15 research outputs found

    Implication de DC-SIGN et DC-SIGNR dans la transmission mère-enfant du VIH-1

    Full text link
    La transmission mère-enfant du VIH-1 (TME) représente le principal mode d’infection chez l’enfant et se produit durant la grossesse (in utero, IU), l’accouchement (intrapartum, IP) ou l’allaitement (postpartum, PP). Les mécanismes qui sous-tendent le passage du VIH-1 à travers le placenta et les muqueuses intestinales du nouveau-né sont encore très peu décrits. « Dendritic cell-specific ICAM-grabbing non-integrin » (DC-SIGN) et son homologue DC-SIGN « related » (DC-SIGNR) sont des récepteurs d’antigènes exprimés au niveau du placenta et capables de capter et de transmettre le VIH-1 aux cellules adjacentes. Ils pourraient donc participer au passage trans placentaire du VIH-1 et le polymorphisme génétique affectant l’expression ou modifiant l’interaction avec le virus aurait une influence sur la TME du VIH-1. Afin d’explorer cette hypothèse, nous avons procédé à une analyse exhaustive du polymorphisme de DC-SIGN et DC-SIGNR dans la population du Zimbabwe. Par la suite, nous avons déterminé l’association entre le polymorphisme de DC-SIGN et DC-SIGNR et la TME du VIH-1 dans une cohorte d’enfants nés de mères VIH-positives à Harare, au Zimbabwe. Enfin, nous avons défini l’impact fonctionnel des mutations associées. Les enfants homozygotes pour les haplotypes H1 et H3 dans le gène de DC-SIGNR sont 4 à 6 fois plus à risque de contracter le VIH-1 par voie IU et IP. H1 et H3 contiennent la mutation du promoteur p-198A et la mutation de l’intron 2, int2-180A, et des études fonctionnelles nous ont permis de démontrer que p-198A diminue l’activité transcriptionnelle du promoteur de DC-SIGNR et l’expression des transcrits d’ARNm dans le placenta, alors que int2-180A modifie le répertoire d’isoformes de DC-SIGNR vers une proportion diminuée d’isoformes membranaires. Les enfants porteurs des haplotypes H4 et H6 de DC-SIGN sont 2 à 6 fois plus à risque de contracter le VIH-1 par voie IU. Ces haplotypes contiennent deux mutations du promoteur (p-336T/C et p-201C/A) et quatre mutations codant pour un changement d’acide aminé dans l’exon 4 (R198Q, E214D, R221Q ou L242V) associées à un risque augmenté de transmission IU, IP et PP du VIH-1. Des études fonctionnelles ont démontré que les mutations du promoteur diminuent l’expression de DC-SIGN dans les macrophages placentaires. Toutefois, l’exposition IU au VIH-1 module le niveau d’expression de DC-SIGN, résultant en des niveaux d’expression similaires entre les macrophages des porteurs des allèles sauvages et mutés. Les mutations de l’exon 4 augmentent l’affinité de DC-SIGN pour le VIH-1 et sa capacité à capturer et à transmettre le virus aux lymphocytes T, favorisant possiblement la dissémination du VIH-1 à travers le placenta. L’association entre les mutations de DC-SIGN et la transmission IP et PP du VIH-1 suggèrent qu’il aurait aussi un rôle à jouer dans les muqueuses intestinales de l’enfant. Notre étude démontre pour la première fois l’implication de DC-SIGN et DC-SIGNR dans la TME du VIH-1. L’augmentation des capacités de capture et de transmission de DC-SIGN résulte en une susceptibilité accrue de l’enfant à l’infection au VIH-1 et concorde avec un rôle dans la dissémination transplacentaire. Toutefois, la diminution préférentielle des transcrits membranaires de DC-SIGNR au placenta augmente la TME du VIH-1 et laisse croire à son implication via un autre mécanisme. Ces mécanismes pourraient aussi s’appliquer à d’autres pathogènes reconnus par DC-SIGN et DC-SIGNR et transmis de la mère à l’enfant.Mother-to-child transmission (MTCT) is the main cause of HIV-1 infection in children worldwide. MTCT of HIV-1 can occur during pregnancy (in utero, IU), delivery (intrapartum, IP) or breastfeeding (postpartum, PP). Dendritic cell-specific ICAM-grabbing non-integrin (DC-SIGN) and its homolog DC-SIGN related (DC-SIGNR) are attachment receptors for HIV-1 and are expressed in the placenta. They have been implicated in viral capture and transmission to T cells. To investigate the potential role of DC-SIGN and DC-SIGNR in MTCT of HIV-1, we carried out a genetic association study in a well-characterized cohort of 197 HIV-infected mothers and their infants recruited in Harare, Zimbabwe. Infants harbouring two copies of DC-SIGNR H1 and/or H3 haplotypes (H1-H1, H1-H3, H3-H3) had a 4-fold increased risk of IU and 6-fold increased risk of IP HIV-1 infection after adjusting for a number of maternal factors. The implicated H1 and H3 haplotypes share two single nucleotide polymorphisms (SNPs) in promoter region (p-198A) and intron 2 (int2-180A) that were associated with increased risk of both IU and IP HIV-1 infection. The promoter variant reduced transcriptional activity in vitro. In homozygous H1 infants bearing both the p-198A and int2-180A mutations, we observed a 4-fold decrease in the level of placental DC-SIGNR transcripts, disproportionately affecting the expression of membrane-bound isoforms compared to infant noncarriers. Infants carrying H4 and H6 haplotypes in DC-SIGN gene were more likely to be HIV-1-infected during pregnancy. These haplotypes contain promoter variants (p-336T/C and p-201C/A) and exon 4 variants (R198Q, E214D, R221Q and L242V) that were all significantly associated with increased risk of MTCT of HIV-1. Compared with wild-type sequence, the promoter variants reduced both the DC-SIGN transcription in vitro and expression (2-fold) in placental macrophages of HIV-1-unexposed infants. However, in HIV-1-exposed infants, the level of DC-SIGN expression in placental macrophages was similar in infants carrying either the promoter wild-type or variant sequences. Exon 4 variants increased HIV-1 capture and transmission to T cells in vitro. Association between DC-SIGN SNPs and HIV-1 IP and PP infection also suggests that DC-SIGN plays an important role in intestinal mucosa. This is the first study reporting on functional impact of DC-SIGN and DC-SIGNR natural polymorphisms on HIV-1 transmission from mother-to-child. Decreased levels of expression of membrane DC-SIGNR isoforms at the placental endothelial cell surface increased child susceptibility to HIV-1. Presence of DC-SIGN variants increasing its affinity for the virus augmented child susceptibility to HIV-1 and may favour viral dissemination across the placental barrier. This study provides compelling evidence to support an important role of DC-SIGN and DC-SIGNR in various modes of MTCT of HIV-1 and shed light on the possible mechanisms involved in HIV-1 passage from mother-to-infant. These findings raise the possibility that similar mechanisms may operate with other human pathogens known to interact with DC-SIGN and DC-SIGNR

    Looking for Evidence of Public Health's Role for Long-Term Evacuees

    Get PDF
    Many Canadians have had personal experience of a major emergency or disaster at some point in their lifetime and close to a third of those affected were evacuated from their homes or communities. Most evacuations have lasted less than 2 weeks, but in some instances, people have been displaced for months or years. For example, hundreds of residents evacuated following flooding in Lake St. Martin, Manitoba in 2011, remain displaced today. In order to learn more about the roles and responses of public health for long-term evacuees (LTEs) in Canada, we conducted a narrative review of published English-language documents, beginning with literature specific to Canada and then expanding to include literature on other high-income countries. We found that while researchers have explored public health considerations in emergency preparedness, acute disaster management, and resettlement in these contexts there is a dearth of published evidence regarding the public health implications of prolonged evacuation and the public health responses to long-term evacuation in Canada and in other high-income countries. Because the public health needs of diverse populations of LTEs have not been fully investigated, it is likely that they are neither well-understood nor adequately addressed in public health policy and practice

    Functional Genetic Variants in DC-SIGNR Are Associated with Mother-to-Child Transmission of HIV-1

    Get PDF
    BACKGROUND: Mother-to-child transmission (MTCT) is the main cause of HIV-1 infection in children worldwide. Given that the C-type lectin receptor, dendritic cell-specific ICAM-grabbing non-integrin-related (DC-SIGNR, also known as CD209L or liver/lymph node-specific ICAM-grabbing non-integrin (L-SIGN)), can interact with pathogens including HIV-1 and is expressed at the maternal-fetal interface, we hypothesized that it could influence MTCT of HIV-1. METHODS AND FINDINGS: To investigate the potential role of DC-SIGNR in MTCT of HIV-1, we carried out a genetic association study of DC-SIGNR in a well-characterized cohort of 197 HIV-infected mothers and their infants recruited in Harare, Zimbabwe. Infants harbouring two copies of DC-SIGNR H1 and/or H3 haplotypes (H1-H1, H1-H3, H3-H3) had a 3.6-fold increased risk of in utero (IU) (P = 0.013) HIV-1 infection and a 5.7-fold increased risk of intrapartum (IP) (P = 0.025) HIV-1 infection after adjusting for a number of maternal factors. The implicated H1 and H3 haplotypes share two single nucleotide polymorphisms (SNPs) in promoter region (p-198A) and intron 2 (int2-180A) that were associated with increased risk of both IU (P = 0.045 and P = 0.003, respectively) and IP (P = 0.025, for int2-180A) HIV-1 infection. The promoter variant reduced transcriptional activity in vitro. In homozygous H1 infants bearing both the p-198A and int2-180A mutations, we observed a 4-fold decrease in the level of placental DC-SIGNR transcripts, disproportionately affecting the expression of membrane-bound isoforms compared to infant noncarriers (P = 0.011). CONCLUSION: These results suggest that DC-SIGNR plays a crucial role in MTCT of HIV-1 and that impaired placental DC-SIGNR expression increases risk of transmission

    HIV-Exposed Seronegative Sex Workers Express Low T-Cell Activation and an Intact Ectocervical Tissue Microenvironment

    No full text
    Immunological correlates of natural resistance to HIV have been identified in HIV-exposed seronegative (HESN) individuals and include a low-inflammatory genital mucosal status. The cervicovaginal epithelium has not been studied for such correlates despite constituting an important barrier against sexual HIV transmission. To fill this gap in knowledge, we collected samples of blood, cervical mononuclear cells, cervicovaginal lavage, and ectocervical tissue from Kenyan HESN sex workers (n = 29) and controls (n = 33). The samples were analyzed by flow cytometry, protein profiling, 16S rRNA gene sequencing, in situ image analysis, and tissue-based RNA sequencing. A significantly higher relative proportion of regulatory T cells in blood (B7+CD25hiFoxP3+CD127loCD4+ and B7+Helios+FoxP3+CD4+), and a significantly lower proportion of activated cervical T cells (CCR5+CD69+CD4+ and CCR5+CD69+CD8+), were found in the HESN group compared with the controls. In contrast, there were no statistically significant differences between the study groups in cervicovaginal protein and microbiome compositions, ectocervical epithelial thickness, E-cadherin expression, HIV receptor expression, and tissue RNA transcriptional profiles. The identification of an intact ectocervical microenvironment in HESN individuals add new data to current knowledge about natural resistance to sexual transmission of HIV

    HIV-Exposed Seronegative Sex Workers Express Low T-Cell Activation and an Intact Ectocervical Tissue Microenvironment

    No full text
    Immunological correlates of natural resistance to HIV have been identified in HIV-exposed seronegative (HESN) individuals and include a low-inflammatory genital mucosal status. The cervicovaginal epithelium has not been studied for such correlates despite constituting an important barrier against sexual HIV transmission. To fill this gap in knowledge, we collected samples of blood, cervical mononuclear cells, cervicovaginal lavage, and ectocervical tissue from Kenyan HESN sex workers (n = 29) and controls (n = 33). The samples were analyzed by flow cytometry, protein profiling, 16S rRNA gene sequencing, in situ image analysis, and tissue-based RNA sequencing. A significantly higher relative proportion of regulatory T cells in blood (B7+CD25hiFoxP3+CD127loCD4+ and B7+Helios+FoxP3+CD4+), and a significantly lower proportion of activated cervical T cells (CCR5+CD69+CD4+ and CCR5+CD69+CD8+), were found in the HESN group compared with the controls. In contrast, there were no statistically significant differences between the study groups in cervicovaginal protein and microbiome compositions, ectocervical epithelial thickness, E-cadherin expression, HIV receptor expression, and tissue RNA transcriptional profiles. The identification of an intact ectocervical microenvironment in HESN individuals add new data to current knowledge about natural resistance to sexual transmission of HIV

    Naturally-Occurring Genetic Variants in Human DC-SIGN Increase HIV-1 Capture, Cell-Transfer and Risk of Mother-To-Child Transmission

    Get PDF
    <div><h3>Background</h3><p>Mother-to-child transmission (MTCT) is the main cause of HIV-1 infection in children worldwide. Dendritic cell–specific ICAM-3 grabbing-nonintegrin (DC-SIGN, also known as CD209) is an HIV-1 receptor that enhances its transmission to T cells and is expressed on placental macrophages.</p> <h3>Methods and Findings</h3><p>We have investigated the association between DC-SIGN genetic variants and risk of MTCT of HIV-1 among Zimbabwean infants and characterized the impact of the associated mutations on DC-SIGN expression and interaction with HIV-1. DC-SIGN promoter (p-336C and p-201A) and exon 4 (198Q and 242V) variants were all significantly associated with increased risk of intrauterine (IU) HIV-1 infection. Promoter variants decreased DC-SIGN expression both in vitro and in placental CD163<sup>+</sup> macrophages (Hofbauer cells) of HIV-1 unexposed infants but not of HIV-1 exposed infants. The exon 4 protein-modifying mutations increased HIV-1 capture and transmission to T cells in vitro.</p> <h3>Conclusion</h3><p>This study provides compelling evidence to support an important role of DC-SIGN in IU HIV-1 infection.</p> </div
    corecore