Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
J Biol Chem ; 299(9): 104927, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37330175

RESUMO

Methicillin-resistant Staphylococcus aureus, or MRSA, is one of the major causative agents of hospital-acquired infections worldwide. Novel antimicrobial strategies efficient against antibiotic-resistant strains are necessary and not only against S. aureus. Among those, strategies that aim at blocking or dismantling proteins involved in the acquisition of essential nutrients, helping the bacteria to colonize the host, are intensively studied. A major route for S. aureus to acquire iron from the host organism is the Isd (iron surface determinant) system. In particular, the hemoglobin receptors IsdH and IsdB located on the surface of the bacterium are necessary to acquire the heme moiety containing iron, making them a plausible antibacterial target. Herein, we obtained an antibody of camelid origin that blocked heme acquisition. We determined that the antibody recognized the heme-binding pocket of both IsdH and IsdB with nanomolar order affinity through its second and third complementary-determining regions. The mechanism explaining the inhibition of acquisition of heme in vitro could be described as a competitive process in which the complementary-determining region 3 from the antibody blocked the acquisition of heme by the bacterial receptor. Moreover, this antibody markedly reduced the growth of three different pathogenic strains of MRSA. Collectively, our results highlight a mechanism for inhibiting nutrient uptake as an antibacterial strategy against MRSA.


Assuntos
Anticorpos Antibacterianos , Staphylococcus aureus Resistente à Meticilina , Receptores de Superfície Celular , Anticorpos de Domínio Único , Humanos , Antibacterianos/farmacologia , Heme/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/uso terapêutico , Anticorpos de Domínio Único/biossíntese , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Anticorpos de Domínio Único/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Antígenos de Bactérias/imunologia , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Camelídeos Americanos , Animais , Ligação Proteica/efeitos dos fármacos , Modelos Moleculares , Simulação de Dinâmica Molecular
2.
J Biol Chem ; 299(10): 105254, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37716701

RESUMO

Listeriosis, caused by infection with Listeria monocytogenes, is a severe disease with a high mortality rate. The L. monocytogenes virulence factor, internalin family protein InlA, which binds to the host receptor E-cadherin, is necessary to invade host cells. Here, we isolated two single-domain antibodies (VHHs) that bind to InlA with picomolar affinities from an alpaca immune library using the phage display method. These InlA-specific VHHs inhibited the binding of InlA to the extracellular domains of E-cadherin in vitro as shown by biophysical interaction analysis. Furthermore, we determined that the VHHs inhibited the invasion of L. monocytogenes into host cells in culture. High-resolution X-ray structure analyses of the complexes of VHHs with InlA revealed that the VHHs bind to the same binding site as E-cadherin against InlA. We conclude that these VHHs have the potential for use as drugs to treat listeriosis.

3.
Biochem Biophys Res Commun ; 714: 149969, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38657446

RESUMO

CD40 is a member of the tumor necrosis factor receptor superfamily, and it is widely expressed on immune and non-immune cell types. The interaction between CD40 and the CD40 ligand (CD40L) plays an essential function in signaling, and the CD40/CD40L complex works as an immune checkpoint molecule. CD40 has become a therapeutic target, and a variety of agonistic/antagonistic anti-CD40 monoclonal antibodies (mAbs) have been developed. To better understand the mode of action of anti-CD40 mAbs, we determined the X-ray crystal structures of dacetuzumab (agonist) and bleselumab (antagonist) in complex with the extracellular domain of human CD40, respectively. The structure reveals that dacetuzumab binds to CD40 on the top of cysteine-rich domain 1 (CRD1), which is the domain most distant from the cell surface, and it does not compete with CD40L binding. The binding interface of bleselumab spread between CRD2 and CRD1, overlapping with the binding surface of the ligand. Our results offer important insights for future structural and functional studies of CD40 and provide clues to understanding the mechanism of biological response. These data can be applied to developing new strategies for designing antibodies with more therapeutic efficacy.


Assuntos
Anticorpos Monoclonais Humanizados , Antígenos CD40 , Humanos , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Sítios de Ligação , Antígenos CD40/química , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Ligante de CD40/química , Ligante de CD40/metabolismo , Ligante de CD40/imunologia , Cristalografia por Raios X , Modelos Moleculares , Ligação Proteica , Conformação Proteica
4.
J Biol Chem ; 298(6): 101995, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35500652

RESUMO

Staphylococcus aureus is a major cause of deadly nosocomial infections, a severe problem fueled by the steady increase of resistant bacteria. The iron surface determinant (Isd) system is a family of proteins that acquire nutritional iron from the host organism, helping the bacterium to proliferate during infection, and therefore represents a promising antibacterial target. In particular, the surface protein IsdH captures hemoglobin (Hb) and acquires the heme moiety containing the iron atom. Structurally, IsdH comprises three distinctive NEAr-iron Transporter (NEAT) domains connected by linker domains. The objective of this study was to characterize the linker region between NEAT2 and NEAT3 from various biophysical viewpoints and thereby advance our understanding of its role in the molecular mechanism of heme extraction. We demonstrate the linker region contributes to the stability of the bound protein, likely influencing the flexibility and orientation of the NEAT3 domain in its interaction with Hb, but only exerts a modest contribution to the affinity of IsdH for heme. Based on these data, we suggest that the flexible nature of the linker facilitates the precise positioning of NEAT3 to acquire heme. In addition, we also found that residues His45 and His89 of Hb located in the heme transfer route toward IsdH do not play a critical role in the transfer rate-determining step. In conclusion, this study clarifies key elements of the mechanism of heme extraction of human Hb by IsdH, providing key insights into the Isd system and other protein systems containing NEAT domains.


Assuntos
Antígenos de Bactérias , Heme , Ferro , Receptores de Superfície Celular , Staphylococcus aureus , Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Heme/metabolismo , Hemoglobinas/química , Humanos , Ferro/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Ligação Proteica , Domínios Proteicos , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo
5.
J Biol Chem ; 298(6): 101962, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35452676

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a disease associated with dysregulation of the immune complement system, especially of the alternative pathway (AP). Complement factor H (CFH), consisting of 20 domains called complement control protein (CCP1-20), downregulates the AP as a cofactor for mediating C3 inactivation by complement factor I. However, anomalies related to CFH are known to cause excessive complement activation and cytotoxicity. In aHUS, mutations and the presence of anti-CFH autoantibodies (AAbs) have been reported as plausible causes of CFH dysfunction, and it is known that CFH-related aHUS carries a high probability of end-stage renal disease. Elucidating the detailed functions of CFH at the molecular level will help to understand aHUS pathogenesis. Herein, we used biophysical data to reveal that a heavy-chain antibody fragment, termed VHH4, recognized CFH with high affinity. Hemolytic assays also indicated that VHH4 disrupted the protective function of CFH on sheep erythrocytes. Furthermore, X-ray crystallography revealed that VHH4 recognized the Leu1181-Leu1189CCP20 loop, a known anti-CFH AAbs epitope. We next analyzed the dynamics of the C-terminal region of CFH and showed that the epitopes recognized by anti-CFH AAbs and VHH4 were the most flexible regions in CCP18-20. Finally, we conducted mutation analyses to elucidate the mechanism of VHH4 recognition of CFH and revealed that VHH4 inserts the Trp1183CCP20 residue of CFH into the pocket formed by the complementary determining region 3 loop. These results suggested that anti-CFH AAbs may adopt a similar molecular mechanism to recognize the flexible loop of Leu1181-Leu1189CCP20, leading to aHUS pathogenesis.


Assuntos
Anticorpos Monoclonais/química , Síndrome Hemolítico-Urêmica Atípica , Fator H do Complemento/química , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Autoanticorpos/imunologia , Ativação do Complemento , Epitopos , Humanos , Mutação
6.
J Cell Sci ; 134(22)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34676411

RESUMO

Diverse genes associated with familial Parkinson's disease (familial Parkinsonism) have been implicated in mitochondrial quality control. One such gene, PARK7 encodes the protein DJ-1, pathogenic mutations of which trigger its translocation from the cytosol to the mitochondrial matrix. The translocation of steady-state cytosolic proteins like DJ-1 to the mitochondrial matrix upon missense mutations is rare, and the underlying mechanism remains to be elucidated. Here, we show that the protein unfolding associated with various DJ-1 mutations drives its import into the mitochondrial matrix. Increasing the structural stability of these DJ-1 mutants restores cytosolic localization. Mechanistically, we show that a reduction in the structural stability of DJ-1 exposes a cryptic N-terminal mitochondrial-targeting signal (MTS), including Leu10, which promotes DJ-1 import into the mitochondrial matrix for subsequent degradation. Our work describes a novel cellular mechanism for targeting a destabilized cytosolic protein to the mitochondria for degradation.


Assuntos
Doença de Parkinson , Humanos , Mitocôndrias/genética , Doença de Parkinson/genética
7.
J Biol Chem ; 297(3): 101054, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34364873

RESUMO

Liver intestine (LI)-cadherin is a member of the cadherin superfamily, which encompasses a group of Ca2+-dependent cell-adhesion proteins. The expression of LI-cadherin is observed on various types of cells in the human body, such as normal small intestine and colon cells, and gastric cancer cells. Because its expression is not observed on normal gastric cells, LI-cadherin is a promising target for gastric cancer imaging. However, because the cell adhesion mechanism of LI-cadherin has remained unknown, rational design of therapeutic molecules targeting this cadherin has been hampered. Here, we have studied the homodimerization mechanism of LI-cadherin. We report the crystal structure of the LI-cadherin homodimer containing its first four extracellular cadherin repeats (EC1-4). The EC1-4 homodimer exhibited a unique architecture different from that of other cadherins reported so far, driven by the interactions between EC2 of one protein chain and EC4 of the second protein chain. The crystal structure also revealed that LI-cadherin possesses a noncanonical calcium ion-free linker between the EC2 and EC3 domains. Various biochemical techniques and molecular dynamics simulations were employed to elucidate the mechanism of homodimerization. We also showed that the formation of the homodimer observed in the crystal structure is necessary for LI-cadherin-dependent cell adhesion by performing cell aggregation assays. Taken together, our data provide structural insights necessary to advance the use of LI-cadherin as a target for imaging gastric cancer.


Assuntos
Caderinas/química , Caderinas/metabolismo , Caderinas/genética , Adesão Celular , Agregação Celular , Cristalografia por Raios X , Dimerização , Humanos , Domínios Proteicos , Estrutura Terciária de Proteína
8.
J Biol Chem ; 296: 100176, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33303630

RESUMO

Proteins are modulated by a variety of posttranslational modifications including methylation. Despite its importance, the majority of protein methylation modifications discovered by mass spectrometric analyses are functionally uncharacterized, partly owing to the difficulty in obtaining reliable methylsite-specific antibodies. To elucidate how functional methylsite-specific antibodies recognize the antigens and lead to the development of a novel method to create such antibodies, we use an immunized library paired with phage display to create rabbit monoclonal antibodies recognizing trimethylated Lys260 of MAP3K2 as a representative substrate. We isolated several methylsite-specific antibodies that contained unique complementarity determining region sequence. We characterized the mode of antigen recognition by each of these antibodies using structural and biophysical analyses, revealing the molecular details, such as binding affinity toward methylated/nonmethylated antigens and structural motif that is responsible for recognition of the methylated lysine residue, by which each antibody recognized the target antigen. In addition, the comparison with the results of Western blotting analysis suggests a critical antigen recognition mode to generate cross-reactivity to protein and peptide antigen of the antibodies. Computational simulations effectively recapitulated our biophysical data, capturing the antibodies of differing affinity and specificity. Our exhaustive characterization provides molecular architectures of functional methylsite-specific antibodies and thus should contribute to the development of a general method to generate functional methylsite-specific antibodies by de novo design.


Assuntos
Anticorpos Monoclonais/química , Antígenos/química , Fragmentos Fab das Imunoglobulinas/química , Lisina/química , MAP Quinase Quinase Quinase 2/química , Peptídeos/química , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/isolamento & purificação , Afinidade de Anticorpos , Especificidade de Anticorpos , Antígenos/genética , Antígenos/imunologia , Sítios de Ligação , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/genética , Regiões Determinantes de Complementaridade/imunologia , Reações Cruzadas , Cristalografia por Raios X , Humanos , Fragmentos Fab das Imunoglobulinas/biossíntese , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Cinética , Lisina/imunologia , MAP Quinase Quinase Quinase 2/genética , MAP Quinase Quinase Quinase 2/imunologia , Metilação , Simulação de Dinâmica Molecular , Biblioteca de Peptídeos , Peptídeos/genética , Peptídeos/imunologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Coelhos
9.
Biochem Biophys Res Commun ; 596: 22-28, 2022 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-35108650

RESUMO

AL amyloidosis is a life-threatening disease characterized by the deposition of amyloidogenic immunoglobulin light chain secreted from clonal plasma cells. Here we established an in-vitro screening system of amyloid inhibition of a variable domain in λ6 light chain mutant (Vλ6), Wil, and screened a food-additive compound library to identify compounds inhibiting the fibril formation. We found gossypetin and isoquercitrin as novel inhibitors. NMR analysis showed that both compounds directly interacted with natively-folded Wil, and proteolysis experiments demonstrated that these compounds conferred proteolytic resistance, suggesting that the compounds enhance the kinetic stability of Wil. Since gossypetin and isoquercitrin specifically interacted with the protein at micromolar concentrations, these compounds could be used as lead to further develop inhibitors against AL amyloidosis.


Assuntos
Amiloide/antagonistas & inibidores , Flavonoides/farmacologia , Amiloidose de Cadeia Leve de Imunoglobulina/metabolismo , Cadeias lambda de Imunoglobulina/metabolismo , Quercetina/análogos & derivados , Amiloide/genética , Amiloide/metabolismo , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Catequina/análogos & derivados , Catequina/metabolismo , Catequina/farmacologia , Relação Dose-Resposta a Droga , Flavonoides/química , Humanos , Amiloidose de Cadeia Leve de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/química , Cadeias lambda de Imunoglobulina/genética , Cinética , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Mutação , Ligação Proteica , Estabilidade Proteica/efeitos dos fármacos , Quercetina/química , Quercetina/farmacologia , Fatores de Tempo
10.
Int J Mol Sci ; 23(18)2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-36142694

RESUMO

Antibody engagement with the membrane-proximal external region (MPER) of the envelope glycoprotein (Env) of HIV-1 constitutes a distinctive molecular recognition phenomenon, the full appreciation of which is crucial for understanding the mechanisms that underlie the broad neutralization of the virus. Recognition of the HIV-1 Env antigen seems to depend on two specific features developed by antibodies with MPER specificity: (i) a large cavity at the antigen-binding site that holds the epitope amphipathic helix; and (ii) a membrane-accommodating Fab surface that engages with viral phospholipids. Thus, besides the main Fab-peptide interaction, molecular recognition of MPER depends on semi-specific (electrostatic and hydrophobic) interactions with membranes and, reportedly, on specific binding to the phospholipid head groups. Here, based on available cryo-EM structures of Fab-Env complexes of the anti-MPER antibody 10E8, we sought to delineate the functional antibody-membrane interface using as the defining criterion the neutralization potency and binding affinity improvements induced by Arg substitutions. This rational, Arg-based mutagenesis strategy revealed the position-dependent contribution of electrostatic interactions upon inclusion of Arg-s at the CDR1, CDR2 or FR3 of the Fab light chain. Moreover, the contribution of the most effective Arg-s increased the potency enhancement induced by inclusion of a hydrophobic-at-interface Phe at position 100c of the heavy chain CDR3. In combination, the potency and affinity improvements by Arg residues delineated a protein-membrane interaction site, whose surface and position support a possible mechanism of action for 10E8-induced neutralization. Functional delineation of membrane-interacting patches could open new lines of research to optimize antibodies of therapeutic interest that target integral membrane epitopes.


Assuntos
HIV-1 , Anticorpos Neutralizantes , Epitopos , Glicoproteínas , Anticorpos Anti-HIV , Proteína gp41 do Envelope de HIV/química , HIV-1/metabolismo , Peptídeos , Fosfolipídeos
11.
Biochem Biophys Res Commun ; 558: 114-119, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33915325

RESUMO

The CH2 domain is a critical element of the human Immunoglobulin G (IgG) constant region. Although the CH2 domain is the least stable domain in IgG, it is also a promising scaffold candidate for developing novel therapeutic approaches. Recently, we succeeded in preparing glycosylated and non-glycosylated CH2 domain in the host organism Pichia pastoris. Herein, we verified that glycosylation of the CH2 domain decreased both, its tendency to aggregate and its immunogenicity in mice, suggesting that aggregation and immunogenicity are related. In addition, we have produced in P. pastoris a stabilized version of the CH2 domain with and without glycan, and their propensity to aggregate evaluated. We found that stabilization alone significantly decreased the aggregation of the CH2 domain. Moreover, the combination of glycosylation and stabilization completely suppressed its aggregation behavior. Since protein aggregation is related to immunogenicity, the combination of glycosylation and stabilization to eliminate the aggregation behavior of a protein could be a fruitful strategy to generate promising immunoglobulin scaffolds.


Assuntos
Imunoglobulina G/química , Imunoglobulina G/metabolismo , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Fenômenos Biofísicos , Feminino , Glicosilação , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Agregados Proteicos/genética , Domínios Proteicos , Engenharia de Proteínas , Estabilidade Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomycetales/genética , Saccharomycetales/metabolismo
12.
Nat Chem Biol ; 15(3): 250-258, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30643284

RESUMO

Irreversible inhibition of disease-associated proteins with small molecules is a powerful approach for achieving increased and sustained pharmacological potency. Here, we introduce α-chlorofluoroacetamide (CFA) as a novel warhead of targeted covalent inhibitor (TCI). Despite weak intrinsic reactivity, CFA-appended quinazoline showed high reactivity toward Cys797 of epidermal growth factor receptor (EGFR). In cells, CFA-quinazoline showed higher target specificity for EGFR than the corresponding Michael acceptors in a wide concentration range (0.1-10 µM). The cysteine adduct of the CFA derivative was susceptible to hydrolysis and reversibly yielded intact thiol but was stable in solvent-sequestered ATP-binding pocket of EGFR. This environment-dependent hydrolysis can potentially reduce off-target protein modification by CFA-based drugs. Oral administration of CFA quinazoline NS-062 significantly suppressed tumor growth in a mouse xenograft model. Further, CFA-appended pyrazolopyrimidine irreversibly inhibited Bruton's tyrosine kinase with higher target specificity. These results demonstrate the utility of CFA as a new class warheads for TCI.


Assuntos
Acetamidas/síntese química , Cisteína/metabolismo , Quinazolinas/síntese química , Acetamidas/química , Acetamidas/farmacologia , Animais , Antineoplásicos , Linhagem Celular , Receptores ErbB , Humanos , Camundongos , Camundongos Nus , Neoplasias , Fosfotransferases/fisiologia , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/antagonistas & inibidores , Quinazolinas/química , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Int J Cancer ; 145(8): 2107-2113, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30515800

RESUMO

Oxaliplatin, which is widely used as chemotherapy for certain solid cancers, frequently causes peripheral neuropathy. Commonly described neuropathic symptoms include aberrant sensations such as mechanical allodynia (hypersensitivity to normally innocuous stimuli). Although oxaliplatin neuropathy is a dose-limiting toxicity, there are no established preventive strategies available at present. By screening several sets of small-molecule chemical libraries (more than 3,000 compounds in total) using a newly established in vitro high-throughput phenotypic assay, we identified fulvestrant, a clinically approved drug for the treatment of breast cancer in postmenopausal women, as having a protective effect on oxaliplatin-induced neuronal damage. Furthermore, histological and behavioural analyses using a rat model of oxaliplatin neuropathy demonstrated the in vivo efficacy of fulvestrant to prevent oxaliplatin-induced axonal degeneration of the sciatic nerve and mechanical allodynia. Furthermore, fulvestrant did not interfere with oxaliplatin-induced cytotoxicity against cancer cells. Thus, our findings reveal a previously unrecognised pharmacological effect of fulvestrant to prevent oxaliplatin-induced painful peripheral neuropathy without impairing its cytotoxicity against cancer cells and may represent a novel prophylactic option for patients receiving oxaliplatin chemotherapy.


Assuntos
Fulvestranto/farmacologia , Hiperalgesia/prevenção & controle , Neurônios/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/prevenção & controle , Animais , Linhagem Celular , Hibridomas , Hiperalgesia/induzido quimicamente , Masculino , Camundongos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Oxaliplatina , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Ratos Sprague-Dawley
14.
Immunol Rev ; 268(1): 201-21, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26497522

RESUMO

The number of studies and the quality of the structural data of Fcγ receptors (FcγRs) has rapidly increased in the last few years. Upon critical examination of the literature, we have extracted general conclusions that could explain differences in affinity and selectivity of FcγRs for immunoglobulin G (IgG) based on structural considerations. FcγRs employ a little conserved asymmetric surface of domain D2 composed of two distinct subsites to recognize the well-conserved lower hinge region of IgG1-Fc. The extent of the contact interface with the antibody in subsite 1 of the receptor (but not in subsite 2), the geometrical complementarity between antibody and receptor, and the number of polar interactions contribute decisively toward strengthening the binding affinity of the antibody for the receptor. In addition, the uncertain role of the N-linked glycan of IgG for the binding and effector responses elicited by FcγRs is discussed. The available data suggest that not only the non-covalent interactions between IgG and FcγRs but also their dynamic features are essential for the immune response elicited through these receptors. We believe that the integration of structural, thermodynamic, and kinetic data will be critical for the design and validation of the next generation of therapeutic antibodies with enhanced effector capabilities.


Assuntos
Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Modelos Moleculares , Conformação Proteica , Receptores de IgG/química , Sequência de Aminoácidos , Aminoácidos/química , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Afinidade de Anticorpos/imunologia , Sítios de Ligação , Variação Genética , Glicosilação , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Polissacarídeos/metabolismo , Ligação Proteica , Engenharia de Proteínas , Domínios e Motivos de Interação entre Proteínas , Receptores de IgG/classificação , Receptores de IgG/genética , Receptores de IgG/metabolismo , Homologia de Sequência de Aminoácidos
15.
Biochemistry ; 57(28): 4177-4185, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-29936828

RESUMO

Protein tyrosine sulfation (PTS) is a post-translational modification regulating numerous biological events. PTS generally occurs at flexible regions of proteins, enhancing intermolecular interactions between proteins. Because of the high flexibility associated with the regions where PTS is generally encountered, an atomic-level understanding has been difficult to achieve by X-ray crystallography or nuclear magnetic resonance techniques. In this study, we focused on the conformational behavior of a flexible sulfated peptide and its interaction with an antibody. Molecular dynamics simulations and thermodynamic analysis indicated that PTS reduced the main-chain fluctuations upon the appearance of sulfate-mediated intramolecular H-bonds. Collectively, our data suggested that one of the mechanisms by which PTS may enhance protein-protein interactions consists of the limitation of conformational dynamics in the unbound state, thus reducing the loss of entropy upon binding and boosting the affinity for its partner.


Assuntos
Anticorpos/metabolismo , Peptídeos/metabolismo , Tirosina/análogos & derivados , Anticorpos/química , Cristalografia por Raios X , Simulação de Dinâmica Molecular , Peptídeos/química , Ligação Proteica , Mapas de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Termodinâmica , Tirosina/química , Tirosina/metabolismo
16.
J Biol Chem ; 292(13): 5571-5583, 2017 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-28213514

RESUMO

The 4E10 antibody displays an extreme breadth of HIV-1 neutralization and therefore constitutes a suitable model system for structure-guided vaccine design and immunotherapeutics against AIDS. In this regard, the relevance of autoreactivity with membrane lipids for the biological function of this antibody is still a subject of controversy. To address this dispute, herein we have compared the membrane partitioning ability of the 4E10 antibody and several of its variants, which were mutated at the region of the paratope surface in contact with the membrane interface. We first employed a physical separation approach (vesicle flotation) and subsequently carried out quantitative fluorescence measurements in an intact system (spectroscopic titration), using 4E10 Fab labeled with a polarity-sensitive fluorescent probe. Moreover, recognition of epitope peptide in membrane was demonstrated by photo-cross-linking assays using a Fab that incorporated the genetically encoded unnatural amino acid p-benzoylphenylalanine. The experimental data ruled out that the proposed stereospecific recognition of viral lipids was necessary for the function of the antibody. In contrast, our data suggest that nonspecific electrostatic interactions between basic residues of 4E10 and acidic phospholipids in the membranes contribute to the observed biological function. Moreover, the energetics of membrane partitioning indicated that 4E10 behaves as a peripheral membrane protein, tightening the binding to the ligand epitope inserted in the viral membrane. The implications of these findings for the natural production and biological function of this antibody are discussed.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Membrana Celular/imunologia , Anticorpos Anti-HIV/imunologia , Vacinas contra a AIDS , Anticorpos Monoclonais/efeitos adversos , Anticorpos Neutralizantes/efeitos adversos , Autoimunidade , Anticorpos Amplamente Neutralizantes , Linhagem Celular , Desenho de Fármacos , Epitopos , Anticorpos Anti-HIV/efeitos adversos , Humanos , Lipídeos de Membrana/imunologia , Proteínas da Matriz Viral/imunologia
18.
Int Immunol ; 29(7): 311-317, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28655198

RESUMO

Antibodies of the IgG class carry a pair of oligosaccharides (N-glycans) in the Fc region. The importance of the N-glycan is clearly demonstrated by its profound effect in the physicochemical and biological properties of antibodies. The term 'glycoengineering' has been coined to describe contemporary strategies to improve the performance of therapeutic monoclonal antibodies on the basis of modifications in the structure and composition of the N-glycan. These methodologies have resulted in the approval and commercialization of a new generation of antibodies with improved therapeutic efficacy. So far, these advances have been driven by herculean efforts in a process of trial-and-error. The collective work of researchers in this field is progressively revealing the molecular basis of N-glycans for the function of antibodies. This knowledge will ultimately be conducive to the application of rational approaches for the successful manipulation of antibodies using glycoengineering strategies. Herein, we review advances in our understanding of the role of the N-glycan in the structural and dynamic integrity, and biological activity, of antibodies. Since the N-glycan has a multifaceted effect in antibodies, in this review we have emphasized the importance of integrating various techniques that address this problem from multiple points of view. In particular, the combination of X-ray crystallography with nuclear magnetic resonance, molecular dynamics simulations and biophysical approaches based on thermodynamic principles, has emerged as a powerful combination that is deepened our understanding of this unique system with critical implications for human well-being.


Assuntos
Anticorpos Monoclonais/genética , Imunoglobulina G/metabolismo , Polissacarídeos/metabolismo , Animais , Cristalografia por Raios X , Glicosilação , Humanos , Imunoglobulina G/química , Imunoglobulina G/imunologia , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Conformação Proteica
19.
J Biol Chem ; 291(37): 19210-19219, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27445331

RESUMO

Pore-forming toxins (PFTs) are cytolytic proteins belonging to the molecular warfare apparatus of living organisms. The assembly of the functional transmembrane pore requires several intermediate steps ranging from a water-soluble monomeric species to the multimeric ensemble inserted in the cell membrane. The non-lytic oligomeric intermediate known as prepore plays an essential role in the mechanism of insertion of the class of ß-PFTs. However, in the class of α-PFTs, like the actinoporins produced by sea anemones, evidence of membrane-bound prepores is still lacking. We have employed single-particle cryo-electron microscopy (cryo-EM) and atomic force microscopy to identify, for the first time, a prepore species of the actinoporin fragaceatoxin C bound to lipid vesicles. The size of the prepore coincides with that of the functional pore, except for the transmembrane region, which is absent in the prepore. Biochemical assays indicated that, in the prepore species, the N terminus is not inserted in the bilayer but is exposed to the aqueous solution. Our study reveals the structure of the prepore in actinoporins and highlights the role of structural intermediates for the formation of cytolytic pores by an α-PFT.


Assuntos
Venenos de Cnidários/química , Membranas Artificiais , Proteínas Citotóxicas Formadoras de Poros/química , Anêmonas-do-Mar/química , Animais , Microscopia Crioeletrônica , Microscopia de Força Atômica
20.
Biochemistry ; 55(34): 4836-49, 2016 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-27490825

RESUMO

Heme oxygenase (HO) is a ubiquitous enzyme with key roles in inflammation, cell signaling, heme disposal, and iron acquisition. HO catalyzes the oxidative conversion of heme to biliverdin (BV) using a conserved histidine to coordinate the iron atom of bound heme. This His-heme interaction has been regarded as being essential for enzyme activity, because His-to-Ala mutants fail to convert heme to biliverdin in vitro. We probed a panel of proximal His mutants of cyanobacterial, human, and plant HO enzymes using a live-cell activity assay based on heterologous co-expression in Escherichia coli of each HO mutant and a fluorescent biliverdin biosensor. In contrast to in vitro studies with purified proteins, we observed that multiple HO mutants retained significant activity within the intracellular environment of bacteria. X-ray crystallographic structures of human HO1 H25R with bound heme and additional functional studies suggest that HO mutant activity inside these cells does not involve heme ligation by a proximal amino acid. Our study reveals unexpected plasticity in the active site binding interactions with heme that can support HO activity within cells, suggests important contributions by the surrounding active site environment to HO catalysis, and can guide efforts to understand the evolution and divergence of HO function.


Assuntos
Heme Oxigenase (Desciclizante)/química , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1/química , Heme Oxigenase-1/metabolismo , Substituição de Aminoácidos , Arabidopsis/enzimologia , Arabidopsis/genética , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biliverdina/metabolismo , Biocatálise , Catalase/metabolismo , Domínio Catalítico/genética , Cristalografia por Raios X , Escherichia coli/enzimologia , Escherichia coli/genética , Heme/metabolismo , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1/genética , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Conformação Proteica , Synechocystis/enzimologia , Synechocystis/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA