Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Publication year range
1.
Nat Commun ; 13(1): 7075, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36400774

ABSTRACT

Resistance to African trypanosomes in humans relies in part on the high affinity targeting of a trypanosome lytic factor 1 (TLF1) to a trypanosome haptoglobin-hemoglobin receptor (HpHbR). While TLF1 avoidance by the inactivation of HpHbR contributes to Trypanosoma brucei gambiense human infectivity, the evolutionary trade-off of this adaptation is unknown, as the physiological function of the receptor remains to be elucidated. Here we show that uptake of hemoglobin via HpHbR constitutes the sole heme import pathway in the trypanosome bloodstream stage. T. b. gambiense strains carrying the inactivating mutation in HpHbR, as well as genetically engineered T. b. brucei HpHbR knock-out lines show only trace levels of intracellular heme and lack hemoprotein-based enzymatic activities, thereby providing an uncommon example of aerobic parasitic proliferation in the absence of heme. We further show that HpHbR facilitates the developmental progression from proliferating long slender forms to cell cycle-arrested stumpy forms in T. b. brucei. Accordingly, T. b. gambiense was found to be poorly competent for slender-to-stumpy differentiation unless a functional HpHbR receptor derived from T. b. brucei was genetically restored. Altogether, we identify heme-deficient metabolism and disrupted cellular differentiation as two distinct HpHbR-dependent evolutionary trade-offs for T. b. gambiense human infectivity.


Subject(s)
Lipoproteins, HDL , Trypanosoma brucei gambiense , Humans , Trypanosoma brucei gambiense/genetics , Trypanosoma brucei gambiense/metabolism , Lipoproteins, HDL/metabolism , Biological Evolution , Heme/metabolism , Cell Differentiation/genetics
2.
Annu Rev Virol ; 7(1): 385-402, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32991263

ABSTRACT

Dendritic cell (DC) subsets are abundantly present in genital and intestinal mucosal tissue and are among the first innate immune cells that encounter human immunodeficiency virus type 1 (HIV-1) after sexual contact. Although DCs have specific characteristics that greatly enhance HIV-1 transmission, it is becoming evident that most DC subsets also have virus restriction mechanisms that exert selective pressure on the viruses during sexual transmission. In this review we discuss the current concepts of the immediate events following viral exposure at genital mucosal sites that lead to selection of specific HIV-1 variants called transmitted founder (TF) viruses. We highlight the importance of the TF HIV-1 phenotype and the role of different DC subsets in establishing infection. Understanding the biology of HIV-1 transmission will contribute to the design of novel treatment strategies preventing HIV-1 dissemination.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Mucous Membrane/immunology , Dendritic Cells/classification , HIV Infections/virology , Humans , Mucous Membrane/cytology , Reproductive Tract Infections/immunology , Reproductive Tract Infections/virology , Skin/cytology , Skin/immunology , Skin/virology
3.
Front Immunol ; 11: 503, 2020.
Article in English | MEDLINE | ID: mdl-32292405

ABSTRACT

Sexually transmitted Hepatitis C virus (HCV) infections and high reinfections are a major concern amongst men who have sex with men (MSM) living with HIV-1 and HIV-negative MSM. Immune activation and/or HIV-1 coinfection enhance HCV susceptibility via sexual contact, suggesting that changes in immune cells or external factors are involved in increased susceptibility. Activation of anal mucosal Langerhans cells (LCs) has been implicated in increased HCV susceptibility as activated but not immature LCs efficiently retain and transmit HCV to other cells. However, the underlying molecular mechanism of transmission remains unclear. Here we identified the Heparan Sulfate Proteoglycan Syndecan 4 as the molecular switch, controlling HCV transmission by LCs. Syndecan 4 was highly upregulated upon activation of LCs and interference with Heparan Sulfate Proteoglycans or silencing of Syndecan 4 abrogated HCV transmission. These data strongly suggest that Syndecan 4 mediates HCV transmission by activated LCs. Notably, our data also identified the C-type lectin receptor langerin as a restriction factor for HCV infection and transmission. Langerin expression abrogated HCV infection in HCV permissive cells, whereas langerin expression on the Syndecan 4 expressing cell line strongly decreased HCV transmission to a target hepatoma cell line. These data suggest that the balanced interplay between langerin restriction and Syndecan 4 transmission determines HCV dissemination. Silencing of langerin enhanced HCV transmission whereas silencing Syndecan 4 on activated LCs decreased transmission. Blocking Heparan Sulfate Proteoglycans abrogated HCV transmission by LCs ex vivo identifying Heparan Sulfate Proteoglycans and Syndecan 4 as potential targets to prevent sexual transmission of HCV. Thus, our data strongly suggest that the interplay between receptors promotes or restricts transmission and further indicate that Syndecan 4 is the molecular switch controlling HCV susceptibility after sexual contact.


Subject(s)
Antigens, CD/metabolism , HIV Infections/metabolism , HIV-1/physiology , Hepacivirus/physiology , Hepatitis C/metabolism , Langerhans Cells/physiology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Sexually Transmitted Diseases/metabolism , Syndecan-4/metabolism , Antigens, CD/genetics , Cell Differentiation , Cell Line , Coinfection , Disease Transmission, Infectious , Homosexuality, Male , Humans , Lectins, C-Type/genetics , Male , Mannose-Binding Lectins/genetics , RNA, Small Interfering/genetics , Syndecan-4/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL