Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Front Microbiol ; 14: 1141652, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36970669

RESUMEN

Human rotaviruses attach to histo-blood group antigens glycans and null alleles of the ABO, FUT2 and FUT3 genes seem to confer diminished risk of gastroenteritis. Yet, the true extent of this protection remains poorly quantified. Here, we conducted a prospective study to evaluate the risk of consulting at the hospital in non-vaccinated pediatric patients according to the ABO, FUT2 (secretor) and FUT3 (Lewis) polymorphisms, in Metropolitan France and French Guiana. At both locations, P genotypes were largely dominated by P [8]-3, with P [6] cases exclusively found in French Guiana. The FUT2 null (nonsecretor) and FUT3 null (Lewis negative) phenotypes conferred near full protection against severe gastroenteritis due to P [8]-3 strains (OR 0.03, 95% CI [0.00-0.21] and 0.1, 95% CI [0.01-0.43], respectively in Metropolitan France; OR 0.08, 95% CI [0.01-0.52] and 0.14, 95%CI [0.01-0.99], respectively in French Guiana). Blood group O also appeared protective in Metropolitan France (OR 0.38, 95% CI [0.23-0.62]), but not in French Guiana. The discrepancy between the two locations was explained by a recruitment at the hospital of less severe cases in French Guiana than in Metropolitan France. Considering the frequencies of the null ABO, Secretor and Lewis phenotypes, the data indicate that in a Western European population, 34% (95% CI [29%; 39%]) of infants are genetically protected against rotavirus gastroenteritis of sufficient severity to lead to hospital visit.

2.
Front Microbiol ; 14: 1123803, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36922975

RESUMEN

The FUT2 α1,2fucosyltransferase contributes to the synthesis of fucosylated glycans used as attachment factors by several pathogens, including noroviruses and rotaviruses, that can induce life-threatening gastroenteritis in young children. FUT2 genetic polymorphisms impairing fucosylation are strongly associated with resistance to dominant strains of both noroviruses and rotaviruses. Interestingly, the wild-type allele associated with viral gastroenteritis susceptibility inversely appears to be protective against several inflammatory or autoimmune diseases for yet unclear reasons, although a FUT2 influence on microbiota composition has been observed. Here, we studied a cohort of young healthy adults and showed that the wild-type FUT2 allele was associated with the presence of anti-RVA antibodies, either neutralizing antibodies or serum IgA, confirming its association with the risk of RVA gastroenteritis. Strikingly, it was also associated with the frequency of gut microbiota-induced regulatory T cells (Tregs), so-called DP8α Tregs, albeit only in individuals who had anti-RVA neutralizing antibodies or high titers of anti-RVA IgAs. DP8α Tregs specifically recognize the human symbiont Faecalibacterium prausnitzii, which strongly supports their induction by this anti-inflammatory bacterium. The proportion of F. prausnitzii in feces was also associated with the FUT2 wild-type allele. These observations link the FUT2 genotype with the risk of RVA gastroenteritis, the microbiota and microbiota-induced DP8α Treg cells, suggesting that the anti-RVA immune response might involve an induction/expansion of these T lymphocytes later providing a balanced immunological state that confers protection against inflammatory diseases.

3.
Front Microbiol ; 12: 641460, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33643275

RESUMEN

Human serum contains large amounts of anti-carbohydrate antibodies, some of which may recognize epitopes on viral glycans. Here, we tested the hypothesis that such antibodies may confer protection against COVID-19 so that patients would be preferentially found among people with low amounts of specific anti-carbohydrate antibodies since individual repertoires vary considerably. After selecting glycan epitopes commonly represented in the human anti-carbohydrate antibody repertoire that may also be expressed on viral glycans, plasma levels of the corresponding antibodies were determined by ELISA in 88 SARS-CoV-2 infected individuals, including 13 asymptomatic, and in 82 non-infected controls. We observed that anti-Tn antibodies levels were significantly lower in patients as compared to non-infected individuals. This was not observed for any of the other tested carbohydrate epitopes, including anti-αGal antibodies used as a negative control since the epitope cannot be synthesized by humans. Owing to structural homologies with blood groups A and B antigens, we also observed that anti-Tn and anti-αGal antibodies levels were lower in blood group A and B, respectively. Analyses of correlations between anti-Tn and the other anti-carbohydrates tested revealed divergent patterns of correlations between patients and controls, suggesting qualitative differences in addition to the quantitative difference. Furthermore, anti-Tn levels correlated with anti-S protein levels in the patients' group, suggesting that anti-Tn might contribute to the development of the specific antiviral response. Overall, this first analysis allows to hypothesize that natural anti-Tn antibodies might be protective against COVID-19.

4.
Viruses ; 13(2)2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499228

RESUMEN

Since the emergence of COVID-19, many publications have reported associations with ABO blood types. Despite between-study discrepancies, an overall consensus has emerged whereby blood group O appears associated with a lower risk of COVID-19, while non-O blood types appear detrimental. Two major hypotheses may explain these findings: First, natural anti-A and anti-B antibodies could be partially protective against SARS-CoV-2 virions carrying blood group antigens originating from non-O individuals. Second, O individuals are less prone to thrombosis and vascular dysfunction than non-O individuals and therefore could be at a lesser risk in case of severe lung dysfunction. Here, we review the literature on the topic in light of these hypotheses. We find that between-study variation may be explained by differences in study settings and that both mechanisms are likely at play. Moreover, as frequencies of ABO phenotypes are highly variable between populations or geographical areas, the ABO coefficient of variation, rather than the frequency of each individual phenotype is expected to determine impact of the ABO system on virus transmission. Accordingly, the ABO coefficient of variation correlates with COVID-19 prevalence. Overall, despite modest apparent risk differences between ABO subtypes, the ABO blood group system might play a major role in the COVID-19 pandemic when considered at the population level.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/sangre , COVID-19/sangre , Susceptibilidad a Enfermedades/sangre , COVID-19/epidemiología , COVID-19/microbiología , Susceptibilidad a Enfermedades/epidemiología , Susceptibilidad a Enfermedades/microbiología , Susceptibilidad a Enfermedades/patología , Humanos , Incidencia , Isoanticuerpos/sangre , Microbiota , Oportunidad Relativa , SARS-CoV-2 , Trombosis/sangre , Trombosis/epidemiología , Trombosis/microbiología
5.
Front Microbiol ; 12: 799519, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069504

RESUMEN

ABO blood groups appear to be associated with the risk of SARS-CoV-2 infection, but the underlying mechanisms and their real importance remain unclear. Two hypotheses have been proposed: ABO compatibility-dependence (neutralization by anti-ABO antibodies) and ABO-dependent intrinsic susceptibility (spike protein attachment to histo-blood group glycans). We tested the first hypothesis through an anonymous questionnaire addressed to hospital staff members. We estimated symptomatic secondary attack rates (SAR) for 333 index cases according to spouse ABO blood group compatibility. Incompatibility was associated with a lower SAR (28% vs. 47%; OR 0.43, 95% CI 0.27-0.69), but no ABO dependence was detected in compatible situations. For the second hypothesis, we detected no binding of recombinant SARS-CoV-2 RBD to blood group-containing glycans. Thus, although no intrinsic differences in susceptibility according to ABO blood type were detected, ABO incompatibility strongly decreased the risk of COVID-19 transmission, suggesting that anti-ABO antibodies contribute to virus neutralization.

6.
Int J Infect Dis ; 104: 242-249, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33326874

RESUMEN

BACKGROUND: Susceptibility to Covid-19 has been found to be associated with the ABO blood group, with O type individuals being at a lower risk. However, the underlying mechanism has not been elucidated. Here, we aimed to test the hypothesis that Covid-19 patients might have lower levels of ABO antibodies than non-infected individuals as they could offer some degree of protection. METHODS: After showing that the viral spike protein harbors the ABO glycan epitopes when produced by cells expressing the relevant glycosyltransferases, like upper respiratory tract epithelial cells, we enrolled 290 patients with Covid-19 and 276 asymptomatic controls to compare their levels of natural ABO blood group antibodies. RESULTS: We found significantly lower IgM anti-A + anti-B agglutination scores in blood group O patients (76.93 vs 88.29, P-value = 0.034) and lower levels of anti-B (24.93 vs 30.40, P-value = 0.028) and anti-A antibodies (28.56 vs 36.50, P-value = 0.048) in blood group A and blood group B patients, respectively, compared to controls. CONCLUSION: In this study, we showed that ABO antibody levels are significantly lower in Covid-19 patients compared to controls. These findings could indicate that patients with low levels of ABO antibodies are at higher risk of being infected.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/inmunología , Anticuerpos/sangre , COVID-19/sangre , Polisacáridos/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/virología , Susceptibilidad a Enfermedades , Células Epiteliales/inmunología , Epítopos/inmunología , Femenino , Galactosiltransferasas , Humanos , Inmunoglobulina M/inmunología , Masculino , Persona de Mediana Edad , Riesgo , Adulto Joven
7.
J Infect Dis ; 222(5): 836-839, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32188998

RESUMEN

In Tunisia, we observed that rotavirus P[8]-3 and P[4] strains in young children with gastroenteritis associate with secretor histo-blood group phenotype. In contrast, the emerging P[8]-4 strain, representing 10% of cases, was exclusively found in nonsecretor patients. Unlike VP8* from P[8]-3 and P[4] strains, the P[8]-4 VP8* protein attached to glycans from saliva samples regardless of the donor's secretor status. Interestingly, a high frequency of FUT2 enzyme deficiency (nonsecretor phenotype) was observed in the population. This may allow cocirculation of P[8]-3 and P[8]-4 strains in secretor and nonsecretor children, respectively.


Asunto(s)
Fucosiltransferasas/genética , Especificidad del Huésped , Proteínas de Unión al ARN/metabolismo , Infecciones por Rotavirus/genética , Rotavirus/genética , Proteínas no Estructurales Virales/metabolismo , Preescolar , Genotipo , Humanos , Lactante , Recién Nacido , Fenotipo , Polisacáridos/metabolismo , Proteínas de Unión al ARN/genética , Rotavirus/fisiología , Saliva , Proteínas no Estructurales Virales/genética , Acoplamiento Viral , Galactósido 2-alfa-L-Fucosiltransferasa
8.
Hum Genet ; 139(6-7): 903-910, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31760489

RESUMEN

Together, norovirus and rotavirus are responsible for the majority of gastroenteritis cases worldwide, leading to a large number of deaths of children in low-income countries. Both attach to glycans of the histo-blood group antigen type (HBGAs) widely expressed in the digestive tract of vertebrates, albeit with interspecies differences. In humans, their synthesis is performed by glycosyltransferases encoded by the highly polymorphic ABO, FUT2 and FUT3 genes that are under long-term balanced selection. The combination of functional and null or weak alleles at these loci provides a diversity of glycan structures that define the ABO, Secretor and Lewis phenotypes. At the initial stage of infection norovirus and rotavirus attach to these glycans, although distinct strains of each virus present different specificities for individual glycans, hence exhibiting preferences for different human phenotypes. Absence or low expression of the recognized glycan motifs due to genetic polymorphism is associated with resistance to the disease, showing that the HBGA polymorphisms provide a population-based innate protection. Epidemiologically dominant strains of either norovirus or rotavirus display specificity for glycan motifs present in large fractions of the population, which may differ between geographical areas in accordance with the frequency of the ABO, FUT2, FUT3 gene polymorphisms. Evidence for virus adaptation to these geographical differences is amounting, indicative of a host-pathogen co-evolution and suggesting that enteric pathogens such as norovirus and rotavirus are likely the driving forces behind the balanced HBGA polymorphisms.


Asunto(s)
Gastroenteritis/etiología , Predisposición Genética a la Enfermedad , Norovirus/metabolismo , Polimorfismo Genético , Polisacáridos/genética , Rotavirus/metabolismo , Azúcares/metabolismo , Infecciones por Caliciviridae/complicaciones , Infecciones por Caliciviridae/virología , Gastroenteritis/patología , Glicosiltransferasas/genética , Humanos , Fenotipo , Polisacáridos/metabolismo , Infecciones por Rotavirus/complicaciones , Infecciones por Rotavirus/virología
9.
Sci Rep ; 8(1): 12961, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30154494

RESUMEN

Human strains of rotavirus A (RVAs) recognize fucosylated glycans belonging to histo-blood group antigens (HBGAs) through their spike protein VP8*. Lack of these ligands due to genetic polymorphisms is associated with resistance to gastroenteritis caused by P[8] genotype RVAs. With the aim to delineate the contribution of HBGAs in the process, we analyzed the glycan specificity of VP8* proteins from various P genotypes. Binding to saliva of VP8* from P[8] and P[4] genotypes required expression of both FUT2 and FUT3 enzymes, whilst binding of VP8* from the P[14] genotype required FUT2 and A enzymes. We further defined a glycan motif, GlcNAcß3Galß4GlcNAc, recognized by P[6] clinical strains. Conversion into Lewis antigens by the FUT3 enzyme impaired recognition, explaining their lower binding to saliva of Lewis positive phenotype. In addition, the presence of neutralizing antibodies was associated with the presence of the FUT2 wild type allele in sera from young healthy adults. Nonetheless, in vitro infection of transformed cell lines was independent of HBGAs expression, indicating that HBGAs are not human RV receptors. The match between results from saliva-based binding assays and the epidemiological data indicates that the polymorphism of human HBGAs controls susceptibility to RVAs, although the exact mechanism remains unclear.


Asunto(s)
Antígenos de Grupos Sanguíneos , Gastroenteritis , Proteínas de Unión al ARN , Infecciones por Rotavirus , Rotavirus , Proteínas no Estructurales Virales , Adolescente , Adulto , Animales , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Células CACO-2 , Chlorocebus aethiops , Femenino , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Gastroenteritis/genética , Gastroenteritis/metabolismo , Gastroenteritis/virología , Humanos , Masculino , Unión Proteica , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Rotavirus/genética , Rotavirus/metabolismo , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/metabolismo , Células Vero , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Galactósido 2-alfa-L-Fucosiltransferasa
10.
J Virol ; 92(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29187537

RESUMEN

Rabbit hemorrhagic disease virus (RHDV) and European brown hare syndrome virus (EBHSV) are two lagoviruses from the family Caliciviridae that cause fatal diseases in two leporid genera, Oryctolagus and Lepus, respectively. In the last few years, several examples of host jumps of lagoviruses among leporids were recorded. In addition, a new pathogenic genotype of RHDV emerged, and many nonpathogenic strains of lagoviruses have been described. The molecular mechanisms behind host shifts and the emergence of virulence are unknown. Since RHDV uses glycans of the histo-blood group antigen type as attachment factors to initiate infection, we studied if glycan specificities of the new pathogenic RHDV genotype, nonpathogenic lagoviruses, and EBHSV potentially play a role in determining the host range and virulence of lagoviruses. We observed binding to A, B, or H antigens of the histo-blood group family for all strains known to primarily infect European rabbits (Oryctolagus cuniculus), which have recently been classified as GI strains. However, we could not explain the emergence of virulence, since similar glycan specificities were found in several pathogenic and nonpathogenic strains. In contrast, EBHSV, recently classified as GII.1, bound to terminal ß-linked N-acetylglucosamine residues of O-glycans. Expression of these attachment factors in the upper respiratory and digestive tracts in three lagomorph species (Oryctolagus cuniculus, Lepuseuropaeus, and Sylvilagus floridanus) showed species-specific patterns regarding susceptibility to infection by these viruses, indicating that species-specific glycan expression is likely a major contributor to lagovirus host specificity and range.IMPORTANCE Lagoviruses constitute a genus of the family Caliciviridae comprising highly pathogenic viruses, RHDV and EBHSV, that infect rabbits and hares, respectively. Recently, nonpathogenic strains were discovered and new pathogenic strains have emerged. In addition, host jumps between lagomorphs have been observed. The mechanisms responsible for the emergence of pathogenicity and host species range are unknown. Previous studies showed that RHDV strains attach to glycans expressed in the upper respiratory and digestive tracts of rabbits, the likely portals of virus entry. Here, we studied the glycan-binding properties of novel pathogenic and nonpathogenic strains looking for a link between glycan binding and virulence or between glycan specificity and host range. We found that glycan binding did not correlate with virulence. However, expression of glycan motifs in the upper respiratory and digestive tracts of lagomorphs revealed species-specific patterns associated with the host ranges of the virus strains, suggesting that glycan diversity contributes to lagovirus host ranges.


Asunto(s)
Infecciones por Caliciviridae/virología , Virus de la Enfermedad Hemorrágica del Conejo/fisiología , Lagomorpha/virología , Lagovirus/fisiología , Polisacáridos/metabolismo , Virulencia , Acoplamiento Viral , Animales , Infecciones por Caliciviridae/metabolismo , Susceptibilidad a Enfermedades , Liebres , Lagomorpha/clasificación , Lagomorpha/metabolismo , Filogenia , Conejos , Especificidad de la Especie
11.
J Gen Virol ; 98(7): 1658-1666, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28714849

RESUMEN

Lagoviruses belong to the Caliciviridae family. They were first recognized as highly pathogenic viruses of the European rabbit (Oryctolagus cuniculus) and European brown hare (Lepus europaeus) that emerged in the 1970-1980s, namely, rabbit haemorrhagic disease virus (RHDV) and European brown hare syndrome virus (EBHSV), according to the host species from which they had been first detected. However, the diversity of lagoviruses has recently expanded to include new related viruses with varying pathogenicity, geographic distribution and host ranges. Together with the frequent recombination observed amongst circulating viruses, there is a clear need to establish precise guidelines for classifying and naming lagovirus strains. Therefore, here we propose a new nomenclature based on phylogenetic relationships. In this new nomenclature, a single species of lagovirus would be recognized and called Lagovirus europaeus. The species would be divided into two genogroups that correspond to RHDV- and EBHSV-related viruses, respectively. Genogroups could be subdivided into genotypes, which could themselves be subdivided into phylogenetically well-supported variants. Based on available sequences, pairwise distance cutoffs have been defined, but with the accumulation of new sequences these cutoffs may need to be revised. We propose that an international working group could coordinate the nomenclature of lagoviruses and any proposals for revision.


Asunto(s)
Lagovirus/clasificación , ARN Viral/genética , Terminología como Asunto , Animales , Infecciones por Caliciviridae/virología , Genotipo , Liebres , Lagovirus/genética , Lagovirus/patogenicidad , Filogenia , Conejos
13.
PLoS Pathog ; 11(4): e1004759, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25875017

RESUMEN

RHDV (rabbit hemorrhagic disease virus), a virulent calicivirus, causes high mortalities in European rabbit populations (Oryctolagus cuniculus). It uses α1,2fucosylated glycans, histo-blood group antigens (HBGAs), as attachment factors, with their absence or low expression generating resistance to the disease. Synthesis of these glycans requires an α1,2fucosyltransferase. In mammals, there are three closely located α1,2fucosyltransferase genes rSec1, rFut2 and rFut1 that arose through two rounds of duplications. In most mammalian species, Sec1 has clearly become a pseudogene. Yet, in leporids, it does not suffer gross alterations, although we previously observed that rabbit Sec1 variants present either low or no activity. Still, a low activity rSec1 allele correlated with survival to an RHDV outbreak. We now confirm the association between the α1,2fucosyltransferase loci and survival. In addition, we show that rabbits express homogenous rFut1 and rFut2 levels in the small intestine. Comparison of rFut1 and rFut2 activity showed that type 2 A, B and H antigens recognized by RHDV strains were mainly synthesized by rFut1, and all rFut1 variants detected in wild animals were equally active. Interestingly, rSec1 RNA levels were highly variable between individuals and high expression was associated with low binding of RHDV strains to the mucosa. Co-transfection of rFut1 and rSec1 caused a decrease in rFut1-generated RHDV binding sites, indicating that in rabbits, the catalytically inactive rSec1 protein acts as a dominant-negative of rFut1. Consistent with neofunctionalization of Sec1 in leporids, gene conversion analysis showed extensive homogenization between Sec1 and Fut2 in leporids, at variance with its limited degree in other mammals. Gene conversion additionally involving Fut1 was also observed at the C-terminus. Thus, in leporids, unlike in most other mammals where it became extinct, Sec1 evolved a new function with a dominant-negative effect on rFut1, contributing to fucosylated glycan diversity, and allowing herd protection from pathogens such as RHDV.


Asunto(s)
Infecciones por Caliciviridae/genética , Fucosiltransferasas/genética , Virus de la Enfermedad Hemorrágica del Conejo/genética , Polimorfismo Genético , Polisacáridos/genética , Animales , Secuencia de Bases , Resistencia a la Enfermedad/genética , Lagomorpha , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Conejos , Transfección
14.
Curr Opin Virol ; 7: 88-94, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25000207

RESUMEN

Noroviruses and rotavirus A bind to polymorphic glycans of the histo-blood group type (HBGAs). Norovirus strains that bind to HBGAs can collectively infect all humans but each strain only infects a subgroup of the population, suggesting a past co-evolution that led to a trade-off where the human population is partly protected whilst the virus circulation is maintained. We termed 'Herd Innate Protection' the host species partial protection provided by the HBGAs polymorphism. Given its recent emergence, high virulence and HBGAs attachment, RHDV provides a model for studying calicivirus-host co-evolution. Field observations documented evolution of the virus ability to recognize the host HBGAs diversity and reciprocal strain-dependent selection of HBGA phenotypes following outbreaks, indicating host-pathogen co-evolution involving glycan polymorphisms.


Asunto(s)
Evolución Biológica , Infecciones por Caliciviridae/genética , Infecciones por Caliciviridae/metabolismo , Caliciviridae/genética , Caliciviridae/metabolismo , Polisacáridos/metabolismo , Receptores Virales/metabolismo , Animales , Infecciones por Caliciviridae/virología , Humanos
15.
J Infect Dis ; 210(2): 183-91, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24459192

RESUMEN

BACKGROUND: Noroviruses (NoVs) represent a considerable public health burden. Despite their enormous genetic diversity, most outbreaks are due to the single GII.4 genotype, but the reasons for this are poorly understood. NoVs use histo-blood group antigens (HBGAs) as attachment factors. Since HBGAs are present in saliva, binding of strains to saliva is commonly used as a surrogate for recognition of the gut surface by specific strains, although the relationship between saliva and gut tissue expression of HBGAs is not well defined. METHODS: The presence of fucosylated HBGAs in saliva and stomach biopsy specimens, as well as that of genogroup I.1 and genogroup II.4 virus-like particles, were compared in a series of 109 donors from Portugal. RESULTS: An overall good concordance between HBGA expression in saliva and stomach surface mucosa was observed. However, unexpected mucosal expression of α(1,2)fucosylated epitopes in nonsecretor individuals was frequently detected, allowing for GII.4 attachment. Although all individuals were infected with Helicobacter pylori, abnormal expression of α(1,2)fucosylated motifs and binding of GII.4 virus-like particles in nonsecretors' mucosa were associated with positivity for the H. pylori CagA virulence factor. CONCLUSIONS: Infection by CagA-positive H. pylori induces expression of GII.4 attachment factors in nonsecretors' mucosa, expanding the host range of these strains and thereby possibly contributing to their epidemiological dominance.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Antígenos de Grupos Sanguíneos/análisis , Mucosa Gástrica/química , Mucosa Gástrica/virología , Norovirus/aislamiento & purificación , Saliva/química , Saliva/virología , Genotipo , Voluntarios Sanos , Infecciones por Helicobacter/microbiología , Helicobacter pylori/aislamiento & purificación , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Norovirus/clasificación , Norovirus/genética , Portugal , Receptores Virales/análisis , Factores de Virulencia/metabolismo
16.
J Infect Dis ; 209(8): 1227-30, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24277741

RESUMEN

Attachment to carbohydrates of the histo-blood group type of several human Rotavirus strains (RVA) has recently been described. Synthesis of these ligands requires a functional FUT2 enzyme, suggesting that FUT2 null homozygote (ie, nonsecretor) individuals may not be recognized by most human RVA strains. Whereas such individuals represent 20% of the control population, this retrospective study determined that none of 51 patients infected by P[8] rotavirus strains were nonsecretors. The lack of α1,2fucosylated carbohydrate motifs in the gut surface mucosa is thus associated with resistance to symptomatic infection and virus attachment to such motifs is essential to the infection process.


Asunto(s)
Heces/virología , Fucosiltransferasas/genética , Gastroenteritis/genética , Polimorfismo Genético , Infecciones por Rotavirus/genética , Rotavirus/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Resistencia a la Enfermedad/genética , Mucosa Gástrica/virología , Gastroenteritis/virología , Genotipo , Humanos , Lactante , Persona de Mediana Edad , Filogenia , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rotavirus/aislamiento & purificación , Infecciones por Rotavirus/virología , Adulto Joven , Galactósido 2-alfa-L-Fucosiltransferasa
17.
Rev Med Virol ; 23(6): 355-66, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23959967

RESUMEN

Noroviruses (NoVs) are recognized as a leading cause of human gastroenteritis worldwide. Infection occurs following the ingestion of contaminated food or, most often, through direct contact from person to person. However, not all individuals are equally sensitive to these viruses. Indeed, NoVs use glycans of the ABH and Lewis histo-blood group antigen family (HBGAs) as attachment factors. At the epithelial level, the synthesis of these HBGAs requires the action of several glycosyltransferases that are encoded by the ABO, FUT2, and FUT3 genes. The combined polymorphism at these three loci dictates sensitivity to NoV infection because the attachment profile to these glycans varies among strains. Structural analysis of the capsid protein interaction with HBGAs reveals distinct modes of binding for strains of genogroups I and II but high conservation within each genogroup, whereas minor amino acid changes are sufficient to generate modifications of HBGA-binding specificities or affinities. Such modifications therefore induce changes in the spectrum of susceptible individuals. Studies of NoV-HBGA interactions together with phylogenetic analyses and the epidemiologic survey of strains indicate that NoV transmission and evolution depend on both the establishment of herd immunity and the genetic resistance of many individuals, which confers herd innate protection by restricting NoV circulation.


Asunto(s)
Antígenos de Grupos Sanguíneos/genética , Infecciones por Caliciviridae/genética , Susceptibilidad a Enfermedades , Antígenos de Histocompatibilidad/genética , Norovirus/fisiología , Polimorfismo Genético , Receptores Virales/genética , Interacciones Huésped-Patógeno , Humanos , Acoplamiento Viral
18.
Virologie (Montrouge) ; 17(4): 264-277, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31910599

RESUMEN

Norovirus (NoVs) are recognized as a leading cause of human gastroenteritis worldwide. Infection takes place following ingestion of contaminated food or most often through direct contact from person to person. However, not all individuals are equally sensitive to these viruses. Indeed, NoVs use ABH and Lewis glycans of the histo-blood group antigen family (HBGAs) as ligands. At the epithelial level synthesis of these HBGAs requires the action of several glycosyltransferases encoded by the ABO, FUT2 and FUT3 genes. Since the attachment profile to these glycans varies from strain to strain, the combined polymorphism at these three loci dictates sensitivity to NoV infection. Studies of the NoV-HBGA interactions together with phylogenetic analyses and the epidemiologic follow-up of strains indicate that NoVs transmission and evolution depends both on the establishment of herd immunity and on the genetic resistance of many individuals that contributes to restrict NoVs circulation, confering a herd innate proctection.

20.
J Biol Chem ; 287(6): 4335-47, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22170069

RESUMEN

Burkholderia ambifaria is generally associated with the rhizosphere of plants where it has biocontrol effects on other microorganisms. It is also a member of the Burkholderia cepacia complex, a group of closely related bacteria that cause lung infections in immunocompromised patients as well as in patients with granulomatous disease or cystic fibrosis. Our previous work indicated that fucose on human epithelia is a frequent target for lectins and adhesins of lung pathogens (Sulák, O., Cioci, G., Lameignère, E., Balloy, V., Round, A., Gutsche, I., Malinovská, L., Chignard, M., Kosma, P., Aubert, D. F., Marolda, C. L., Valvano, M. A., Wimmerová, M., and Imberty, A. (2011) PLoS Pathog. 7, e1002238). Analysis of the B. ambifaria genome identified BambL as a putative fucose-binding lectin. The 87-amino acid protein was produced recombinantly and demonstrated to bind to fucosylated oligosaccharides with a preference for αFuc1-2Gal epitopes. Crystal structures revealed that it associates as a trimer with two fucose-binding sites per monomer. The overall fold is a six-bladed ß-propeller formed by oligomerization as in the Ralstonia solanacearum lectin and not by sequential domains like the fungal fucose lectin from Aleuria aurantia. The affinity of BambL for small fucosylated glycans is very high as demonstrated by microcalorimetry (K(D) < 1 µM). Plant cell wall oligosaccharides and human histo-blood group oligosaccharides H-type 2 and Lewis Y are bound with equivalent efficiency. Binding to artificial glycosphingolipid-containing vesicles, human saliva, and lung tissues confirmed that BambL could recognize a wide spectrum of fucosylated epitopes, albeit with a lower affinity for biological material from nonsecretor individuals.


Asunto(s)
Proteínas Bacterianas/química , Burkholderia/química , Epítopos/química , Fucosa/química , Lectinas/química , Oligosacáridos/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Burkholderia/metabolismo , Epítopos/metabolismo , Fucosa/metabolismo , Humanos , Lectinas/metabolismo , Oligosacáridos/metabolismo , Unión Proteica , Pliegue de Proteína , Estructura Cuaternaria de Proteína , Rizoma/microbiología , Saliva/química , Saliva/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...