Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(6): e1012351, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924030

ABSTRACT

AXL+ Siglec-6+ dendritic cells (ASDC) are novel myeloid DCs which can be subdivided into CD11c+ and CD123+ expressing subsets. We showed for the first time that these two ASDC subsets are present in inflamed human anogenital tissues where HIV transmission occurs. Their presence in inflamed tissues was supported by single cell RNA analysis of public databases of such tissues including psoriasis diseased skin and colorectal cancer. Almost all previous studies have examined ASDCs as a combined population. Our data revealed that the two ASDC subsets differ markedly in their functions when compared with each other and to pDCs. Relative to their cell functions, both subsets of blood ASDCs but not pDCs expressed co-stimulatory and maturation markers which were more prevalent on CD11c+ ASDCs, thus inducing more T cell proliferation and activation than their CD123+ counterparts. There was also a significant polarisation of naïve T cells by both ASDC subsets toward Th2, Th9, Th22, Th17 and Treg but less toward a Th1 phenotype. Furthermore, we investigated the expression of chemokine receptors that facilitate ASDCs and pDCs migration from blood to inflamed tissues, their HIV binding receptors, and their interactions with HIV and CD4 T cells. For HIV infection, within 2 hours of HIV exposure, CD11c+ ASDCs showed a trend in more viral transfer to T cells than CD123+ ASDCs and pDCs for first phase transfer. However, for second phase transfer, CD123+ ASDCs showed a trend in transferring more HIV than CD11c+ ASDCs and there was no viral transfer from pDCs. As anogenital inflammation is a prerequisite for HIV transmission, strategies to inhibit ASDC recruitment into inflamed tissues and their ability to transmit HIV to CD4 T cells should be considered.


Subject(s)
Dendritic Cells , HIV Infections , Humans , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/virology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Axl Receptor Tyrosine Kinase , Male , HIV-1/immunology , Female , Myeloid Cells/metabolism , Myeloid Cells/immunology , Middle Aged , Adult
2.
PLoS Pathog ; 20(6): e1012267, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38857290

ABSTRACT

HSV infects keratinocytes in the epidermis of skin via nectin-1. We established a human foreskin explant infection model to investigate HSV entry and spread. HSV1 entry could only be achieved by the topical application of virus via high density microarray projections (HD-MAPs) to the epidermis, which penetrated beyond one third of its thickness, simulating in vivo microtrauma. Rapid lateral spread of HSV1 to a mean of 13 keratinocytes wide occurred after 24 hours and free virus particles were observed between keratinocytes, consistent with an intercellular route of spread. Nectin-1 staining was markedly decreased in foci of infection in the epidermis and in the human keratinocyte HaCaT cell line. Nectin-1 was redistributed, at the protein level, in adjacent uninfected cells surrounding infection, inducible by CCL3, IL-8 (or CXCL8), and possibly CXCL10 and IL-6, thus facilitating spread. These findings provide the first insights into HSV1 entry and spread in human inner foreskin in situ.


Subject(s)
Chemokines , Foreskin , Herpes Simplex , Herpesvirus 1, Human , Keratinocytes , Nectins , Humans , Male , Keratinocytes/virology , Keratinocytes/metabolism , Foreskin/virology , Foreskin/cytology , Nectins/metabolism , Herpes Simplex/virology , Herpes Simplex/metabolism , Chemokines/metabolism , Herpesvirus 1, Human/physiology , Cell Adhesion Molecules/metabolism , Virus Internalization
3.
Cell Rep ; 43(4): 113977, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38512869

ABSTRACT

Next-generation vaccines may be delivered via the skin and mucosa. The stratified squamous epithelium (SSE) represents the outermost layer of the skin (epidermis) and type II mucosa (epithelium). Langerhans cells (LCs) have been considered the sole antigen-presenting cells (APCs) to inhabit the SSE; however, it is now clear that dendritic cells (DCs) are also present. Importantly, there are functional differences in how LCs and DCs take up and process pathogens as well as their ability to activate and polarize T cells, though whether DCs participate in neuroimmune interactions like LCs is yet to be elucidated. A correct definition and functional characterization of APCs in the skin and anogenital tissues are of utmost importance for the design of better vaccines and blocking pathogen transmission. Here, we provide a historical perspective on the evolution of our understanding of the APCs that inhabit the SSE, including a detailed review of the most recent literature.


Subject(s)
Dendritic Cells , Langerhans Cells , Vaccines , Langerhans Cells/immunology , Humans , Dendritic Cells/immunology , Animals , Vaccines/immunology , Mucous Membrane/immunology , Mucous Membrane/cytology , Epithelial Cells/immunology , Skin/immunology
4.
Cytometry A ; 103(11): 851-856, 2023 11.
Article in English | MEDLINE | ID: mdl-37772977

ABSTRACT

There is a great need to understand human immune cells within tissue, where disease manifests and infection occurs. Tissue-resident memory T cells (TRMs) were discovered over a decade ago, there is a great need to understand their role in human disease. We developed a 24-color flow cytometry panel to comprehensively interrogate CD4+ and CD8+ TRMs isolated from human tissues. When interrogating cells within human tissue, enzymatic methods used to liberate cells from within the tissue can cause cleavage of cell surface markers needed to phenotype these cells. Here we carefully select antibody clones and evaluate the effect of enzymatic digestion on the expression of markers relevant to the identification of T cell residency, as well as markers relevant to the activation and immunoregulation status of these cells. We have designed this panel to be applicable across a range of human tissues including skin, intestine, and type II mucosae such as the vagina.


Subject(s)
CD8-Positive T-Lymphocytes , Intestines , Female , Humans , Flow Cytometry , CD4-Positive T-Lymphocytes , Mucous Membrane , Immunologic Memory
5.
Proc Natl Acad Sci U S A ; 120(31): e2217795120, 2023 08.
Article in English | MEDLINE | ID: mdl-37487076

ABSTRACT

The healthy human cornea is a uniquely transparent sensory tissue where immune responses are tightly controlled to preserve vision. The cornea contains immune cells that are widely presumed to be intraepithelial dendritic cells (DCs). Corneal immune cells have diverse cellular morphologies and morphological alterations are used as a marker of inflammation and injury. Based on our imaging of corneal T cells in mice, we hypothesized that many human corneal immune cells commonly defined as DCs are intraepithelial lymphocytes (IELs). To investigate this, we developed functional in vivo confocal microscopy (Fun-IVCM) to investigate cell dynamics in the human corneal epithelium and stroma. We show that many immune cells resident in the healthy human cornea are T cells. These corneal IELs are characterized by rapid, persistent motility and interact with corneal DCs and sensory nerves. Imaging deeper into the corneal stroma, we show that crawling macrophages and rare motile T cells patrol the tissue. Furthermore, we identify altered immune cell behaviors in response to short-term contact lens wear (acute inflammatory stimulus), as well as in individuals with allergy (chronic inflammatory stimulus) that was modulated by therapeutic intervention. These findings redefine current understanding of immune cell subsets in the human cornea and reveal how resident corneal immune cells respond and adapt to chronic and acute stimuli.


Subject(s)
Cornea , Epithelium, Corneal , Animals , Humans , Mice , Afferent Pathways , Inflammation , Intravital Microscopy
7.
Cell Rep ; 40(12): 111385, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130503

ABSTRACT

The initial immune response to HIV determines transmission. However, due to technical limitations we still do not have a comparative map of early mucosal transmission events. By combining RNAscope, cyclic immunofluorescence, and image analysis tools, we quantify HIV transmission signatures in intact human colorectal explants within 2 h of topical exposure. We map HIV enrichment to mucosal dendritic cells (DCs) and submucosal macrophages, but not CD4+ T cells, the primary targets of downstream infection. HIV+ DCs accumulate near and within lymphoid aggregates, which act as early sanctuaries of high viral titers while facilitating HIV passage to the submucosa. Finally, HIV entry induces recruitment and clustering of target cells, facilitating DC- and macrophage-mediated HIV transfer and enhanced infection of CD4+ T cells. These data demonstrate a rapid response to HIV structured to maximize the likelihood of mucosal infection and provide a framework for in situ studies of host-pathogen interactions and immune-mediated pathologies.


Subject(s)
Colorectal Neoplasms , HIV Infections , HIV-1 , CD4-Positive T-Lymphocytes , Colorectal Neoplasms/pathology , Dendritic Cells , Host-Pathogen Interactions , Humans
8.
Mucosal Immunol ; 15(4): 542-550, 2022 04.
Article in English | MEDLINE | ID: mdl-35173293

ABSTRACT

In tissue, mononuclear phagocytes (MNP) are comprised of Langerhans cells, dendritic cells, macrophages and monocyte-derived cells. They are the first immune cells to encounter HIV during transmission and transmit the virus to CD4 T cells as a consequence of their antigen presenting cell function. To understand the role these cells play in transmission, their phenotypic and functional characterisation is important. With advancements in high parameter single cell technologies, new MNPs subsets are continuously being discovered and their definition and classification is in a state of flux. This has important implications for our knowledge of HIV transmission, which requires a deeper understanding to design effective vaccines and better blocking strategies. Here we review the historical research of the role MNPs play in HIV transmission up to the present day and revaluate these studies in the context of our most recent understandings of the MNP system.


Subject(s)
HIV Infections , Macrophages , CD4-Positive T-Lymphocytes , Dendritic Cells , Humans , Langerhans Cells , Mononuclear Phagocyte System , Phagocytes
9.
Cytometry A ; 101(3): 196-202, 2022 03.
Article in English | MEDLINE | ID: mdl-35018731

ABSTRACT

We developed a 25-color flow cytometry panel to comprehensively interrogate innate lymphoid cells (ILC), mucosal-associated invariant T (MAIT) cells, natural killer (NK) cells and γδ T cells in human tissues. The ability to isolate and interrogate these cells from fresh human tissue is crucial in understanding the role these cells play at immune-privileged mucosal surfaces like the intestine in health and disease settings. However, liberating these cells from tissue is extremely challenging as many key surface identification markers are susceptible to enzymatic cleavage. Choosing the correct enzyme-antibody clone combination within a high-parameter panel is, therefore, a critical consideration. Here, we present a comprehensive, in-depth analysis of the effect different common digestive enzyme blends have on key surface markers used to identify these cell types. In addition, we compared multiple antibody clones for surface markers that are highly susceptible to enzymatic cleavage, such as CD127 and NKp44, to achieve the most consistent and superior staining patterns among donors.


Subject(s)
Mucosal-Associated Invariant T Cells , Biomarkers , Flow Cytometry , Humans , Immunity, Innate , Intestines , Killer Cells, Natural
10.
Front Immunol ; 12: 727952, 2021.
Article in English | MEDLINE | ID: mdl-34566985

ABSTRACT

The human intestine contains numerous mononuclear phagocytes (MNP), including subsets of conventional dendritic cells (cDC), macrophages (Mf) and monocytes, each playing their own unique role within the intestinal immune system and homeostasis. The ability to isolate and interrogate MNPs from fresh human tissue is crucial if we are to understand the role of these cells in homeostasis, disease settings and immunotherapies. However, liberating these cells from tissue is problematic as many of the key surface identification markers they express are susceptible to enzymatic cleavage and they are highly susceptible to cell death. In addition, the extraction process triggers immunological activation/maturation which alters their functional phenotype. Identifying the evolving, complex and highly heterogenous repertoire of MNPs by flow cytometry therefore requires careful selection of digestive enzyme blends that liberate viable cells and preserve recognition epitopes involving careful selection of antibody clones to enable analysis and sorting for functional assays. Here we describe a method for the anatomical separation of mucosa and submucosa as well as isolating lymphoid follicles from human jejunum, ileum and colon. We also describe in detail the optimised enzyme digestion methods needed to acquire functionally immature and biologically functional intestinal MNPs. A comprehensive list of screened antibody clones is also presented which allows for the development of high parameter flow cytometry panels to discriminate all currently identified human tissue MNP subsets including pDCs, cDC1, cDC2 (langerin+ and langerin-), newly described DC3, monocytes, Mf1, Mf2, Mf3 and Mf4. We also present a novel method to account for autofluorescent signal from tissue macrophages. Finally, we demonstrate that these methods can successfully be used to sort functional, immature intestinal DCs that can be used for functional assays such as cytokine production assays.


Subject(s)
Cell Separation , Colon/cytology , Flow Cytometry , Ileum/cytology , Intestinal Mucosa/cytology , Jejunum/cytology , Phagocytes/metabolism , Biomarkers/metabolism , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Phagocytes/immunology , Phenotype
11.
Nat Commun ; 12(1): 2147, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846309

ABSTRACT

Tissue mononuclear phagocytes (MNP) are specialised in pathogen detection and antigen presentation. As such they deliver HIV to its primary target cells; CD4 T cells. Most MNP HIV transmission studies have focused on epithelial MNPs. However, as mucosal trauma and inflammation are now known to be strongly associated with HIV transmission, here we examine the role of sub-epithelial MNPs which are present in a diverse array of subsets. We show that HIV can penetrate the epithelial surface to interact with sub-epithelial resident MNPs in anogenital explants and define the full array of subsets that are present in the human anogenital and colorectal tissues that HIV may encounter during sexual transmission. In doing so we identify two subsets that preferentially take up HIV, become infected and transmit the virus to CD4 T cells; CD14+CD1c+ monocyte-derived dendritic cells and langerin-expressing conventional dendritic cells 2 (cDC2).


Subject(s)
Anal Canal/cytology , Antigens, CD/metabolism , Dendritic Cells/metabolism , Genitalia/cytology , HIV-1/physiology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Monocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Shape , Collagenases/metabolism , Dermis/metabolism , HIV Infections/immunology , HIV Infections/virology , Humans , Lipopolysaccharide Receptors/metabolism , Mucous Membrane/metabolism , Phagocytes/metabolism , Phenotype , Receptors, CCR5/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , Transcription, Genetic
12.
PLoS Pathog ; 17(4): e1009536, 2021 04.
Article in English | MEDLINE | ID: mdl-33905459

ABSTRACT

Skin mononuclear phagocytes (MNPs) provide the first interactions of invading viruses with the immune system. In addition to Langerhans cells (LCs), we recently described a second epidermal MNP population, Epi-cDC2s, in human anogenital epidermis that is closely related to dermal conventional dendritic cells type 2 (cDC2) and can be preferentially infected by HIV. Here we show that in epidermal explants topically infected with herpes simplex virus (HSV-1), both LCs and Epi-cDC2s interact with HSV-1 particles and infected keratinocytes. Isolated Epi-cDC2s support higher levels of infection than LCs in vitro, inhibited by acyclovir, but both MNP subtypes express similar levels of the HSV entry receptors nectin-1 and HVEM, and show similar levels of initial uptake. Using inhibitors of endosomal acidification, actin and cholesterol, we found that HSV-1 utilises different entry pathways in each cell type. HSV-1 predominantly infects LCs, and monocyte-derived MNPs, via a pH-dependent pathway. In contrast, Epi-cDC2s are mainly infected via a pH-independent pathway which may contribute to the enhanced infection of Epi-cDC2s. Both cells underwent apoptosis suggesting that Epi-cDC2s may follow the same dermal migration and uptake by dermal MNPs that we have previously shown for LCs. Thus, we hypothesize that the uptake of HSV and infection of Epi-cDC2s will stimulate immune responses via a different pathway to LCs, which in future may help guide HSV vaccine development and adjuvant targeting.


Subject(s)
Herpesvirus 1, Human/physiology , Langerhans Cells/virology , Virus Internalization , Adolescent , Animals , Cells, Cultured , Child , Child, Preschool , Chlorocebus aethiops , Epidermis/pathology , Epidermis/virology , HaCaT Cells , HeLa Cells , Herpes Simplex/pathology , Herpes Simplex/virology , Humans , Infant , Signal Transduction/physiology , Vero Cells
13.
Bioinformatics ; 37(4): 559-567, 2021 05 01.
Article in English | MEDLINE | ID: mdl-32931552

ABSTRACT

MOTIVATION: Autofluorescence is a long-standing problem that has hindered the analysis of images of tissues acquired by fluorescence microscopy. Current approaches to mitigate autofluorescence in tissue are lab-based and involve either chemical treatment of sections or specialized instrumentation and software to 'unmix' autofluorescent signals. Importantly, these approaches are pre-emptive and there are currently no methods to deal with autofluorescence in acquired fluorescence microscopy images. RESULTS: To address this, we developed Autofluorescence Identifier (AFid). AFid identifies autofluorescent pixels as discrete objects in multi-channel images post-acquisition. These objects can then be tagged for exclusion from downstream analysis. We validated AFid using images of FFPE human colorectal tissue stained for common immune markers. Further, we demonstrate its utility for image analysis where its implementation allows the accurate measurement of HIV-Dendritic cell interactions in a colorectal explant model of HIV transmission. Therefore, AFid represents a major leap forward in the extraction of useful data from images plagued by autofluorescence by offering an approach that is easily incorporated into existing workflows and that can be used with various samples, staining panels and image acquisition methods. We have implemented AFid in ImageJ, Matlab and R to accommodate the diverse image analysis community. AVAILABILITY AND IMPLEMENTATION: AFid software is available at https://ellispatrick.github.io/AFid. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Image Processing, Computer-Assisted , Software , Histological Techniques , Humans , Microscopy, Fluorescence , Workflow
14.
Methods Mol Biol ; 2060: 31-56, 2020.
Article in English | MEDLINE | ID: mdl-31617171

ABSTRACT

Herpes simplex viruses (HSV) types 1 and 2 are ubiquitous. They both cause genital herpes, occasionally severe disease in the immunocompromised, and facilitate much HIV acquisition globally. Despite more than 60 years of research, there is no licensed prophylactic HSV vaccine and some doubt as to whether this can be achieved. Nevertheless, a previous HSV vaccine candidate did have partial success in preventing genital herpes and HSV acquisition and another immunotherapeutic candidate reduced viral shedding and recurrent lesions, inspiring further research. However, the entry pathway of HSV into the anogenital mucosa and the subsequent cascade of immune responses need further elucidation so that these responses could be mimicked or improved by a vaccine, to prevent viral entry and colonization of the neuronal ganglia. For an effective novel vaccine against genital herpes the choice of antigen and adjuvant may be critical. The incorporation of adjuvants of the vaccine candidates in the past, may account for their partial efficacy. It is likely that they can be improved by understanding the mechanisms of immune responses elicited by different adjuvants and comparing these to natural immune responses. Here we review the history of vaccines for HSV, those in development and compare them to successful vaccines for chicken pox or herpes zoster. We also review what is known of the natural immune control of herpes lesions, via interacting innate immunity and CD4 and CD8 T cells and the lessons they provide for development of new, more effective vaccines.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Herpes Genitalis , Herpes Simplex Virus Vaccines , Herpes Simplex , Herpesvirus 1, Human/immunology , Herpesvirus 2, Human/immunology , Animals , Herpes Genitalis/immunology , Herpes Genitalis/pathology , Herpes Genitalis/prevention & control , Herpes Simplex/immunology , Herpes Simplex/pathology , Herpes Simplex/prevention & control , Herpes Simplex Virus Vaccines/immunology , Herpes Simplex Virus Vaccines/therapeutic use , Humans
15.
Front Immunol ; 10: 2263, 2019.
Article in English | MEDLINE | ID: mdl-31616434

ABSTRACT

Mononuclear phagocytes are antigen presenting cells that play a key role in linking the innate and adaptive immune systems. In tissue, these consist of Langerhans cells, dendritic cells and macrophages, all of which express the key HIV entry receptors CD4 and CCR5 making them directly infectible with HIV. Mononuclear phagocytes are the first cells of the immune system to interact with invading pathogens such as HIV. Each cell type expresses a specific repertoire of pathogen binding receptors which triggers pathogen uptake and the release of innate immune cytokines. Langerhans cells and dendritic cells migrate to lymph nodes and present antigens to CD4 T cells, whereas macrophages remain tissue resident. Here we review how HIV-1 manipulates these cells by blocking their ability to produce innate immune cytokines and taking advantage of their antigen presenting cell function in order to gain transport to its primary target cells, CD4 T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Phagocytes/immunology , CD4 Antigens/immunology , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Humans , Langerhans Cells/immunology , Langerhans Cells/metabolism , Langerhans Cells/virology , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Phagocytes/metabolism , Phagocytes/virology , Receptors, CCR5/immunology , Receptors, CCR5/metabolism
16.
Nat Commun ; 10(1): 2759, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227717

ABSTRACT

Langerhans cells (LC) are thought to be the only mononuclear phagocyte population in the epidermis where they detect pathogens. Here, we show that CD11c+ dendritic cells (DCs) are also present. These cells are transcriptionally similar to dermal cDC2 but are more efficient antigen-presenting cells. Compared to LCs, epidermal CD11c+ DCs are enriched in anogenital tissues where they preferentially interact with HIV, express the higher levels of HIV entry receptor CCR5, support the higher levels of HIV uptake and replication and are more efficient at transmitting the virus to CD4 T cells. Importantly, these findings are observed using both a lab-adapted and transmitted/founder strain of HIV. We also describe a CD33low cell population, which is transcriptionally similar to LCs but does not appear to function as antigen-presenting cells or acts as HIV target cells. Our findings reveal that epidermal DCs in anogenital tissues potentially play a key role in sexual transmission of HIV.


Subject(s)
Dendritic Cells/virology , Epidermal Cells/virology , HIV Infections/transmission , HIV-1/immunology , Antigen Presentation/immunology , CD11c Antigen/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epidermal Cells/immunology , Epidermal Cells/metabolism , Epidermis/immunology , Female , HIV Infections/immunology , HIV Infections/virology , HIV-1/pathogenicity , Healthy Volunteers , Humans , Male , Primary Cell Culture , Receptors, CCR5/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , T-Lymphocytes/immunology , Virus Internalization
17.
J Leukoc Biol ; 101(6): 1393-1403, 2017 06.
Article in English | MEDLINE | ID: mdl-28270408

ABSTRACT

Mononuclear phagocytes are present in skin and mucosa and represent one of the first lines of defense against invading pathogens, which they detect via an array of pathogen-binding receptors expressed on their surface. However, their extraction from tissue is difficult, and the isolation technique used has functional consequences on the cells obtained. Here, we compare mononuclear phagocytes isolated from human skin using either enzymatic digestion or spontaneous migration. Cells isolated via enzymatic digestion are in an immature state, and all subsets are easily defined. However, cells isolated by spontaneous migration are in a mature state, and CD141 cross-presenting DCs (cDC1) are more difficult to define. Different pathogen-binding receptors are susceptible to cleavage by blends of collagenase, demonstrating that great care must be taken in choosing the correct enzyme blend to digest tissue if carrying out pathogen-interaction assays. Finally, we have optimized mononuclear phagocyte culture conditions to enhance their survival after liberation from the tissue.


Subject(s)
Cell Separation/methods , Enzymes/metabolism , Monocytes/cytology , Phagocytes/cytology , Skin/cytology , Cell Movement , Humans , Monocytes/immunology , Monocytes/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Phenotype , Skin/immunology , Skin/metabolism
18.
Rev Med Virol ; 27(2)2017 03.
Article in English | MEDLINE | ID: mdl-28044388

ABSTRACT

Langerhans cells (LCs) situated in stratified squamous epithelium of the skin and mucosal tissue are amongst the first cells that sexually transmitted pathogens encounter during transmission. They are potent antigen presenting cells and play a key role in the host mounting an appropriate immune response. As such, viruses have evolved complex strategies to manipulate these cells to facilitate successful transmission. One of best studied examples is HIV, which manipulates the natural function of these cells to interact with CD4 T cells, which are the main target cell for HIV in which rapid replication occurs. However, there is controversy in the literature as to the role that LCs play in this process. Langerhans cells also play a key role in the way the body mounts an immune response to HSV, and there is also a complex interplay between the transmission of HSV and HIV that involves LCs. In this article, we review both past and present literatures with a particular focus on a few very recent studies that shed new light on the role that LCs play in the transmission and immune response to these 2 pathogens.


Subject(s)
HIV Infections/transmission , Herpes Genitalis/transmission , Host-Pathogen Interactions , Langerhans Cells/immunology , Langerhans Cells/virology , Humans
19.
Clin Transl Immunology ; 5(7): e94, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27525067

ABSTRACT

Incremental advances in our knowledge of how natural immune control of herpes simplex virus (HSV) develops have yielded insight as to why previous vaccine attempts have only been partially successful, however, our understanding of these pathways, particularly in humans, is still incomplete. Further elucidation of the innate immune events that are responsible for stimulating these effector responses is required to accurately inform vaccine design. An enhanced understanding of the mechanism of action of novel adjuvants will also facilitate the rational choice of adjuvant to optimise such responses. Here we review the reasons for the hitherto partial HSV vaccine success and align these with our current knowledge of how natural HSV immunity develops. In particular, we focus on the innate immune response and the role of dendritic cells in inducing protective T-cell responses and how these pathways might be recapitulated in a vaccine setting.

20.
PLoS Pathog ; 11(4): e1004812, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25875649

ABSTRACT

The mechanism by which immunity to Herpes Simplex Virus (HSV) is initiated is not completely defined. HSV initially infects mucosal epidermis prior to entering nerve endings. In mice, epidermal Langerhans cells (LCs) are the first dendritic cells (DCs) to encounter HSV, but it is CD103(+) dermal DCs that carry viral antigen to lymph nodes for antigen presentation, suggesting DC cross-talk in skin. In this study, we compared topically HSV-1 infected human foreskin explants with biopsies of initial human genital herpes lesions to show LCs are initially infected then emigrate into the dermis. Here, LCs bearing markers of maturation and apoptosis formed large cell clusters with BDCA3(+) dermal DCs (thought to be equivalent to murine CD103(+) dermal DCs) and DC-SIGN(+) DCs/macrophages. HSV-expressing LC fragments were observed inside the dermal DCs/macrophages and the BDCA3(+) dermal DCs had up-regulated a damaged cell uptake receptor CLEC9A. No other infected epidermal cells interacted with dermal DCs. Correspondingly, LCs isolated from human skin and infected with HSV-1 in vitro also underwent apoptosis and were taken up by similarly isolated BDCA3(+) dermal DCs and DC-SIGN(+) cells. Thus, we conclude a viral antigen relay takes place where HSV infected LCs undergo apoptosis and are taken up by dermal DCs for subsequent antigen presentation. This provides a rationale for targeting these cells with mucosal or perhaps intradermal HSV immunization.


Subject(s)
Dendritic Cells/virology , Herpesvirus 1, Human/physiology , Langerhans Cells/virology , Simplexvirus/pathogenicity , Skin/virology , Cell Movement , Flow Cytometry , Humans , Microscopy, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...