Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
2.
Nature ; 552(7683): 101-105, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29186116

RESUMO

Malaria is among the most serious infectious diseases affecting humans, accounting for approximately half a million deaths each year. Plasmodium falciparum causes most life-threatening cases of malaria. Acquired immunity to malaria is inefficient, even after repeated exposure to P. falciparum, but the immune regulatory mechanisms used by P. falciparum remain largely unknown. Here we show that P. falciparum uses immune inhibitory receptors to achieve immune evasion. RIFIN proteins are products of a polymorphic multigene family comprising approximately 150-200 genes per parasite genome that are expressed on the surface of infected erythrocytes. We found that a subset of RIFINs binds to either leucocyte immunoglobulin-like receptor B1 (LILRB1) or leucocyte-associated immunoglobulin-like receptor 1 (LAIR1). LILRB1-binding RIFINs inhibit activation of LILRB1-expressing B cells and natural killer (NK) cells. Furthermore, P. falciparum-infected erythrocytes isolated from patients with severe malaria were more likely to interact with LILRB1 than erythrocytes from patients with non-severe malaria, although an extended study with larger sample sizes is required to confirm this finding. Our results suggest that P. falciparum has acquired multiple RIFINs to evade the host immune system by targeting immune inhibitory receptors.


Assuntos
Evasão da Resposta Imune/imunologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/imunologia , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Receptores Imunológicos/imunologia , Sequência de Aminoácidos , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células CHO , Cricetulus , Eritrócitos/imunologia , Eritrócitos/parasitologia , Células HEK293 , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/química , Ligantes , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Receptores Imunológicos/química , Tamanho da Amostra
3.
J Struct Biol ; 201(3): 247-251, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29170031

RESUMO

Several gene fusion technologies have been successfully applied to label particular subunits or domains within macromolecular complexes to enable positional mapping of electron microscopy (EM) density maps, but exogenous fusion of a protein domain into the target polypeptide can cause unwanted structural and functional outcomes. Fab fragments from antibodies can be used as labeling reagents during EM visualization without gene manipulation of the target protein, but this method requires a panel of high-affinity antibodies that recognize a wide variety of epitopes. Linear peptide tags and their anti-tag antibodies can be used but they have a limited mapping ability as their placement is usually limited to the terminal regions of a protein. The PA dodecapeptide epitope tag (GVAMPGAEDDVV), forms a tight ß-turn in the antigen binding pocket of its antibody (NZ-1). This capability allows for insertion of the PA tag into various surface-exposed loops within a multi-domain cell adhesion receptor, αIIbß3 integrin. We confirmed that the purified PA-tagged integrin ectodomain fragments can form a stable complex with NZ-1 Fab. Negative stain EM of the various integrin-NZ-1 complexes revealed that a majority of the particles exhibited a clear density corresponding to the NZ-1 Fab; and the positions of the bound Fab were in good agreement with the predicted location of the inserted PA tag. The high-affinity and insertion-compatibility of the PA tag system allowed us to develop a new EM labeling methodology applicable to proteins for which good antibodies are not available.


Assuntos
Epitopos/química , Integrinas/química , Microscopia Eletrônica/métodos , Proteínas/química , Epitopos/metabolismo , Fragmentos Fab das Imunoglobulinas/química , Substâncias Macromoleculares/química , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/imunologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Domínios Proteicos
4.
J Biol Chem ; 292(7): 2785-2794, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28035004

RESUMO

The human MutT homolog 1 (hMTH1, human NUDT1) hydrolyzes oxidatively damaged nucleoside triphosphates and is the main enzyme responsible for nucleotide sanitization. hMTH1 recently has received attention as an anticancer target because hMTH1 blockade leads to accumulation of oxidized nucleotides in the cell, resulting in mutations and death of cancer cells. Unlike Escherichia coli MutT, which shows high substrate specificity for 8-oxoguanine nucleotides, hMTH1 has broad substrate specificity for oxidized nucleotides, including 8-oxo-dGTP and 2-oxo-dATP. However, the reason for this broad substrate specificity remains unclear. Here, we determined crystal structures of hMTH1 in complex with 8-oxo-dGTP or 2-oxo-dATP at neutral pH. These structures based on high quality data showed that the base moieties of two substrates are located on the similar but not the same position in the substrate binding pocket and adopt a different hydrogen-bonding pattern, and both triphosphate moieties bind to the hMTH1 Nudix motif (i.e. the hydrolase motif) similarly and align for the hydrolysis reaction. We also performed kinetic assays on the substrate-binding Asp-120 mutants (D120N and D120A), and determined their crystal structures in complex with the substrates. Analyses of bond lengths with high-resolution X-ray data and the relationship between the structure and enzymatic activity revealed that hMTH1 recognizes the different oxidized nucleotides via an exchange of the protonation state at two neighboring aspartate residues (Asp-119 and Asp-120) in its substrate binding pocket. To our knowledge, this mechanism of broad substrate recognition by enzymes has not been reported previously and may have relevance for anticancer drug development strategies targeting hMTH1.


Assuntos
Enzimas Reparadoras do DNA/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Cristalografia por Raios X , Enzimas Reparadoras do DNA/química , Enzimas Reparadoras do DNA/genética , Humanos , Cinética , Mutação , Monoéster Fosfórico Hidrolases/química , Monoéster Fosfórico Hidrolases/genética , Conformação Proteica , Especificidade por Substrato
5.
J Cell Sci ; 129(7): 1512-22, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26872787

RESUMO

Placement of a tag sequence is usually limited to either terminal end of the target protein, reducing the potential of epitope tags for various labeling applications. The PA tag is a dodecapeptide (GVAMPGAEDDVV) that is recognized by a high-affinity antibody NZ-1. We determined the crystal structure of the PA-tag-NZ-1 complex and found that NZ-1 recognizes a central segment of the PA tag peptide in a tight ß-turn configuration, suggesting that it is compatible with the insertion into a loop. This possibility was tested and confirmed using multiple integrin subunits and semaphorin. More specifically, the PA tag can be inserted at multiple locations within the integrin αIIb subunit (encoded by ITGA2B) of the fibrinogen receptor αIIbß3 integrin (of which the ß3 subunit is encoded by ITGB3) without affecting the structural and functional integrity, while maintaining its high affinity for NZ-1. The large choice of the sites for 'epitope grafting' enabled the placement of the PA tag at a location whose accessibility is modulated during the biological action of the receptor. Thus, we succeeded in converting a general anti-tag antibody into a special anti-integrin antibody that can be classified as a ligand-induced binding site antibody.


Assuntos
Anticorpos Monoclonais/imunologia , Sítios de Ligação de Anticorpos/imunologia , Integrina alfa2/metabolismo , Integrina beta3/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Integrina alfa2/genética , Integrina beta3/genética , Glicoproteínas de Membrana/imunologia , Conformação Proteica , Semaforinas/genética , Semaforinas/metabolismo
6.
J Biol Chem ; 288(26): 18696-706, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23658014

RESUMO

Chitinase C from Ralstonia sp. A-471 (Ra-ChiC) has a catalytic domain sequence similar to goose-type (G-type) lysozymes and, unlike other chitinases, belongs to glycohydrolase (GH) family 23. Using NMR spectroscopy, however, Ra-ChiC was found to interact only with the chitin dimer but not with the peptidoglycan fragment. Here we report the crystal structures of wild-type, E141Q, and E162Q of the catalytic domain of Ra-ChiC with or without chitin oligosaccharides. Ra-ChiC has a substrate-binding site including a tunnel-shaped cavity, which determines the substrate specificity. Mutation analyses based on this structural information indicated that a highly conserved Glu-141 acts as a catalytic acid, and that Asp-226 located at the roof of the tunnel activates a water molecule as a catalytic base. The unique arrangement of the catalytic residues makes a clear contrast to the other GH23 members and also to inverting GH19 chitinases.


Assuntos
Proteínas de Bactérias/química , Quitina/química , Quitinases/química , Glicosídeo Hidrolases/química , Ralstonia/enzimologia , Sequência de Aminoácidos , Animais , Domínio Catalítico , Cristalografia por Raios X , Gadus morhua , Hidrólise , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Proteínas de Plantas , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
7.
Structure ; 32(5): 536-549.e5, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38460519

RESUMO

Overexpression of human epidermal growth factor receptor 2 (HER2) in breast and gastric cancers is associated with a poor prognosis, making it an important therapeutic target. Here, we establish a novel cancer-specific anti-HER2 antibody, H2Mab-214. H2Mab-214 reacts with HER2 on cancer cells, but unlike the therapeutic antibody trastuzumab, it does not react with HER2 on normal cells in flow cytometry measurements. A crystal structure suggests that H2Mab-214 recognizes a structurally disrupted region in the HER2 domain IV, which normally forms a ß-sheet. We show that this misfolding is inducible by site-directed mutagenesis mimicking the disulfide bond defects that also may occur in cancer cells, indicating that the local misfolding in the Cys-rich domain IV governs the cancer-specificity of H2Mab-214. Furthermore, we show that H2Mab-214 effectively suppresses tumor growth in xenograft mouse models. Our findings offer a potential strategy for developing cancer-specific therapeutic antibodies that target partially misfolded cell surface receptors.


Assuntos
Dobramento de Proteína , Receptor ErbB-2 , Ensaios Antitumorais Modelo de Xenoenxerto , Receptor ErbB-2/metabolismo , Receptor ErbB-2/química , Humanos , Animais , Camundongos , Linhagem Celular Tumoral , Modelos Moleculares , Feminino , Cristalografia por Raios X , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/química , Trastuzumab/farmacologia , Trastuzumab/química , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/tratamento farmacológico
8.
J Synchrotron Radiat ; 20(Pt 6): 884-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24121333

RESUMO

The saccharification process is essential for bioethanol production from woody biomass including celluloses. Cold-adapted cellulase, which has sufficient activity at low temperature (<293 K), is capable of reducing heating costs during the saccharification process and is suitable for simultaneous saccharification and fermentation. Endo-1,4-ß-glucanase from the earthworm Eisenia fetida (EF-EG2) belonging to glycoside hydrolase family 9 has been shown to have the highest activity at 313 K, and also retained a comparatively high activity at 283 K. The recombinant EF-EG2 was purified expressed in Pichia pastoris, and then grew needle-shaped crystals with dimensions of 0.02 × 0.02 × 1 mm. The crystals belonged to the space group P3221 with unit-cell parameters of a = b = 136 Å, c = 55.0 Å. The final model of EF-EG2, including 435 residues, two ions, seven crystallization reagents and 696 waters, was refined to a crystallographic R-factor of 14.7% (free R-factor of 16.8%) to 1.5 Å resolution. The overall structure of EF-EG2 has an (α/α)6 barrel fold which contains a putative active-site cleft and a negatively charged surface. This structural information helps us understand the catalytic and cold adaptation mechanisms of EF-EG2.


Assuntos
Celulase/química , Oligoquetos/enzimologia , Animais , Sequência de Bases , Domínio Catalítico , Cristalografia por Raios X , Primers do DNA , Reação em Cadeia da Polimerase , Conformação Proteica
9.
Nucleic Acids Res ; 39(20): 8972-83, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21768126

RESUMO

Human NUDT5 (hNUDT5) hydrolyzes various modified nucleoside diphosphates including 8-oxo-dGDP, 8-oxo-dADP and ADP-ribose (ADPR). However, the structural basis of the broad substrate specificity remains unknown. Here, we report the crystal structures of hNUDT5 complexed with 8-oxo-dGDP and 8-oxo-dADP. These structures reveal an unusually different substrate-binding mode. In particular, the positions of two phosphates (α and ß phosphates) of substrate in the 8-oxo-dGDP and 8-oxo-dADP complexes are completely inverted compared with those in the previously reported hNUDT5-ADPR complex structure. This result suggests that the nucleophilic substitution sites of the substrates involved in hydrolysis reactions differ despite the similarities in the chemical structures of the substrates and products. To clarify this hypothesis, we employed the isotope-labeling method and revealed that 8-oxo-dGDP is attacked by nucleophilic water at Pß, whereas ADPR is attacked at Pα. This observation reveals that the broad substrate specificity of hNUDT5 is achieved by a diversity of not only substrate recognition, but also hydrolysis mechanisms and leads to a novel aspect that enzymes do not always catalyze the reaction of substrates with similar chemical structures by using the chemically equivalent reaction site.


Assuntos
Pirofosfatases/química , Adenosina Difosfato Ribose/química , Biocatálise , Cristalografia por Raios X , Nucleotídeos de Desoxiguanina/química , Proteínas de Escherichia coli/química , Guanosina Monofosfato/análogos & derivados , Guanosina Monofosfato/química , Humanos , Hidrólise , Modelos Moleculares , Ligação Proteica , Especificidade por Substrato
10.
Sci Immunol ; 8(81): eadc9324, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-37000855

RESUMO

Celastrol, a bioactive molecule extracted from the Tripterygium wilfordii plant, has been shown to exhibit anti-inflammatory properties. However, its mechanism of action has not been fully elucidated. Here, we show that celastrol suppresses humoral immune responses and autoimmunity by disabling a protein complex consisting of copper metabolism MURR1 domain-containing (COMMD) 3 and COMMD8 (COMMD3/8 complex), a signaling adaptor for chemoattractant receptors. Having demonstrated the involvement of the COMMD3/8 complex in a mouse model of rheumatoid arthritis, we identified celastrol as a compound that covalently bound to and dissociated the COMMD3/8 complex. Celastrol inhibited B cell migration, reduced antibody responses, and blocked arthritis progression, recapitulating deficiency of the COMMD3/8 complex. These effects of celastrol were abolished in mice expressing a celastrol-resistant mutant of the COMMD3/8 complex. These findings establish that celastrol exerts immunosuppressive activity by targeting the COMMD3/8 complex. Our study suggests that the COMMD3/8 complex is a potentially druggable target in autoimmune diseases and points to celastrol as a lead pharmacologic candidate in this capacity.


Assuntos
Doenças Autoimunes , Imunidade Humoral , Camundongos , Animais , Autoimunidade , Triterpenos Pentacíclicos
11.
Trends Pharmacol Sci ; 43(10): 838-851, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35902282

RESUMO

Decoy receptor proteins that trick viruses to bind to them should be resistant to viral escape because viruses that require entry receptors cannot help but bind decoy receptors. Angiotensin-converting enzyme 2 (ACE2) is the major receptor for coronavirus cell entry. Recombinant soluble ACE2 was previously developed as a biologic against acute respiratory distress syndrome (ARDS) and verified to be safe in clinical studies. The emergence of COVID-19 reignited interest in soluble ACE2 as a potential broad-spectrum decoy receptor against coronaviruses. In this review, we summarize recent developments in preclinical studies using various high-affinity mutagenesis and Fc fusion approaches to achieve therapeutic efficacy of recombinant ACE2 decoy receptor against coronaviruses. We also highlight the relevance of stimulating effector immune cells through Fc-receptor engagement and the potential of using liquid aerosol delivery of ACE2 decoy receptors for defense against ACE2-utilizing coronaviruses.


Assuntos
Enzima de Conversão de Angiotensina 2 , Tratamento Farmacológico da COVID-19 , Receptores Virais , Humanos , Ligação Proteica , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/metabolismo
12.
Cell Rep Methods ; 2(10): 100301, 2022 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-36313806

RESUMO

Image reconstruction by integrating exchangeable single-molecule localization (IRIS) achieves multiplexed super-resolution imaging by high-density labeling with fast exchangeable fluorescent probes. However, previous methods to develop probes for individual targets required a great amount of time and effort. Here, we introduce a method for generating recombinant IRIS probes with a new mutagenesis strategy that can be widely applied to existing antibody sequences. Several conserved tyrosine residues at the base of complementarity-determining regions were identified as candidate sites for site-directed mutagenesis. With a high probability, mutations at candidate sites accelerated the off rate of recombinant antibody-based probes without compromising specific binding. We were able to develop IRIS probes from five monoclonal antibodies and three single-domain antibodies. We demonstrate multiplexed localization of endogenous proteins in primary neurons that visualizes small synaptic connections with high binding density. It is now practically feasible to generate fast-dissociating fluorescent probes for multitarget super-resolution imaging.


Assuntos
Corantes Fluorescentes , Proteínas , Microscopia de Fluorescência/métodos , Corantes Fluorescentes/química , Anticorpos , Fragmentos de Imunoglobulinas
13.
Structure ; 30(10): 1411-1423.e4, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-35981535

RESUMO

Signaling by single-pass transmembrane receptors often involves a formation of ligand-induced receptor dimers with particular conformation, and bivalent receptor binders can modulate receptor functions by inducing different receptor dimer conformations, although such agents are difficult to design. Here, we describe the generation of both antagonistic and agonistic receptor dimerizers toward PlexinB1 (PlxnB1), a receptor for semaphorin 4D (Sema4D), by grafting two different PlxnB1-binding peptides onto the human immunoglobulin G1 (IgG1) Fc protein. The function-modulating activity of a peptide Fc was strongly dependent on the type of the peptide as well as the grafting site, with the best variants showing activity at an nM concentration range. Structural analysis of each peptide-PlxnB1 complex revealed that the agonistic Fc dimerizes PlxnB1 in a face-to-face fashion similar to that induced by Sema4D, whereas antagonistic Fc would induce signaling-incompetent PlxnB1 dimer conformation, enforcing the idea that plexin activation is primarily controlled by the receptor orientation within the dimer.


Assuntos
Receptores de Superfície Celular , Semaforinas , Proteínas Ativadoras de GTPase , Humanos , Imunoglobulina G , Ligantes , Peptídeos , Receptores de Superfície Celular/metabolismo , Receptores Fc , Semaforinas/genética , Semaforinas/metabolismo
14.
Sci Transl Med ; 14(650): eabn7737, 2022 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-35471044

RESUMO

The Omicron (B.1.1.529) SARS-CoV-2 variant contains an unusually high number of mutations in the spike protein, raising concerns of escape from vaccines, convalescent serum, and therapeutic drugs. Here, we analyzed the degree to which Omicron pseudo-virus evades neutralization by serum or therapeutic antibodies. Serum samples obtained 3 months after two doses of BNT162b2 vaccination exhibited 18-fold lower neutralization titers against Omicron than parental virus. Convalescent serum samples from individuals infected with the Alpha and Delta variants allowed similar frequencies of Omicron breakthrough infections. Domain-wise analysis using chimeric spike proteins revealed that this efficient evasion was primarily achieved by mutations clustered in the receptor binding domain but that multiple mutations in the N-terminal domain contributed as well. Omicron escaped a therapeutic cocktail of imdevimab and casirivimab, whereas sotrovimab, which targets a conserved region to avoid viral mutation, remains effective. Angiotensin-converting enzyme 2 (ACE2) decoys are another virus-neutralizing drug modality that are free, at least in theory, from complete escape. Deep mutational analysis demonstrated that an engineered ACE2 molecule prevented escape for each single-residue mutation in the receptor binding domain, similar to immunized serum. Engineered ACE2 neutralized Omicron comparably to the Wuhan strain and also showed a therapeutic effect against Omicron infection in hamsters and human ACE2 transgenic mice. Similar to previous SARS-CoV-2 variants, some sarbecoviruses showed high sensitivity against engineered ACE2, confirming the therapeutic value against diverse variants, including those that are yet to emerge.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Animais , Anticorpos Monoclonais Humanizados , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Vacina BNT162 , COVID-19/terapia , Humanos , Imunização Passiva , Camundongos , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , SARS-CoV-2 , Soroterapia para COVID-19
15.
Artigo em Inglês | MEDLINE | ID: mdl-21505250

RESUMO

Chitinase from the moderately thermophilic bacterium Ralstonia sp. A-471 (Ra-ChiC) is divided into two domains: a chitin-binding domain (residues 36-80) and a catalytic domain (residues 103-252). Although the catalytic domain of Ra-ChiC has homology to goose-type lysozyme, Ra-ChiC does not show lysozyme activity but does show chitinase activity. The catalytic domain with part of an interdomain loop (Ra-ChiC(89-252)) was crystallized under several different conditions using polyethylene glycol as a precipitant. The crystals diffracted to 1.85 Šresolution and belonged to space group P6(1)22 or P6(5)22, with unit-cell parameters a = b = 100, c = 243 Å. The calculated Matthews coefficient was approximately 3.2, 2.4 or 1.9 Å(3) Da(-1) assuming the presence of three, four or five Ra-ChiC(89-252) molecules in the asymmetric unit, respectively.


Assuntos
Domínio Catalítico , Quitinases/química , Ralstonia/enzimologia , Cristalização , Cristalografia por Raios X
16.
Nat Commun ; 12(1): 4012, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34188035

RESUMO

Recognition of laminin by integrin receptors is central to the epithelial cell adhesion to basement membrane, but the structural background of this molecular interaction remained elusive. Here, we report the structures of the prototypic laminin receptor α6ß1 integrin alone and in complex with three-chain laminin-511 fragment determined via crystallography and cryo-electron microscopy, respectively. The laminin-integrin interface is made up of several binding sites located on all five subunits, with the laminin γ1 chain C-terminal portion providing focal interaction using two carboxylate anchor points to bridge metal-ion dependent adhesion site of integrin ß1 subunit and Asn189 of integrin α6 subunit. Laminin α5 chain also contributes to the affinity and specificity by making electrostatic interactions with large surface on the ß-propeller domain of α6, part of which comprises an alternatively spliced X1 region. The propeller sheet corresponding to this region shows unusually high mobility, suggesting its unique role in ligand capture.


Assuntos
Células Epiteliais/metabolismo , Integrina alfa6/metabolismo , Integrina alfa6beta1/metabolismo , Integrina beta1/metabolismo , Laminina/metabolismo , Sequência de Aminoácidos , Membrana Basal/metabolismo , Sítios de Ligação/fisiologia , Adesão Celular/fisiologia , Microscopia Crioeletrônica , Cristalografia por Raios X , Humanos , Conformação Proteica , Domínios Proteicos/fisiologia , Eletricidade Estática
17.
J Biochem ; 169(6): 701-708, 2021 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-33585933

RESUMO

Small-angle neutron scattering (SANS) and small- angle X-ray scattering (SAXS) are powerful techniques for the structural characterization of biomolecular complexes. In particular, SANS enables a selective observation of specific components in complexes by selective deuteration with contrast-matching techniques. In most cases, however, biomolecular interaction systems with heterogeneous oligomers often contain unfavorable aggregates and unbound species, hampering data interpretation. To overcome these problems, SAXS has been recently combined with size exclusion chromatography (SEC), which enables the isolation of the target complex in a multi-component system. By contrast, SEC-SANS is only at a preliminary stage. Hence, we herein perform a feasibility study of this method based on our newly developed inverse contrast-matching (iCM) SANS technique using antibody interactions as model systems. Immunoglobulin G (IgG) or its Fc fragment was mixed with 75% deuterated Fc-binding proteins, i.e. a mutated form of IgG-degrading enzyme of Streptococcus pyogenes and a soluble form of Fcγ receptor IIIb, and subjected to SEC-SANS as well as SEC-SAXS as reference. We successfully observe SANS from the non-deuterated IgG or Fc formed in complex with these binding partners, which were unobservable in terms of SANS in D2O, hence demonstrating the potential utility of the SEC-iCM-SANS approach.


Assuntos
Cromatografia em Gel/métodos , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/metabolismo , Receptores de IgG/metabolismo , Espalhamento a Baixo Ângulo , Streptococcus pyogenes/metabolismo , Difração de Raios X/métodos , Proteínas de Transporte/metabolismo , Estudos de Viabilidade , Modelos Biológicos , Streptococcus pyogenes/imunologia
18.
Nat Commun ; 12(1): 1543, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33750839

RESUMO

Protein engineering has great potential for devising multifunctional recombinant proteins to serve as next-generation protein therapeutics, but it often requires drastic modifications of the parental protein scaffolds e.g., additional domains at the N/C-terminus or replacement of a domain by another. A discovery platform system, called RaPID (Random non-standard Peptides Integrated Discovery) system, has enabled rapid discovery of small de novo macrocyclic peptides that bind a target protein with high binding specificity and affinity. Capitalizing on the optimized binding properties of the RaPID-derived peptides, here we show that RaPID-derived pharmacophore sequences can be readily implanted into surface-exposed loops on recombinant proteins and maintain both the parental peptide binding function(s) and the host protein function. We refer to this protein engineering method as lasso-grafting and demonstrate that it can endow specific binding capacity toward various receptors into a diverse set of scaffolds that includes IgG, serum albumin, and even capsid proteins of adeno-associated virus, enabling us to rapidly formulate and produce bi-, tri-, and even tetra-specific binder molecules.


Assuntos
Peptídeos/química , Peptídeos/farmacologia , Engenharia de Proteínas/métodos , Proteínas do Capsídeo/química , Proteínas de Transporte/química , Linhagem Celular , Dependovirus , Humanos , Imunoglobulina G/química , Modelos Moleculares , Albumina Sérica/química , Bibliotecas de Moléculas Pequenas
19.
Nat Commun ; 12(1): 3802, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-34155214

RESUMO

SARS-CoV-2 has mutated during the global pandemic leading to viral adaptation to medications and vaccinations. Here we describe an engineered human virus receptor, ACE2, by mutagenesis and screening for binding to the receptor binding domain (RBD). Three cycles of random mutagenesis and cell sorting achieved sub-nanomolar affinity to RBD. Our structural data show that the enhanced affinity comes from better hydrophobic packing and hydrogen-bonding geometry at the interface. Additional disulfide mutations caused the fixing of a closed ACE2 conformation to avoid off-target effects of protease activity, and also improved structural stability. Our engineered ACE2 neutralized SARS-CoV-2 at a 100-fold lower concentration than wild type; we also report that no escape mutants emerged in the co-incubation after 15 passages. Therapeutic administration of engineered ACE2 protected hamsters from SARS-CoV-2 infection, decreased lung virus titers and pathology. Our results provide evidence of a therapeutic potential of engineered ACE2.


Assuntos
Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/farmacologia , Tratamento Farmacológico da COVID-19 , Mutação , SARS-CoV-2/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , COVID-19/metabolismo , COVID-19/virologia , Células Cultivadas , Cricetinae , Cristalografia por Raios X , Modelos Animais de Doenças , Humanos , Masculino , Simulação de Dinâmica Molecular , Ligação Proteica , Engenharia de Proteínas/métodos , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/metabolismo
20.
J Biochem ; 170(3): 379-387, 2021 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-34185078

RESUMO

Advanced glycation end-products (AGEs) are a heterogeneous group of compounds formed by non-enzymatic reaction between reducing-sugar and Arg/Lys in proteins and are involved in various diabetic complications. GA-pyridine is derived from glycolaldehyde and is one of the most cytotoxic AGEs. Here, we established a single-chain Fv (scFv) antibody against GA-pyridine, 73MuL9-scFv, and examined the details of its specificity and antigen recognition by using various techniques involving biophysics, chemical biology and structural biology. We also synthesized several compounds that differ slightly in regard to the position and number of GA-pyridine substituent groups, and revealed that GA-pyridine was specifically bound to 73MuL9-scFv. Thermodynamic analysis revealed that the association of GA-pyridine to 73MuL9-scFv was an exothermic and enthalpy driven reaction, and thus that the antigen recognition involved multiple specific interactions. Crystallographic analysis of the Fv fragment of 73MuL9-scFv revealed that several CH-π and hydrogen bond interactions took place between the Fv-fragment and GA-pyridine, which was consistent with the results of thermodynamic analysis. Further studies using 73MuL9-scFv as a tool to clarify the relevance of GA-pyridine to diabetic complications are warranted.


Assuntos
Produtos Finais de Glicação Avançada/imunologia , Piridinas/imunologia , Anticorpos de Cadeia Única/metabolismo , Acetaldeído/análogos & derivados , Acetaldeído/química , Acetaldeído/imunologia , Sequência de Aminoácidos , Antígenos/química , Antígenos/metabolismo , Biofísica , Cristalografia/métodos , Produtos Finais de Glicação Avançada/química , Humanos , Ligação de Hidrogênio , Piridinas/química , Anticorpos de Cadeia Única/química , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA