarce. Here we use high-resolution solid-state NMR spectroscopy to determine the side-chain conformation and dynamics of Trp(41) in the M2 transmembrane peptide by measuring the Trp chemical shifts, His(37)-Trp(41) distances, and indole dynamics at high and low pH. The interatomic distances constrain the Trp41 side-chain conformation to trans for χ1 and 120-135° for χ2. This t90 rotamer points the Nε1-Cε2-Cζ2 side of the indole toward the aqueous pore. The precise χ1 and χ2 angles differ by ∼20° between high and low pH. These differences, together with the known changes in the helix tilt angle between high and low pH, push the imidazole and indole rings closer together at low pH. Moreover, the measured order parameters indicate that the indole rings undergo simultaneous χ1 and χ2 torsional fluctuations at acidic pH, but only restricted χ1 fluctuations at high pH. As a result, the Trp(41) side chain periodically experiences strong cation-π interactions with His(37) at low pH as the indole sweeps through its trajectory, whereas at high pH the indole ring is further away from the imidazole. These results provide the structural basis for understanding how the His(37)-water proton exchange rate measured by NMR is reduced to the small proton flux measured in biochemical experiments. The indole dynamics, together with the known motion of the imidazolium, indicate that this compact ion channel uses economical side-chain dynamics to regulate proton conduction and gating.


Assuntos
Ativação do Canal Iônico , Proteínas da Matriz Viral/química , Sequência de Aminoácidos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Estrutura Terciária de Proteína , Triptofano/química
11.
J Am Chem Soc ; 135(26): 9885-97, 2013 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-23758317

RESUMO

The M2 protein of influenza A viruses forms a tetrameric proton channel that is targeted by the amantadine class of antiviral drugs. A S31N mutation in the transmembrane (TM) domain of the protein has caused widespread amantadine resistance in most of the currently circulating flu viruses. Recently, a new family of compounds based on amantadine- and aryl-substituted isoxazole were discovered to inhibit the S31N channel activity and reduce replication of S31N-harboring viruses. We now use solid-state NMR spectroscopy to investigate the effects of one of these isoxazole compounds, WJ352, on the conformation of the S31N TM segment and the dynamics of the proton-selective residue, His37. Chemical shift perturbations show that WJ352 changes the conformational equilibrium of multiple TM residues, with the maximal perturbation occurring at the crucial Asn31. (13)C-(2)H distance measurements and (1)H-(1)H NOE cross peaks indicate that the adamantane moiety of the drug is bound in the spacious pore between Asn31 and Gly34 while the phenyl tail is located near Val27. Thus, the polar amine points to the channel exterior rather than to His37, in contrast to amantadine and rimantadine in the wild-type channel, suggesting that the drug is significantly stabilized by hydrophobic interactions between the adamantane and the TM peptide. (15)N and (13)C chemical shifts indicate that at low pH, His37 undergoes fast exchange among the τ tautomer, the π tautomer, and the cationic state due to proton transfer with water. The exchange rate is higher than the wild-type channel, consistent with the larger single-channel conductance of the mutant. Drug binding at acidic pH largely suppresses this exchange, reverting the histidines to a similar charge distribution as that of the high-pH closed state.


Assuntos
Amantadina/farmacologia , Canais Iônicos/antagonistas & inibidores , Isoxazóis/farmacologia , Simulação de Dinâmica Molecular , Ressonância Magnética Nuclear Biomolecular , Proteínas da Matriz Viral/antagonistas & inibidores , Amantadina/química , Canais Iônicos/química , Canais Iônicos/metabolismo , Isoxazóis/química , Modelos Moleculares , Estrutura Molecular , Mutação , Conformação Proteica/efeitos dos fármacos , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/metabolismo
12.
Protein Sci ; 32(1): e4508, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36369695

RESUMO

Fibrillar collagen-integrin interactions in the extracellular matrix (ECM) regulate a multitude of cellular processes and cell signalling. Collagen I fibrils serve as the molecular scaffolding for connective tissues throughout the human body and are the most abundant protein building blocks in the ECM. The ECM environment is diverse, made up of several ECM proteins, enzymes, and proteoglycans. In particular, glycosaminoglycans (GAGs), anionic polysaccharides that decorate proteoglycans, become depleted in the ECM with natural aging and their mis-regulation has been linked to cancers and other diseases. The impact of GAG depletion in the ECM environment on collagen I protein interactions and on mechanical properties is not well understood. Here, we integrate ELISA protein binding assays with liquid high-resolution atomic force microscopy (AFM) to assess the effects of GAG depletion on the interaction of collagen I fibrils with the integrin α2I domain using separate rat tails. ELISA binding assays demonstrate that α2I preferentially binds to GAG-depleted collagen I fibrils in comparison to native fibrils. By amplitude modulated AFM in air and in solution, we find that GAG-depleted collagen I fibrils retain structural features of the native fibrils, including their characteristic D-banding pattern, a key structural motif. AFM fast force mapping in solution shows that GAG depletion reduces the stiffness of individual fibrils, lowering the indentation modulus by half compared to native fibrils. Together these results shed new light on how GAGs influence collagen I fibril-integrin interactions and may aid in strategies to treat diseases that result from GAG mis-regulation.


Assuntos
Matriz Extracelular , Glicosaminoglicanos , Ratos , Humanos , Animais , Glicosaminoglicanos/análise , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Matriz Extracelular/química , Proteoglicanas/análise , Proteoglicanas/metabolismo , Microscopia de Força Atômica , Colágeno/química
13.
J Am Chem Soc ; 134(36): 14753-5, 2012 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-22931093

RESUMO

The influenza M2 protein conducts protons through a critical histidine (His) residue, His37. Whether His37 only interacts with water to relay protons into the virion or whether a low-barrier hydrogen bond (LBHB) also exists between the histidines to stabilize charges before proton conduction is actively debated. To address this question, we have measured the imidazole (1)H(N) chemical shifts of His37 at different temperatures and pH using 2D (15)N-(1)H correlation solid-state NMR. At low temperature, the H(N) chemical shifts are 8-15 ppm at all pH values, indicating that the His37 side chain forms conventional hydrogen bonds (H-bonds) instead of LBHBs. At ambient temperature, the dynamically averaged H(N) chemical shifts are 4.8 ppm, indicating that the H-bonding partner of the imidazole is water instead of another histidine in the tetrameric channel. These data show that His37 forms H-bonds only to water, with regular strength, thus supporting the His-water proton exchange model and ruling out the low-barrier H-bonded dimer model.


Assuntos
Histidina/química , Prótons , Proteínas da Matriz Viral/química , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Modelos Moleculares , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular
14.
bioRxiv ; 2020 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-33299989

RESUMO

Three-dimensional structures of SARS-CoV-2 and other coronaviral proteins archived in the Protein Data Bank were used to analyze viral proteome evolution during the first six months of the COVID-19 pandemic. Analyses of spatial locations, chemical properties, and structural and energetic impacts of the observed amino acid changes in >48,000 viral proteome sequences showed how each one of the 29 viral study proteins have undergone amino acid changes. Structural models computed for every unique sequence variant revealed that most substitutions map to protein surfaces and boundary layers with a minority affecting hydrophobic cores. Conservative changes were observed more frequently in cores versus boundary layers/surfaces. Active sites and protein-protein interfaces showed modest numbers of substitutions. Energetics calculations showed that the impact of substitutions on the thermodynamic stability of the proteome follows a universal bi-Gaussian distribution. Detailed results are presented for six drug discovery targets and four structural proteins comprising the virion, highlighting substitutions with the potential to impact protein structure, enzyme activity, and functional interfaces. Characterizing the evolution of the virus in three dimensions provides testable insights into viral protein function and should aid in structure-based drug discovery efforts as well as the prospective identification of amino acid substitutions with potential for drug resistance.

15.
Sci Rep ; 9(1): 17579, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31772376

RESUMO

Alpha-synuclein (αS) fibrils are toxic to cells and contribute to the pathogenesis and progression of Parkinson's disease and other synucleinopathies. ß-Synuclein (ßS), which co-localizes with αS, has been shown to provide a neuroprotective effect, but the molecular mechanism by which this occurs remains elusive. Here we show that αS fibrils formed in the presence of ßS are less cytotoxic, exhibit reduced cell seeding capacity and are more resistant to fibril shedding compared to αS fibrils alone. Using solid-state NMR, we found that the overall structure of the core of αS fibrils when co-incubated with ßS is minimally perturbed, however, the dynamics of Lys and Thr residues, located primarily in the imperfect KTKEGV repeats of the αS N-terminus, are increased. Our results suggest that amyloid fibril dynamics may play a key role in modulating toxicity and seeding. Thus, enhancing the dynamics of amyloid fibrils may be a strategy for future therapeutic targeting of neurodegenerative diseases.


Assuntos
Amiloide/metabolismo , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , Amiloide/ultraestrutura , Encéfalo/metabolismo , Linhagem Celular Tumoral , Humanos , Espectroscopia de Ressonância Magnética , Microscopia de Força Atômica , Microscopia Confocal , Microscopia de Fluorescência , Agregação Patológica de Proteínas/metabolismo
16.
Annu Rev Biophys ; 47: 201-222, 2018 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-29498890

RESUMO

Solid-state nuclear magnetic resonance (SSNMR) spectroscopy elucidates membrane protein structures and dynamics in atomic detail to yield mechanistic insights. By interrogating membrane proteins in phospholipid bilayers that closely resemble biological membranes, SSNMR spectroscopists have revealed ion conduction mechanisms, substrate transport dynamics, and oligomeric interfaces of seven-transmembrane helix proteins. Research has also identified conformational plasticity underlying virus-cell membrane fusions by complex protein machineries, and ß-sheet folding and assembly by amyloidogenic proteins bound to lipid membranes. These studies collectively show that membrane proteins exhibit extensive structural plasticity to carry out their functions. Because of the inherent dependence of NMR frequencies on molecular orientations and the sensitivity of NMR frequencies to dynamical processes on timescales from nanoseconds to seconds, SSNMR spectroscopy is ideally suited to elucidate such structural plasticity, local and global conformational dynamics, protein-lipid and protein-ligand interactions, and protonation states of polar residues. New sensitivity-enhancement techniques, resolution enhancement by ultrahigh magnetic fields, and the advent of 3D and 4D correlation NMR techniques are increasingly aiding these mechanistically important structural studies.


Assuntos
Proteínas de Membrana/química , Ressonância Magnética Nuclear Biomolecular/métodos , Humanos
17.
J Mol Biol ; 430(16): 2360-2371, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-29782835

RESUMO

The intrinsically disordered protein ß-synuclein is known to inhibit the aggregation of its intrinsically disordered homolog, α-synuclein, which is implicated in Parkinson's disease. While ß-synuclein itself does not form fibrils at the cytoplasmic pH 7.4, alteration of pH and other environmental perturbations are known to induce its fibrilization. However, the sequence and structural determinants of ß-synuclein inhibition and self-aggregation are not well understood. We have utilized a series of domain-swapped chimeras of α-synuclein and ß-synuclein to probe the relative contributions of the N-terminal, C-terminal, and the central non-amyloid-ß component domains to the inhibition of α-synuclein aggregation. Changes in the rates of α-synuclein fibril formation in the presence of the chimeras indicate that the non-amyloid-ß component domain is the primary determinant of self-association leading to fibril formation, while the N- and C-terminal domains play critical roles in the fibril inhibition process. Our data provide evidence that all three domains of ß-synuclein together contribute to providing effective inhibition, and support a model of transient, multi-pronged interactions between IDP chains in both processes. Inclusion of such multi-site inhibitory interactions spread over the length of synuclein chains may be critical for the development of therapeutics that are designed to mimic the inhibitory effects of ß-synuclein.


Assuntos
Agregação Patológica de Proteínas/metabolismo , alfa-Sinucleína/química , beta-Sinucleína/química , beta-Sinucleína/metabolismo , Sítios de Ligação , Citoplasma/química , Citoplasma/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Ligação Proteica , Espectrometria de Massas por Ionização por Electrospray
18.
Sci Rep ; 7(1): 7954, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28801573

RESUMO

Natural enzymes use local environments to tune the reactivity of amino acid side chains. In searching for small peptides with similar properties, we discovered a four-residue π-clamp motif (Phe-Cys-Pro-Phe) for regio- and chemoselective arylation of cysteine in ribosomally produced proteins. Here we report mutational, computational, and structural findings directed toward elucidating the molecular factors that drive π-clamp-mediated arylation. We show the significance of a trans conformation prolyl amide bond for the π-clamp reactivity. The π-clamp cysteine arylation reaction enthalpy of activation (ΔH‡) is significantly lower than a non-π-clamp cysteine. Solid-state NMR chemical shifts indicate the prolyl amide bond in the π-clamp motif adopts a 1:1 ratio of the cis and trans conformation, while in the reaction product Pro3 was exclusively in trans. In two structural models of the perfluoroarylated product, distinct interactions at 4.7 Å between Phe1 side chain and perfluoroaryl electrophile moiety are observed. Further, solution 19F NMR and isothermal titration calorimetry measurements suggest interactions between hydrophobic side chains in a π-clamp mutant and the perfluoroaryl probe. These studies led us to design a π-clamp mutant with an 85-fold rate enhancement. These findings will guide us toward the discovery of small reactive peptides to facilitate abiotic chemistry in water.


Assuntos
Cisteína/química , Proteínas/química , Proteínas/genética , Motivos de Aminoácidos , Calorimetria , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Mutação , Conformação Proteica , Termodinâmica
19.
J Magn Reson ; 247: 118-127, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25228502

RESUMO

Water plays an essential role in the structure and function of proteins, lipid membranes and other biological macromolecules. Solid-state NMR heteronuclear-detected (1)H polarization transfer from water to biomolecules is a versatile approach for studying water-protein, water-membrane, and water-carbohydrate interactions in biology. We review radiofrequency pulse sequences for measuring water polarization transfer to biomolecules, the mechanisms of polarization transfer, and the application of this method to various biological systems. Three polarization transfer mechanisms, chemical exchange, spin diffusion and NOE, manifest themselves at different temperatures, magic-angle-spinning frequencies, and pulse irradiations. Chemical exchange is ubiquitous in all systems examined so far, and spin diffusion plays the key role in polarization transfer within the macromolecule. Tightly bound water molecules with long residence times are rare in proteins at ambient temperature. The water polarization-transfer technique has been used to study the hydration of microcrystalline proteins, lipid membranes, and plant cell wall polysaccharides, and to derive atomic-resolution details of the kinetics and mechanism of ion conduction in channels and pumps. Using this approach, we have measured the water polarization transfer to the transmembrane domain of the influenza M2 protein to obtain information on the structure of this tetrameric proton channel. At short mixing times, the polarization transfer rates are site-specific and depend on the pH, labile protons, sidechain conformation, as well as the radial position of the residues in this four-helix bundle. Despite the multiple dependences, the initial transfer rates reflect the periodic nature of the residue positions from the water-filled pore, thus this technique provides a way of gleaning secondary structure information, helix tilt angle, and the oligomeric structure of membrane proteins.


Assuntos
Proteínas de Membrana/química , Ressonância Magnética Nuclear Biomolecular/métodos , Carboidratos/química , Hidrogênio/química , Lipídeos de Membrana/química , Prótons , Proteínas da Matriz Viral/química , Água/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA