Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Publication year range
2.
Nat Immunol ; 20(7): 824-834, 2019 07.
Article in English | MEDLINE | ID: mdl-31209403

ABSTRACT

Multiple genome-wide studies have identified associations between outcome of human immunodeficiency virus (HIV) infection and polymorphisms in and around the gene encoding the HIV co-receptor CCR5, but the functional basis for the strongest of these associations, rs1015164A/G, is unknown. We found that rs1015164 marks variation in an activating transcription factor 1 binding site that controls expression of the antisense long noncoding RNA (lncRNA) CCR5AS. Knockdown or enhancement of CCR5AS expression resulted in a corresponding change in CCR5 expression on CD4+ T cells. CCR5AS interfered with interactions between the RNA-binding protein Raly and the CCR5 3' untranslated region, protecting CCR5 messenger RNA from Raly-mediated degradation. Reduction in CCR5 expression through inhibition of CCR5AS diminished infection of CD4+ T cells with CCR5-tropic HIV in vitro. These data represent a rare determination of the functional importance of a genome-wide disease association where expression of a lncRNA affects HIV infection and disease progression.


Subject(s)
Gene Expression Regulation , Genetic Variation , HIV Infections/genetics , HIV Infections/virology , HIV-1 , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , Receptors, CCR5/genetics , 3' Untranslated Regions , Alleles , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Membrane/metabolism , Genes, Reporter , Genotype , HIV Infections/metabolism , Humans , Linkage Disequilibrium , Polymorphism, Single Nucleotide , Population Groups/genetics , Prognosis , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR5/metabolism , Viral Load
3.
Nature ; 620(7976): 1025-1030, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532928

ABSTRACT

HIV-1 remains a global health crisis1, highlighting the need to identify new targets for therapies. Here, given the disproportionate HIV-1 burden and marked human genome diversity in Africa2, we assessed the genetic determinants of control of set-point viral load in 3,879 people of African ancestries living with HIV-1 participating in the international collaboration for the genomics of HIV3. We identify a previously undescribed association signal on chromosome 1 where the peak variant associates with an approximately 0.3 log10-transformed copies per ml lower set-point viral load per minor allele copy and is specific to populations of African descent. The top associated variant is intergenic and lies between a long intergenic non-coding RNA (LINC00624) and the coding gene CHD1L, which encodes a helicase that is involved in DNA repair4. Infection assays in iPS cell-derived macrophages and other immortalized cell lines showed increased HIV-1 replication in CHD1L-knockdown and CHD1L-knockout cells. We provide evidence from population genetic studies that Africa-specific genetic variation near CHD1L associates with HIV replication in vivo. Although experimental studies suggest that CHD1L is able to limit HIV infection in some cell types in vitro, further investigation is required to understand the mechanisms underlying our observations, including any potential indirect effects of CHD1L on HIV spread in vivo that our cell-based assays cannot recapitulate.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Genetic Variation , HIV Infections , HIV-1 , Viral Load , Humans , Cell Line , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HIV Infections/genetics , HIV-1/growth & development , HIV-1/physiology , Viral Load/genetics , Africa , Chromosomes, Human, Pair 1/genetics , Alleles , RNA, Long Noncoding/genetics , Virus Replication
4.
Nat Immunol ; 16(6): 577-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25988890

ABSTRACT

The outcome after infection with the human immunodeficiency virus type 1 (HIV-1) is a complex phenotype determined by interactions among the pathogen, the human host and the surrounding environment. An impact of host genetic variation on HIV-1 susceptibility was identified early in the pandemic, with a major role attributed to the genes encoding class I human leukocyte antigens (HLA) and the chemokine receptor CCR5. Studies using genome-wide data sets have underscored the strength of these associations relative to variants located throughout the rest of the genome. However, the extent to which additional polymorphisms influence HIV-1 disease progression, and how much of the variability in outcome can be attributed to host genetics, remain largely unclear. Here we discuss findings concerning the functional impact of associated variants, outline methods for quantifying the host genetic component and examine how available genome-wide data sets may be leveraged to discover gene variants that affect the outcome of HIV-1 infection.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , HLA Antigens/genetics , Host-Pathogen Interactions/genetics , Receptors, CCR5/genetics , Animals , Disease Progression , Gene-Environment Interaction , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , HIV Infections/genetics , Humans
5.
Nat Rev Genet ; 22(10): 645-657, 2021 10.
Article in English | MEDLINE | ID: mdl-34168330

ABSTRACT

Over the past four decades, research on the natural history of HIV infection has described how HIV wreaks havoc on human immunity and causes AIDS. HIV host genomic research, which aims to understand how human genetic variation affects our response to HIV infection, has progressed from early candidate gene studies to recent multi-omic efforts, benefiting from spectacular advances in sequencing technology and data science. In addition to invading cells and co-opting the host machinery for replication, HIV also stably integrates into our own genome. The study of the complex interactions between the human and retroviral genomes has improved our understanding of pathogenic mechanisms and suggested novel preventive and therapeutic approaches against HIV infection.


Subject(s)
Genetic Variation , Genome, Human , HIV Infections/pathology , HIV-1/pathogenicity , Host-Pathogen Interactions , HIV Infections/genetics , HIV Infections/virology , Humans
6.
J Virol ; 97(10): e0095423, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37747237

ABSTRACT

IMPORTANCE: It has been previously shown that genetic variants near CHD1L on chromosome 1 are associated with reduced HIV VL in African populations. However, the impact of these variants on viral diversity and how they restrict viral replication are unknown. We report on a regional association analysis in a South African population and show evidence of selective pressure by variants near CHD1L on HIV RT and gag. Our findings provide further insight into how genetic variability at this locus contributes to host control of HIV in a South African population.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Genetic Loci , Genetic Variation , HIV Infections , HIV-1 , Humans , DNA Helicases/genetics , DNA-Binding Proteins/genetics , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , South Africa , Viral Load/genetics , Virus Replication , HIV Reverse Transcriptase/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism
8.
Proc Natl Acad Sci U S A ; 116(3): 944-949, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30602460

ABSTRACT

Genetic variation in the peptide-binding groove of the highly polymorphic HLA class I molecules has repeatedly been associated with HIV-1 control and progression to AIDS, accounting for up to 12% of the variation in HIV-1 set point viral load (spVL). This suggests a key role in disease control for HLA presentation of HIV-1 epitopes to cytotoxic T cells. However, a comprehensive understanding of the relevant HLA-bound HIV epitopes is still elusive. Here we describe a peptidome-wide association study (PepWAS) approach that integrates HLA genotypes and spVL data from 6,311 HIV-infected patients to interrogate the entire HIV-1 proteome (3,252 unique peptides) for disease-relevant peptides. This PepWAS approach predicts a core set of epitopes associated with spVL, including known epitopes but also several previously uncharacterized disease-relevant peptides. More important, each patient presents only a small subset of these predicted core epitopes through their individual HLA-A and HLA-B variants. Eventually, the individual differences in these patient-specific epitope repertoires account for the variation in spVL that was previously associated with HLA genetic variation. PepWAS thus enables a comprehensive functional interpretation of the robust but little-understood association between HLA and HIV-1 control, prioritizing a short list of disease-associated epitopes for the development of targeted therapy.


Subject(s)
Acquired Immunodeficiency Syndrome , Antigens, Viral , Epitopes , Genetic Variation , HIV-1 , HLA Antigens , Proteome , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Epitopes/genetics , Epitopes/immunology , Genome-Wide Association Study , HIV-1/genetics , HIV-1/immunology , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Proteome/genetics , Proteome/immunology
9.
Mol Biol Evol ; 37(3): 639-650, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31651980

ABSTRACT

Pathogen-mediated balancing selection is regarded as a key driver of host immunogenetic diversity. A hallmark for balancing selection in humans is the heterozygote advantage at genes of the human leukocyte antigen (HLA), resulting in improved HIV-1 control. However, the actual mechanism of the observed heterozygote advantage is still elusive. HLA heterozygotes may present a broader array of antigenic viral peptides to immune cells, possibly resulting in a more efficient cytotoxic T-cell response. Alternatively, heterozygosity may simply increase the chance to carry the most protective HLA alleles, as individual HLA alleles are known to differ substantially in their association with HIV-1 control. Here, we used data from 6,311 HIV-1-infected individuals to explore the relative contribution of quantitative and qualitative aspects of peptide presentation in HLA heterozygote advantage against HIV. Screening the entire HIV-1 proteome, we observed that heterozygous individuals exhibited a broader array of HIV-1 peptides presented by their HLA class I alleles. In addition, viral load was negatively correlated with the breadth of the HIV-1 peptide repertoire bound by an individual's HLA variants, particularly at HLA-B. This suggests that heterozygote advantage at HLA-B is at least in part mediated by quantitative peptide presentation. We also observed higher HIV-1 sequence diversity among HLA-B heterozygous individuals, suggesting stronger evolutionary pressure from HLA heterozygosity. However, HLA heterozygotes were also more likely to carry certain HLA alleles, including the highly protective HLA-B*57:01 variant, indicating that HLA heterozygote advantage ultimately results from a combination of quantitative and qualitative effects in antigen presentation.


Subject(s)
HIV Infections/immunology , HIV-1/physiology , HLA Antigens/genetics , Peptides/immunology , Viral Proteins/chemistry , Antigen Presentation , Genetic Variation , Genome, Viral , HIV Infections/genetics , HIV Infections/virology , HIV-1/immunology , HLA Antigens/immunology , HLA-B Antigens/genetics , Heterozygote , Humans , Viral Load
10.
Haematologica ; 106(8): 2233-2241, 2021 08 01.
Article in English | MEDLINE | ID: mdl-32675224

ABSTRACT

Human immunodeficiency virus (HIV) infection is associated with an increased risk of non-Hodgkin lymphoma (NHL). Even in the era of suppressive antiretroviral treatment, HIV-infected individuals remain at higher risk of developing NHL compared to the general population. To identify potential genetic risk loci, we performed case-control genome-wide association studies and a meta-analysis across three cohorts of HIV+ patients of European ancestry, including a total of 278 cases and 1924 matched controls. We observed a significant association with NHL susceptibility in the C-X-C motif chemokine ligand 12 (CXCL12) region on chromosome 10. A fine mapping analysis identified rs7919208 as the most likely causal variant (P = 4.77e-11), with the G>A polymorphism creating a new transcription factor binding site for BATF and JUND. These results suggest a modulatory role of CXCL12 regulation in the increased susceptibility to NHL observed in the HIV-infected population.


Subject(s)
HIV Infections , Lymphoma, AIDS-Related , Lymphoma, Non-Hodgkin , Anti-Retroviral Agents/therapeutic use , Case-Control Studies , Chemokine CXCL12 , Genome-Wide Association Study , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/genetics , Humans , Lymphoma, AIDS-Related/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/genetics , Polymorphism, Genetic
11.
Hum Genet ; 139(6-7): 865-875, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32409920

ABSTRACT

Genome-wide association studies (GWAS) have been successful in identifying and confirming novel genetic variants that are associated with diverse HIV phenotypes. However, these studies have predominantly focused on European cohorts. HLA molecules have been consistently associated with HIV outcomes, some of which have been found to be population specific, underscoring the need for diversity in GWAS. Recently, there has been a concerted effort to address this gap that leads to health care (disease prevention, diagnosis, treatment) disparities with marginal improvement. As precision medicine becomes more utilized, non-European individuals will be more and more disadvantaged, as the genetic variants identified in genomic research based on European populations may not accurately reflect that of non-European individuals. Leveraging pre-existing, large, multiethnic cohorts, such as the UK Biobank, 23andMe, and the National Institute of Health's All of Us Research Program, can contribute in raising genomic research in non-European populations and ultimately lead to better health outcomes.


Subject(s)
Genetic Markers , Genetic Predisposition to Disease , Genetics, Population , HIV Infections/genetics , HIV/genetics , Human Genetics , Polymorphism, Single Nucleotide , Genome-Wide Association Study , HIV/isolation & purification , HIV Infections/epidemiology , HIV Infections/virology , Humans
12.
Proc Natl Acad Sci U S A ; 114(31): 8342-8347, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28716935

ABSTRACT

Viral respiratory infections are usually mild and self-limiting; still they exceptionally result in life-threatening infections in previously healthy children. To investigate a potential genetic cause, we recruited 120 previously healthy children requiring support in intensive care because of a severe illness caused by a respiratory virus. Using exome and transcriptome sequencing, we identified and characterized three rare loss-of-function variants in IFIH1, which encodes an RIG-I-like receptor involved in the sensing of viral RNA. Functional testing of the variants IFIH1 alleles demonstrated that the resulting proteins are unable to induce IFN-ß, are intrinsically less stable than wild-type IFIH1, and lack ATPase activity. In vitro assays showed that IFIH1 effectively restricts replication of human respiratory syncytial virus and rhinoviruses. We conclude that IFIH1 deficiency causes a primary immunodeficiency manifested in extreme susceptibility to common respiratory RNA viruses.


Subject(s)
Genetic Predisposition to Disease/genetics , Immunologic Deficiency Syndromes/genetics , Interferon-Induced Helicase, IFIH1/genetics , Interferon-beta/biosynthesis , Respiratory Syncytial Viruses/immunology , Respiratory Tract Infections/virology , Rhinovirus/immunology , Adenosine Triphosphatases/genetics , Child, Preschool , Critical Care , Female , Genetic Variation/genetics , Humans , Immunologic Deficiency Syndromes/immunology , Infant , Infant, Newborn , Interferon-beta/immunology , Male , Prospective Studies , Protein Isoforms/genetics , Respiratory Tract Infections/immunology , Virus Replication/immunology
13.
Am J Hum Genet ; 97(5): 738-43, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26456283

ABSTRACT

The magnitude of the human antibody response to viral antigens is highly variable. To explore the human genetic contribution to this variability, we performed genome-wide association studies of the immunoglobulin G response to 14 pathogenic viruses in 2,363 immunocompetent adults. Significant associations were observed in the major histocompatibility complex region on chromosome 6 for influenza A virus, Epstein-Barr virus, JC polyomavirus, and Merkel cell polyomavirus. Using local imputation and fine mapping, we identified specific amino acid residues in human leucocyte antigen (HLA) class II proteins as the most probable causal variants underlying these association signals. Common HLA-DRß1 haplotypes showed virus-specific patterns of humoral-response regulation. We observed an overlap between variants affecting the humoral response to influenza A and EBV and variants previously associated with autoimmune diseases related to these viruses. The results of this study emphasize the central and pathogen-specific role of HLA class II variation in the modulation of humoral immune response to viral antigens in humans.


Subject(s)
Autoimmune Diseases/immunology , Histocompatibility Antigens Class II/genetics , Immunity, Humoral/immunology , Immunocompromised Host/immunology , Polymorphism, Single Nucleotide/genetics , Schizophrenia/immunology , Virus Diseases/immunology , Viruses/immunology , Adult , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Case-Control Studies , Female , Follow-Up Studies , Genome-Wide Association Study , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin G/blood , Male , Prognosis , Protein Conformation , Schizophrenia/genetics , Schizophrenia/pathology , Virus Diseases/genetics , Virus Diseases/virology
14.
PLoS Comput Biol ; 13(2): e1005339, 2017 02.
Article in English | MEDLINE | ID: mdl-28182649

ABSTRACT

We evaluated the fraction of variation in HIV-1 set point viral load attributable to viral or human genetic factors by using joint host/pathogen genetic data from 541 HIV infected individuals. We show that viral genetic diversity explains 29% of the variation in viral load while host factors explain 8.4%. Using a joint model including both host and viral effects, we estimate a total of 30% heritability, indicating that most of the host effects are reflected in viral sequence variation.


Subject(s)
Genome, Human/genetics , Genome, Viral/genetics , HIV Infections/genetics , HIV/genetics , Models, Genetic , Viral Load/genetics , AIDS Vaccines/therapeutic use , Genetic Association Studies/methods , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , HIV Infections/therapy , Host-Pathogen Interactions/genetics , Humans
15.
J Biomed Inform ; 79: 1-6, 2018 03.
Article in English | MEDLINE | ID: mdl-29331453

ABSTRACT

PURPOSE: Protecting patient privacy is a major obstacle for the implementation of genomic-based medicine. Emerging privacy-enhancing technologies can become key enablers for managing sensitive genetic data. We studied physicians' attitude toward this kind of technology in order to derive insights that might foster their future adoption for clinical care. METHODS: We conducted a questionnaire-based survey among 55 physicians of the Swiss HIV Cohort Study who tested the first implementation of a privacy-preserving model for delivering genomic test results. We evaluated their feedback on three different aspects of our model: clinical utility, ability to address privacy concerns and system usability. RESULTS: 38/55 (69%) physicians participated in the study. Two thirds of them acknowledged genetic privacy as a key aspect that needs to be protected to help building patient trust and deploy new-generation medical information systems. All of them successfully used the tool for evaluating their patients' pharmacogenomics risk and 90% were happy with the user experience and the efficiency of the tool. Only 8% of physicians were unsatisfied with the level of information and wanted to have access to the patient's actual DNA sequence. CONCLUSION: This survey, although limited in size, represents the first evaluation of privacy-preserving models for genomic-based medicine. It has allowed us to derive unique insights that will improve the design of these new systems in the future. In particular, we have observed that a clinical information system that uses homomorphic encryption to provide clinicians with risk information based on sensitive genetic test results can offer information that clinicians feel sufficient for their needs and appropriately respectful of patients' privacy. The ability of this kind of systems to ensure strong security and privacy guarantees and to provide some analytics on encrypted data has been assessed as a key enabler for the management of sensitive medical information in the near future. Providing clinically relevant information to physicians while protecting patients' privacy in order to comply with regulations is crucial for the widespread use of these new technologies.


Subject(s)
Computer Security , Confidentiality , HIV Infections/genetics , HIV Infections/therapy , Adult , Aged , Cohort Studies , Electronic Health Records , Female , Genetic Variation , Genomics , Genotype , Humans , Male , Medical Informatics , Middle Aged , Physicians , Software , Surveys and Questionnaires , Switzerland , User-Computer Interface
16.
Proc Natl Acad Sci U S A ; 112(47): 14658-63, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26553974

ABSTRACT

Previous genome-wide association studies (GWAS) of HIV-1-infected populations have been underpowered to detect common variants with moderate impact on disease outcome and have not assessed the phenotypic variance explained by genome-wide additive effects. By combining the majority of available genome-wide genotyping data in HIV-infected populations, we tested for association between ∼8 million variants and viral load (HIV RNA copies per milliliter of plasma) in 6,315 individuals of European ancestry. The strongest signal of association was observed in the HLA class I region that was fully explained by independent effects mapping to five variable amino acid positions in the peptide binding grooves of the HLA-B and HLA-A proteins. We observed a second genome-wide significant association signal in the chemokine (C-C motif) receptor (CCR) gene cluster on chromosome 3. Conditional analysis showed that this signal could not be fully attributed to the known protective CCR5Δ32 allele and the risk P1 haplotype, suggesting further causal variants in this region. Heritability analysis demonstrated that common human genetic variation-mostly in the HLA and CCR5 regions-explains 25% of the variability in viral load. This study suggests that analyses in non-European populations and of variant classes not assessed by GWAS should be priorities for the field going forward.


Subject(s)
Genetic Predisposition to Disease , HIV-1/genetics , Host-Pathogen Interactions/genetics , Polymorphism, Single Nucleotide/genetics , Viral Load/genetics , Adult , Alleles , Amino Acids/genetics , Chromosomes, Human, Pair 3/genetics , Genome-Wide Association Study , HLA-B Antigens/genetics , Humans , Inheritance Patterns/genetics , Physical Chromosome Mapping , Receptors, CCR5/genetics
17.
J Infect Dis ; 216(9): 1063-1069, 2017 11 27.
Article in English | MEDLINE | ID: mdl-28968755

ABSTRACT

Background: Previous genetic association studies of human immunodeficiency virus-1 (HIV-1) progression have focused on common human genetic variation ascertained through genome-wide genotyping. Methods: We sought to systematically assess the full spectrum of functional variation in protein coding gene regions on HIV-1 progression through exome sequencing of 1327 individuals. Genetic variants were tested individually and in aggregate across genes and gene sets for an influence on HIV-1 viral load. Results: Multiple single variants within the major histocompatibility complex (MHC) region were observed to be strongly associated with HIV-1 outcome, consistent with the known impact of classical HLA alleles. However, no single variant or gene located outside of the MHC region was significantly associated with HIV progression. Set-based association testing focusing on genes identified as being essential for HIV replication in genome-wide small interfering RNA (siRNA) and clustered regularly interspaced short palindromic repeats (CRISPR) studies did not reveal any novel associations. Conclusions: These results suggest that exonic variants with large effect sizes are unlikely to have a major contribution to host control of HIV infection.


Subject(s)
Exome Sequencing , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions/genetics , Viral Load/genetics , Adult , Female , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
18.
Ann Hum Genet ; 81(1): 27-34, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28084001

ABSTRACT

Common single-nucleotide variation in the host accounts for 25% of the variability in the plasma levels of HIV during the clinical latency stage (viral load set point). However, the role of rare variants and copy number variants remains relatively unexplored. Previous work has suggested copy number variation of a cluster of ß-defensin genes affects HIV load in treatment-naïve sub-Saharan Africans and rate of response to antiretroviral treatment. Here we analyse a total of 1827 individuals from two cohorts of HIV-infected individuals from Europe and sub-Saharan Africa to investigate the role of ß-defensin copy number variation on HIV load at set point. We find no evidence for association of copy number with viral load. We also compare distribution of ß-defensin copy number between European cases and controls and find no differences, arguing against a role of ß-defensin copy number in HIV acquisition. Taken together, our data argue against an effect of copy number variation of the ß-defensin region in the spontaneous control of HIV infection.


Subject(s)
Acquired Immunodeficiency Syndrome/genetics , HIV-1/physiology , beta-Defensins/genetics , Acquired Immunodeficiency Syndrome/virology , Adult , DNA Copy Number Variations , Disease Progression , Female , Gene Dosage , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Viral Load
19.
PLoS Biol ; 12(9): e1001951, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25226169

ABSTRACT

In ecology, "disease tolerance" is defined as an evolutionary strategy of hosts against pathogens, characterized by reduced or absent pathogenesis despite high pathogen load. To our knowledge, tolerance has to date not been quantified and disentangled from host resistance to disease in any clinically relevant human infection. Using data from the Swiss HIV Cohort Study, we investigated if there is variation in tolerance to HIV in humans and if this variation is associated with polymorphisms in the human genome. In particular, we tested for associations between tolerance and alleles of the Human Leukocyte Antigen (HLA) genes, the CC chemokine receptor 5 (CCR5), the age at which individuals were infected, and their sex. We found that HLA-B alleles associated with better HIV control do not confer tolerance. The slower disease progression associated with these alleles can be fully attributed to the extent of viral load reduction in carriers. However, we observed that tolerance significantly varies across HLA-B genotypes with a relative standard deviation of 34%. Furthermore, we found that HLA-B homozygotes are less tolerant than heterozygotes. Lastly, tolerance was observed to decrease with age, resulting in a 1.7-fold difference in disease progression between 20 and 60-y-old individuals with the same viral load. Thus, disease tolerance is a feature of infection with HIV, and the identification of the mechanisms involved may pave the way to a better understanding of pathogenesis.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , HLA-B Antigens/immunology , Immune Tolerance/genetics , Immunity, Innate/genetics , Polymorphism, Genetic , Adult , Age Factors , Alleles , CD4 Lymphocyte Count , Cohort Studies , Disease Progression , Female , Gene Frequency , Genome, Human , HIV Infections/genetics , HIV Infections/pathology , HIV Infections/virology , HLA-B Antigens/genetics , Heterozygote , Homozygote , Host-Pathogen Interactions , Humans , Male , Middle Aged , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Sex Factors , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/virology , Viral Load
20.
PLoS Pathog ; 10(7): e1004146, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25033082

ABSTRACT

Dendritic cells (DCs) are essential in order to combat invading viruses and trigger antiviral responses. Paradoxically, in the case of HIV-1, DCs might contribute to viral pathogenesis through trans-infection, a mechanism that promotes viral capture and transmission to target cells, especially after DC maturation. In this review, we highlight recent evidence identifying sialyllactose-containing gangliosides in the viral membrane and the cellular lectin Siglec-1 as critical determinants for HIV-1 capture and storage by mature DCs and for DC-mediated trans-infection of T cells. In contrast, DC-SIGN, long considered to be the main receptor for DC capture of HIV-1, plays a minor role in mature DC-mediated HIV-1 capture and trans-infection.


Subject(s)
Dendritic Cells/immunology , Glycolipids/immunology , HIV Infections/immunology , HIV-1/immunology , Sialic Acid Binding Ig-like Lectin 1/immunology , T-Lymphocytes/immunology , Animals , Dendritic Cells/pathology , Dendritic Cells/virology , HIV Infections/pathology , Humans , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL