Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 210
Filtrar
1.
Phys Chem Chem Phys ; 26(19): 14160-14170, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38712976

RESUMO

Protonated ions of fucose-containing oligosaccharides are prone to undergo internal glycan rearrangement which results in chimeric fragments that obfuscate mass-spectrometric analysis. Lack of accessible tools that would facilitate systematic analysis of glycans in the gas phase limits our understanding of this phenomenon. In this work, we use density functional theory modeling to interpret cryogenic IR spectra of Lewis a and blood group type H1 trisaccharides and to establish whether these trisaccharides undergo the rearrangement during gas-phase analysis. Structurally unconstrained search reveals that none of the parent ions constitute a thermodynamic global minimum. In contrast, predicted collision cross sections and anharmonic IR spectra provide a good match to available experimental data which allowed us to conclude that fucose migration does not occur in these antigens. By comparing the predicted structures with those obtained for Lewis x and blood group type H2 epitopes, we demonstrate that the availability of the mobile proton and a large difference in the relative stability of the parent ions and rearrangement products constitute the prerequisites for the rearrangement reaction.


Assuntos
Antígenos do Grupo Sanguíneo de Lewis , Antígenos do Grupo Sanguíneo de Lewis/química , Epitopos/química , Termodinâmica , Polissacarídeos/química , Teoria da Densidade Funcional , Antígenos de Grupos Sanguíneos/química , Espectrofotometria Infravermelho , Oligossacarídeos/química , Trissacarídeos/química
2.
Glycobiology ; 34(6)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38590172

RESUMO

Human noroviruses, globally the main cause of viral gastroenteritis, show strain specific affinity for histo-blood group antigens (HBGA) and can successfully be propagated ex vivo in human intestinal enteroids (HIEs). HIEs established from jejunal stem cells of individuals with different ABO, Lewis and secretor geno- and phenotypes, show varying susceptibility to such infections. Using bottom-up glycoproteomic approaches we have defined and compared the N-linked glycans of glycoproteins of seven jejunal HIEs. Membrane proteins were extracted, trypsin digested, and glycopeptides enriched by hydrophilic interaction liquid chromatography and analyzed by nanoLC-MS/MS. The Byonic software was used for glycopeptide identification followed by hands-on verifications and interpretations. Glycan structures and attachment sites were identified from MS2 spectra obtained by higher-energy collision dissociation through analysis of diagnostic saccharide oxonium ions (B-ions), stepwise glycosidic fragmentation of the glycans (Y-ions), and peptide sequence ions (b- and y-ions). Altogether 694 unique glycopeptides from 93 glycoproteins were identified. The N-glycans encompassed pauci- and oligomannose, hybrid- and complex-type structures. Notably, polyfucosylated HBGA-containing glycopeptides of the four glycoproteins tetraspanin-8, carcinoembryonic antigen-related cell adhesion molecule 5, sucrose-isomaltase and aminopeptidase N were especially prominent and were characterized in detail and related to donor ABO, Lewis and secretor types of each HIE. Virtually no sialylated N-glycans were identified for these glycoproteins suggesting that terminal sialylation was infrequent compared to fucosylation and HBGA biosynthesis. This approach gives unique site-specific information on the structural complexity of N-linked glycans of glycoproteins of human HIEs and provides a platform for future studies on the role of host glycoproteins in gastrointestinal infectious diseases.


Assuntos
Glicoproteínas , Humanos , Glicoproteínas/metabolismo , Glicoproteínas/química , Proteômica/métodos , Antígenos de Grupos Sanguíneos/metabolismo , Antígenos de Grupos Sanguíneos/química , Polissacarídeos/química , Polissacarídeos/metabolismo , Fucose/metabolismo , Fucose/química , Fenótipo , Glicosilação , Sistema ABO de Grupos Sanguíneos/metabolismo , Sistema ABO de Grupos Sanguíneos/química
3.
J Biol Chem ; 300(3): 105667, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38272228

RESUMO

The aggregation of α-Synuclein (α-Syn) into amyloid fibrils is the hallmark of Parkinson's disease. Under stress or other pathological conditions, the accumulation of α-Syn oligomers is the main contributor to the cytotoxicity. A potential approach for treating Parkinson's disease involves preventing the accumulation of these α-Syn oligomers. In this study, we present a novel mechanism involving a conserved group of disorderly proteins known as small EDRK-rich factor (SERF), which promotes the aggregation of α-Syn through a cophase separation process. Using diverse methods like confocal microscopy, fluorescence recovery after photobleaching assays, solution-state NMR spectroscopy, and Western blot, we determined that the N-terminal domain of SERF1a plays a role in the interactions that occur during cophase separation. Within these droplets, α-Syn undergoes a gradual transformation from solid condensates to amyloid fibrils, while SERF1a is excluded from the condensates and dissolves into the solution. Notably, in vivo experiments show that SERF1a cophase separation with α-Syn significantly reduces the deposition of α-Syn oligomers and decreases its cellular toxicity under stress. These findings suggest that SERF1a accelerates the conversion of α-Syn from highly toxic oligomers to less toxic fibrils through cophase separation, thereby mitigating the biological damage of α-Syn aggregation.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Humanos , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Amiloide/química , Doença de Parkinson/metabolismo , Separação de Fases , Agregados Proteicos , Agregação Patológica de Proteínas/metabolismo , Fatores de Transcrição , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Células HeLa , Eletricidade Estática
4.
Mar Drugs ; 21(6)2023 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-37367667

RESUMO

Noroviruses, the major cause of acute viral gastroenteritis, are known to bind to histo-blood group antigens (HBGAs), including ABH groups and Lewis-type epitopes, which decorate the surface of erythrocytes and epithelial cells of their host tissues. The biosynthesis of these antigens is controlled by several glycosyltransferases, the distribution and expression of which varies between tissues and individuals. The use of HBGAs as ligands by viruses is not limited to humans, as many animal species, including oysters, which synthesize similar glycan epitopes that act as a gateway for viruses, become vectors for viral infection in humans. Here, we show that different oyster species synthesize a wide range of N-glycans that share histo-blood A-antigens but differ in the expression of other terminal antigens and in their modification by O-methyl groups. In particular, we show that the N-glycans isolated from Crassostrea gigas and Ostrea edulis exhibit exquisite methylation patterns in their terminal N-acetylgalactosamine and fucose residues in terms of position and number, adding another layer of complexity to the post-translational glycosylation modifications of glycoproteins. Furthermore, modeling of the interactions between norovirus capsid proteins and carbohydrate ligands strongly suggests that methylation has the potential to fine-tune the recognition events of oysters by virus particles.


Assuntos
Antígenos de Grupos Sanguíneos , Crassostrea , Norovirus , Ostrea , Humanos , Animais , Crassostrea/metabolismo , Ostrea/metabolismo , Metilação , Ligantes , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Epitopos/metabolismo
5.
J Virol ; 97(4): e0183322, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-36971561

RESUMO

Noroviruses are the leading cause of outbreaks of acute gastroenteritis. These viruses usually interact with histo-blood group antigens (HBGAs), which are considered essential cofactors for norovirus infection. This study structurally characterizes nanobodies developed against the clinically important GII.4 and GII.17 noroviruses with a focus on the identification of novel nanobodies that efficiently block the HBGA binding site. Using X-ray crystallography, we have characterized nine different nanobodies that bound to the top, side, or bottom of the P domain. The eight nanobodies that bound to the top or side of the P domain were mainly genotype specific, while one nanobody that bound to the bottom cross-reacted against several genotypes and showed HBGA blocking potential. The four nanobodies that bound to the top of the P domain also inhibited HBGA binding, and structural analysis revealed that these nanobodies interacted with several GII.4 and GII.17 P domain residues that commonly engaged HBGAs. Moreover, these nanobody complementarity-determining regions (CDRs) extended completely into the cofactor pockets and would likely impede HBGA engagement. The atomic level information for these nanobodies and their corresponding binding sites provide a valuable template for the discovery of additional "designer" nanobodies. These next-generation nanobodies would be designed to target other important genotypes and variants, while maintaining cofactor interference. Finally, our results clearly demonstrate for the first time that nanobodies directly targeting the HBGA binding site can function as potent norovirus inhibitors. IMPORTANCE Human noroviruses are highly contagious and a major problem in closed institutions, such as schools, hospitals, and cruise ships. Reducing norovirus infections is challenging on multiple levels and includes the frequent emergence of antigenic variants, which complicates designing effective, broadly reactive capsid therapeutics. We successfully developed and characterized four norovirus nanobodies that bound at the HBGA pockets. Compared with previously developed norovirus nanobodies that inhibited HBGA through disrupted particle stability, these four novel nanobodies directly inhibited HBGA engagement and interacted with HBGA binding residues. Importantly, these new nanobodies specifically target two genotypes that have caused the majority of outbreaks worldwide and consequently would have an enormous benefit if they could be further developed as norovirus therapeutics. To date, we have structurally characterized 16 different GII nanobody complexes, a number of which block HBGA binding. These structural data could be used to design multivalent nanobody constructs with improved inhibition properties.


Assuntos
Antígenos de Grupos Sanguíneos , Norovirus , Anticorpos de Domínio Único , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Norovirus/efeitos dos fármacos , Norovirus/metabolismo , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/farmacologia , Sítios de Ligação/efeitos dos fármacos , Reações Cruzadas , Termodinâmica , Cristalografia por Raios X , Domínios Proteicos , Ligação Proteica , Modelos Moleculares
6.
Glycoconj J ; 39(5): 633-651, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35962217

RESUMO

Galα1 → and GalNAcα1 → are the two essential key sugars in human blood group AB active glycotopes, in which GalNAcα1 → related sequences are located at both sides of the nonreducing and the reducing ends of human blood group A active O-glycans. It is also found at the nonreducing ends of GlcNAc N-glycans and glycosphingolipid(GSL) of human blood group A active glycotopes (Ah) and Forssman antigen (Fp). When monosaccharides and their α, ß anomers are involved in basic units to express the complex size of the combining sites of the GalNAcα1 → specific lectins, they can be divided into a cavity site to accommodate the GalNAcα → key sugar and a subsite with a wide and broad range of recognition area to adopt the rest part of sugar sequences or glycotopes. The function of the subsite is assumed to act as an enhancement factor to increase its affinity power. The following three points are the theme of this mini review: (1) the loci and distribution of the GalNAcα1 → related glycotopes in mammalian glycoconjugates are illustrated and their chemical structures are advanced by the expression of the disaccharide units and code system; (2) the sizes and motifs of GalNAcα1 → specific lectin-glycan interactions are given and (3) the role of the polyvalent blood group Ah and Bh glycotopes as blood group AB antigens are proposed. These three highlights should provide an essential background required for the advances in this field.


Assuntos
Antígenos de Grupos Sanguíneos , Lectinas , Animais , Antígenos de Grupos Sanguíneos/química , Dissacarídeos/química , Glicoconjugados/metabolismo , Humanos , Lectinas/genética , Lectinas/metabolismo , Mamíferos/metabolismo , Polissacarídeos/química
7.
FEBS Open Bio ; 12(8): 1489-1497, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35674188

RESUMO

Human norovirus (HuNoV) is the primary viral pathogen that causes acute gastroenteritis (AGE) in humans. The protruding (P) domain of HuNoV interacts with cell surface histo-blood group antigens (HBGAs) to initiate infection. Owing to the lack of an effective in vitro culture method and a robust animal model, our understanding of HuNoVs is limited, and as a result, there are no commercial vaccines or antivirals available at present against the virus. In an attempt to develop a preventative measure, we previously identified that bovine colostrum (bCM) contains functional factors that inhibit the binding of HuNoV P domain to its HBGA receptors. In this study, a candidate functional factor in bCM was identified as immunoglobulin M (IgM) using mass spectrometry, followed by database comparison. The natural antibody IgM was further verified to be a functional protein that inhibited HuNoV P protein binding to HBGA receptors through receptor-binding inhibition experiments using bCM, commercial IgM, and fetal bovine serum. Our findings provide a foundation for future development of natural IgM into an antiviral drug, which may help to prevent and/or treat HuNoV infection.


Assuntos
Antígenos de Grupos Sanguíneos , Norovirus , Animais , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Humanos , Imunoglobulina M , Norovirus/fisiologia , Ligação Proteica , Domínios Proteicos
8.
FEBS Open Bio ; 12(3): 560-570, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35038379

RESUMO

Noroviruses have been identified as major causative agents of acute nonbacterial gastroenteritis in humans. Histo-blood group antigens (HBGAs) are thought to play a major role among the host cellular factors influencing norovirus infection. Genogroup I, genotype 9 (GI.9) is the most recently identified genotype within genogroup I, whose representative strain is the Vancouver 730 norovirus. However, the molecular interactions between host antigens and the GI.9 capsid protein have not been investigated in detail. In this study, we demonstrate that the GI.9 norovirus preferentially binds Lewis antigens over blood group A, B, and H antigens, as revealed by an HBGA binding assay using virus-like particles. We determined the crystal structures of the protruding domain of the GI.9 capsid protein in the presence or absence of Lewis antigens. Our analysis demonstrated that Lewis fucose (α1-3/4 fucose) represents a key moiety for the GI.9 protein-HBGA interaction, thus suggesting that Lewis antigens might play a critical role during norovirus infection. In addition to previously reported findings, our observations may support the future design of antiviral agents and vaccines against noroviruses.


Assuntos
Antígenos de Grupos Sanguíneos , Norovirus , Sítios de Ligação , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Cristalografia por Raios X , Fucose/química , Fucose/metabolismo , Humanos , Modelos Moleculares , Norovirus/química , Norovirus/genética , Norovirus/metabolismo , Ligação Proteica
9.
Biochem Soc Trans ; 50(1): 347-359, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-34940787

RESUMO

Infection with human noroviruses requires attachment to histo blood group antigens (HBGAs) via the major capsid protein VP1 as a primary step. Several crystal structures of VP1 protruding domain dimers, so called P-dimers, complexed with different HBGAs have been solved to atomic resolution. Corresponding binding affinities have been determined for HBGAs and other glycans exploiting different biophysical techniques, with mass spectrometry (MS) and nuclear magnetic resonance (NMR) spectroscopy being most widely used. However, reported binding affinities are inconsistent. At the extreme, for the same system MS detects binding whereas NMR spectroscopy does not, suggesting a fundamental source of error. In this short essay, we will explain the reason for the observed differences and compile reliable and reproducible binding affinities. We will then highlight how a combination of MS techniques and NMR experiments affords unique insights into the process of HBGA binding by norovirus capsid proteins.


Assuntos
Antígenos de Grupos Sanguíneos , Norovirus , Sítios de Ligação , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Humanos , Norovirus/química , Norovirus/metabolismo , Polissacarídeos/metabolismo , Ligação Proteica
10.
Viruses ; 13(10)2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34696500

RESUMO

Recognition of cell-surface glycans is an important step in the attachment of several viruses to susceptible host cells. The molecular basis of glycan interactions and their functional consequences are well studied for human norovirus (HuNoV), an important gastrointestinal pathogen. Histo-blood group antigens (HBGAs), a family of fucosylated carbohydrate structures that are present on the cell surface, are utilized by HuNoVs to initially bind to cells. In this review, we describe the discovery of HBGAs as genetic susceptibility factors for HuNoV infection and review biochemical and structural studies investigating HuNoV binding to different HBGA glycans. Recently, human intestinal enteroids (HIEs) were developed as a laboratory cultivation system for HuNoV. We review how the use of this novel culture system has confirmed that fucosylated HBGAs are necessary and sufficient for infection by several HuNoV strains, describe mechanisms of antibody-mediated neutralization of infection that involve blocking of HuNoV binding to HBGAs, and discuss the potential for using the HIE model to answer unresolved questions on viral interactions with HBGAs and other glycans.


Assuntos
Antígenos de Grupos Sanguíneos/metabolismo , Infecções por Caliciviridae/metabolismo , Polissacarídeos/metabolismo , Animais , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/genética , Infecções por Caliciviridae/epidemiologia , Fucosiltransferases/genética , Glicoconjugados , Interações entre Hospedeiro e Microrganismos , Humanos , Intestinos , Modelos Moleculares , Norovirus/genética , Polissacarídeos/genética , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Ligação Viral , Galactosídeo 2-alfa-L-Fucosiltransferase
11.
J Biol Chem ; 295(52): 18426-18435, 2020 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-33127644

RESUMO

α-Linked galactose is a common carbohydrate motif in nature that is processed by a variety of glycoside hydrolases from different families. Terminal Galα1-3Gal motifs are found as a defining feature of different blood group and tissue antigens, as well as the building block of the marine algal galactan λ-carrageenan. The blood group B antigen and linear α-Gal epitope can be processed by glycoside hydrolases in family GH110, whereas the presence of genes encoding GH110 enzymes in polysaccharide utilization loci from marine bacteria suggests a role in processing λ-carrageenan. However, the structure-function relationships underpinning the α-1,3-galactosidase activity within family GH110 remain unknown. Here we focus on a GH110 enzyme (PdGH110B) from the carrageenolytic marine bacterium Pseudoalteromonas distincta U2A. We showed that the enzyme was active on Galα1-3Gal but not the blood group B antigen. X-ray crystal structures in complex with galactose and unhydrolyzed Galα1-3Gal revealed the parallel ß-helix fold of the enzyme and the structural basis of its inverting catalytic mechanism. Moreover, an examination of the active site reveals likely adaptations that allow accommodation of fucose in blood group B active GH110 enzymes or, in the case of PdGH110, accommodation of the sulfate groups found on λ-carrageenan. Overall, this work provides insight into the first member of a predominantly marine clade of GH110 enzymes while also illuminating the structural basis of α-1,3-galactoside processing by the family as a whole.


Assuntos
Antígenos de Grupos Sanguíneos/metabolismo , Carragenina/metabolismo , Galactosídeos/metabolismo , Glicosídeo Hidrolases/química , Pseudoalteromonas/enzimologia , Antígenos de Grupos Sanguíneos/química , Carragenina/química , Domínio Catalítico , Cristalografia por Raios X , Galactosídeos/química , Glicosídeo Hidrolases/classificação , Glicosídeo Hidrolases/metabolismo , Hidrólise , Modelos Moleculares , Filogenia , Conformação Proteica , Especificidade por Substrato
12.
Blood Transfus ; 18(5): 366-373, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32931415

RESUMO

BACKGROUND: Alloanti-Dia can be implicated in mild to severe blood transfusion reactions. Given the concomitance of a high prevalence of the Dia antigen and antibody circulating in some populations, an anti-Dia typing reagent is required in order to enable safe blood transfusions. Limitations of hybridoma technology to produce such a reagent led to the use of phage display technology to generate an anti-Dia monoclonal antibody. MATERIALS AND METHODS: A library of phages displaying murine single-chain variable fragment antibody (scFv-phages) was consecutively adsorbed with different panels of Di(a-b+) red cells to eliminate scFc-phages that potentially bind irrelevant blood group antigens. Thereafter, the subtractive library was specifically selected for the scFv-phages that bound Dia antigen by sequentially biopanning the library with Di(a+b+) cell ghosts and Di(a+b-) intact red cells. A specific interaction between the selected scFv-phages and Dia epitope was validated with the Dia peptide by a competitive haemagglutination inhibition assay and confirmed with the red cells by flow cytometry. RESULTS: An scFv-phage clone specifically bound the Dia epitope, as shown by its binding competition with the human anti-Dia to the Dia peptide in a haemagglutination inhibition test. Moreover, it was highly reactive to Di(a+b+) red cells but not to Di(a-b+) red cells, as determined by flow cytometry. DISCUSSION: In this study, a Dia-specific scFv-phage antibody was successfully produced. The selection protocol might be a prototypic platform for producing monoclonal antibodies to relevant blood group antigens. The scFv-phage produced in this way warrants further development for use as a reagent for Dia red cell typing.


Assuntos
Especificidade de Anticorpos/genética , Antígenos de Grupos Sanguíneos , Epitopos , Biblioteca de Peptídeos , Peptídeos , Anticorpos de Cadeia Única , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/imunologia , Epitopos/química , Epitopos/imunologia , Humanos , Peptídeos/química , Peptídeos/imunologia , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia
13.
Transfusion ; 60(10): 2408-2418, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32870515

RESUMO

BACKGROUND: All antigens described in the KN blood group system are located in the long homologous repeat D (LHR-D) of complement receptor 1 (CR1). While there have been reports that some sera react only with the long homologous repeat C (LHR-C), the antigens in LHR-C are unknown. STUDY DESIGN AND METHODS: Recombinant LHR-C and LHR-D were used to identify antibodies directed against LHR-C of CR1, into which a point mutation was introduced to characterize the underlying blood group antigens. In addition, database studies to define haplotypes of CR1 were performed. RESULTS: Several antisera were identified that were specific against CR1 p.1208His and against CR1 p.1208Arg, located in LHR-C. Fifteen KN haplotypes were found in the Ensembl genome browser. It was shown that due to a linkage disequilibrium anti-CR1 p.1208His may be mistaken for anti-KCAM. CONCLUSION: A novel antithetical KN blood group antigen pair was found at position p.1208 of CR1, for which the names DACY and YCAD are proposed. Antibodies against these two novel antigens seem to contribute to more than a quarter of all KN sera in Europe.


Assuntos
Antígenos de Grupos Sanguíneos , Mutação Puntual , Polimorfismo Genético , Receptores de Complemento 3b , Substituição de Aminoácidos , Anticorpos/química , Anticorpos/imunologia , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/imunologia , Europa (Continente) , Humanos , Domínios Proteicos , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
14.
Nat Commun ; 11(1): 3569, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32678083

RESUMO

The clinically important MAM blood group antigen is present on haematopoietic cells of all humans except rare MAM-negative individuals. Its molecular basis is unknown. By whole-exome sequencing we identify EMP3, encoding epithelial membrane protein 3 (EMP3), as a candidate gene, then demonstrate inactivating mutations in ten known MAM-negative individuals. We show that EMP3, a purported tumour suppressor in various solid tumours, is expressed in erythroid cells. Disruption of EMP3 by CRISPR/Cas9 gene editing in an immortalised human erythroid cell line (BEL-A2) abolishes MAM expression. We find EMP3 to associate with, and stabilise, CD44 in the plasma membrane. Furthermore, cultured erythroid progenitor cells from MAM-negative individuals show markedly increased proliferation and higher reticulocyte yields, suggesting an important regulatory role for EMP3 in erythropoiesis and control of cell production. Our data establish MAM as a new blood group system and demonstrate an interaction of EMP3 with the cell surface signalling molecule CD44.


Assuntos
Antígenos de Grupos Sanguíneos/genética , Proliferação de Células , Células Eritroides/citologia , Glicoproteínas de Membrana/genética , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Plaquetas/metabolismo , Células Cultivadas , Membrana Eritrocítica/metabolismo , Células Eritroides/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Mutação , Fenótipo , Ligação Proteica , Sequenciamento do Exoma
15.
J Biotechnol ; 318: 31-38, 2020 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-32387450

RESUMO

Norovirus infections cause severe gastroenteritis in millions of people every year. Infection requires the recognition of histo-blood group antigens (HBGAs), but such interactions can be inhibited by human milk oligosaccharides (HMOs), which act as structurally-similar decoys. HMO supplements could help to prevent norovirus infections, but the industrial production of complex HMOs is challenging. Here we describe a large-scale fermentation process that yields several kilograms of lacto-N-fucopentaose I (LNFP I). The product was synthesized in Escherichia coli BL21(DE3) cells expressing a recombinant N-acetylglucosaminyltransferase, ß(1,3)galactosyltransferase and α(1,2)fucosyltransferase. Subsequent in vitro enzymatic conversion produced HBGA types A1 and B1 for norovirus inhibition assays. These carbohydrates inhibited the binding of GII.17 virus-like particles (VLPs) to type A1 and B1 trisaccharides more efficiently than simpler fucosylated HMOs, which were in turn more effective than any non-fucosylated structures. However, we found that the simpler fucosylated HMOs were more effective than complex molecules such as LNFP I when inhibiting the binding of GII.17 and GII.4 VLPs to human gastric mucins and mucins from human amniotic fluid. Our results show that complex fucosylated HMOs can be produced by large-scale fermentation and that a combination of simple and complex fucosylated structures is more likely to prevent norovirus infections.


Assuntos
Norovirus/efeitos dos fármacos , Oligossacarídeos/metabolismo , Oligossacarídeos/farmacologia , Receptores Virais/metabolismo , Biotecnologia , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/metabolismo , Antígenos de Grupos Sanguíneos/farmacologia , Fermentação , Humanos , Concentração Inibidora 50 , Leite Humano/química , Mucinas/metabolismo , Norovirus/fisiologia , Oligossacarídeos/química , Trissacarídeos/metabolismo
16.
ACS Appl Mater Interfaces ; 12(20): 22426-22432, 2020 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-32347090

RESUMO

Production of red blood cells (RBCs) without immunogenicity of blood group antigens is of special interest in blood transfusion therapy in clinical chemistry. In this study, a selective cell surface engineering method was developed for the preparation of antigen-shielded RBCs based on molecular imprinting. Using an epitope imprinting method, biocompatible molecularly imprinted nanogels (MIgels) were prepared with a high affinity to the blood group antigens of RBCs. The antigen-shielded RBCs could avoid the agglutination caused by blood group mismatch, resulting in the antigen-shielded RBCs in efficiently substituting RBCs in case of a shortage of blood supply. Moreover, the antigen-shielded RBCs could maintain the normal physiological structure and functions of the original RBCs. We believe that the selective cell surface engineering presented in this work may offer significant benefits in specific cell protection for biomedical application.


Assuntos
Antígenos de Grupos Sanguíneos/química , Eritrócitos/química , Géis/química , Hemaglutinação/efeitos dos fármacos , Anticorpos Monoclonais/imunologia , Antígenos de Grupos Sanguíneos/imunologia , Engenharia Celular/métodos , Humanos , Impressão Molecular
17.
Sci Rep ; 10(1): 4661, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32170122

RESUMO

Noroviruses are known to bind to histo-blood group antigens (HBGAs) and the specific binding patterns depend on the virus genotype. However, the development of point-of-care diagnostic assays based on this binding has been challenging due to low assay sensitivity. This study utilized a well-defined stool collection from a GII.2 Snow Mountain Virus (SMV) human challenge study to investigate virus recovery from stool and emesis samples using HBGA-coated beads. SMV was recovered from H type III-coated beads for 13 stool specimens out of 27 SMV-positive specimens tested. After adjusting for non-specific binding to PEG-coated beads, the mean percent recovery by H type III-coated beads was 308.11% +/- 861.61. Recovery by H type III ligands was subject-specific and weakly correlated with stool consistency. Input virus titer was not correlated with SMV recovery. The results suggest that the generally low virus recovery we observed may be due to bead saturation or hindrance by existing glycans in the matrix that precluded the virus from being captured by the synthetic glycans. These results indicate a strong role for subject-specific and matrix effects in HBGA binding by SMV. Further investigation of the nature of this interference is needed to facilitate development of high sensitivity diagnostic assays.


Assuntos
Infecções por Caliciviridae/diagnóstico , Glicoconjugados/síntese química , Glicoconjugados/metabolismo , Norovirus/isolamento & purificação , Antígenos de Grupos Sanguíneos/química , Fezes/virologia , Glicoconjugados/química , Humanos , Estrutura Molecular , Norovirus/fisiologia , Sistemas Automatizados de Assistência Junto ao Leito , Polissacarídeos , Biologia Sintética , Ligação Viral
18.
Molecules ; 25(2)2020 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-31936166

RESUMO

Lectins mediate adhesion of pathogens to host tissues, filling in a key role in the first steps of infection. Belonging to the opportunistic pathogen Burkholderia cenocepacia, BC2L-C is a superlectin with dual carbohydrate specificity, believed to mediate cross-linking between bacteria and host cells. Its C-terminal domain binds to bacterial mannosides while its N-terminal domain (BCL2-CN) recognizes fucosylated human epitopes. BC2L-CN presents a tumor necrosis factor alpha (TNF-) fold previously unseen in lectins with a novel fucose binding mode. We report, here, the production of a novel recombinant form of BC2L-CN (rBC2L-CN2), which allowed better protein stability and unprecedented co-crystallization with oligosaccharides. Isothermal calorimetry measurements showed no detrimental effect on ligand binding and data were obtained on the binding of Globo H hexasaccharide and l-galactose. Crystal structures of rBC2L-CN2 were solved in complex with two blood group antigens: H-type 1 and H-type 3 (Globo H) by X-ray crystallography. They provide new structural information on the binding site, of importance for the structural-based design of glycodrugs as new antimicrobials with antiadhesive properties.


Assuntos
Antígenos de Grupos Sanguíneos/química , Burkholderia cenocepacia/química , Lectinas/química , Oligossacarídeos/química , Antígenos de Diferenciação/química , Antígenos Glicosídicos Associados a Tumores/química , Sítios de Ligação , Burkholderia cenocepacia/genética , Burkholderia cenocepacia/metabolismo , Cristalografia por Raios X , Epitopos/química , Fucose/química , Expressão Gênica , Humanos , Manosídeos/química , Modelos Moleculares , Ligação Proteica , Proteínas Recombinantes/genética , Fator de Necrose Tumoral alfa/química
19.
J Agric Food Chem ; 68(5): 1207-1212, 2020 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-31755264

RESUMO

Human noroviruses (HuNoVs) are among the main pathogens causing acute nonbacterial gastroenteritis. Histo-blood group antigens (HBGAs) are widely accepted receptors for HuNoV specific binding. HBGA-like substances in produce are also considered as the critical ligands for capture of HuNoVs. However, the composition of viral ligands from food substrates remains unknown. In this study, an oligosaccharide (H2N2F2) was captured and isolated from romaine lettuce extract by a bacterial surface display system. Using electrospray ionization mass spectrometry and tandem mass spectrometry, it was shown that H2N2F2 was most likely to be a chimera of type A, H, and Lewis a HBGAs. The composition was consistent with our ELISA results using a panel of monoclonal antibodies against HBGAs. Our results revealed a possible interaction mechanism between HuNoVs and romaine lettuce. Better understanding of the interaction of HuNoVs with easily contaminated produce will ultimately aid in the control of and reduction in disease outbreaks.


Assuntos
Antígenos de Plantas/metabolismo , Antígenos de Grupos Sanguíneos/metabolismo , Lactuca/virologia , Norovirus/fisiologia , Receptores Virais/metabolismo , Ligação Viral , Antígenos de Plantas/química , Antígenos de Plantas/genética , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/genética , Infecções por Caliciviridae/genética , Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/virologia , Humanos , Lactuca/química , Lactuca/genética , Lactuca/metabolismo , Espectrometria de Massas , Norovirus/genética , Oligossacarídeos/química , Oligossacarídeos/genética , Oligossacarídeos/metabolismo , Ligação Proteica , Receptores Virais/química , Receptores Virais/genética
20.
J Mol Model ; 25(12): 369, 2019 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-31781987

RESUMO

Norovirus, also called winter vomiting bug, is the most common cause for gastroenteritis and severe childhood diarrhea disease. High mutation rates cause drug resistance and thus complicate the development of an effective therapeutics against virus infection. The virus protein enters the host cell via the interaction with histo-blood group antigens (HBGAs), formed by oligosaccharides. To date, the crystal structures of numerous complexes of virus proteins with different antigens have been reported. The HBGAs bind to the two distinct regions of the virus proteins. Herein, the affinity of different variants of virus protein to some common glycans has been computationally analyzed. Molecular docking studies as combination of docking scores and rmsd values revealed that the binding region 1 is more attractive for the ligands in variants of categories 1-5, but selectivity is drastically shifted to region 2 due to in category 6. In addition, molecular dynamics simulations were unraveled when the region 1 is hindered (in category 6); the blocking loop has less fluctuation than that of unblocked in other categories.


Assuntos
Antígenos de Grupos Sanguíneos/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Norovirus/metabolismo , Proteínas Virais/metabolismo , Sítios de Ligação , Antígenos de Grupos Sanguíneos/química , Mutação , Norovirus/genética , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Proteínas Virais/química , Proteínas Virais/genética , Internalização do Vírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA