Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.753
Filter
1.
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
3.
Clin Exp Dermatol ; 49(5): 450-458, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38173286

ABSTRACT

The CD1 and MR1 protein families present lipid antigens and small molecules to T cells, complementing well-studied major histocompatibility complex-peptide mechanisms. The CD1a subtype is highly and continuously expressed within the skin, most notably on Langerhans cells, and has been demonstrated to present self and foreign lipids to T cells, highlighting its cutaneous sentinel role. Alteration of CD1a-dependent T-cell responses has recently been discovered to contribute to the pathogenesis of several inflammatory skin diseases. In this review, we overview the structure and role of CD1a and outline the current evidence implicating CD1a in the development of psoriasis, atopic dermatitis and allergic contact dermatitis.


Subject(s)
Antigens, CD1 , Skin Diseases , T-Lymphocytes , Humans , Antigens, CD1/metabolism , Antigens, CD1/immunology , Dermatitis, Allergic Contact/immunology , Dermatitis, Atopic/immunology , Langerhans Cells/immunology , Psoriasis/immunology , Skin/immunology , Skin/pathology , T-Lymphocytes/immunology , Skin Diseases/drug therapy , Skin Diseases/metabolism , Skin Diseases/pathology
4.
Elife ; 112022 10 12.
Article in English | MEDLINE | ID: mdl-36223176

ABSTRACT

Skin is an active immune organ where professional antigen-presenting cells such as epidermal Langerhans cells (LCs) link innate and adaptive immune responses. While Reticulon 1A (RTN1A) was recently identified in LCs and dendritic cells in cutaneous and lymphoid tissues of humans and mice, its function is still unclear. Here, we studied the involvement of this protein in cytoskeletal remodeling and immune responses toward pathogens by stimulation of Toll-like receptors (TLRs) in resident LCs (rLCs) and emigrated LCs (eLCs) in human epidermis ex vivo and in a transgenic THP-1 RTN1A+ cell line. Hampering RTN1A functionality through an inhibitory antibody induced significant dendrite retraction of rLCs and inhibited their emigration. Similarly, expression of RTN1A in THP-1 cells significantly altered their morphology, enhanced aggregation potential, and inhibited the Ca2+ flux. Differentiated THP-1 RTN1A+ macrophages exhibited long cell protrusions and a larger cell body size in comparison to wild-type cells. Further, stimulation of epidermal sheets with bacterial lipoproteins (TLR1/2 and TLR2 agonists) and single-stranded RNA (TLR7 agonist) resulted in the formation of substantial clusters of rLCs and a significant decrease of RTN1A expression in eLCs. Together, our data indicate involvement of RTN1A in dendrite dynamics and structural plasticity of primary LCs. Moreover, we discovered a relation between activation of TLRs, clustering of LCs, and downregulation of RTN1A within the epidermis, thus indicating an important role of RTN1A in LC residency and maintaining tissue homeostasis.


Subject(s)
Dendrites , Langerhans Cells , Nerve Tissue Proteins , Animals , Dendrites/immunology , Epidermis/metabolism , Humans , Immunity , Langerhans Cells/immunology , Lipoproteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , RNA/metabolism , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptors/metabolism
5.
Cell Rep ; 40(1): 111032, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35793635

ABSTRACT

How mechanistic target of rapamycin complex 1 (mTORC1), a key regulator of cellular metabolism, affects dendritic cell (DC) metabolism and T cell-priming capacity has primarily been investigated in vitro, but how mTORC1 regulates this in vivo remains poorly defined. Here, using mice deficient for mTORC1 component raptor in DCs, we find that loss of mTORC1 negatively affects glycolytic and fatty acid metabolism and maturation of conventional DCs, particularly cDC1s. Nonetheless, antigen-specific CD8+ T cell responses to infection are not compromised and are even enhanced following skin immunization. This is associated with increased activation of Langerhans cells and a subpopulation of EpCAM-expressing cDC1s, of which the latter show an increased physical interaction with CD8+ T cells in situ. Together, this work reveals that mTORC1 limits CD8+ T cell priming in vivo by differentially orchestrating the metabolism and immunogenicity of distinct antigen-presenting cell subsets, which may have implications for clinical use of mTOR inhibitors.


Subject(s)
CD8-Positive T-Lymphocytes , Mechanistic Target of Rapamycin Complex 1 , Skin , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Langerhans Cells/immunology , Langerhans Cells/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Signal Transduction , Skin/immunology , Skin/metabolism
6.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046040

ABSTRACT

Inflammatory pain, such as hypersensitivity resulting from surgical tissue injury, occurs as a result of interactions between the immune and nervous systems with the orchestrated recruitment and activation of tissue-resident and circulating immune cells to the site of injury. Our previous studies identified a central role for Ly6Clow myeloid cells in the pathogenesis of postoperative pain. We now show that the chemokines CCL17 and CCL22, with their cognate receptor CCR4, are key mediators of this response. Both chemokines are up-regulated early after tissue injury by skin-resident dendritic and Langerhans cells to act on peripheral sensory neurons that express CCR4. CCL22, and to a lesser extent CCL17, elicit acute mechanical and thermal hypersensitivity when administered subcutaneously; this response abrogated by pharmacological blockade or genetic silencing of CCR4. Electrophysiological assessment of dissociated sensory neurons from naïve and postoperative mice showed that CCL22 was able to directly activate neurons and enhance their excitability after injury. These responses were blocked using C 021 and small interfering RNA (siRNA)-targeting CCR4. Finally, our data show that acute postoperative pain is significantly reduced in mice lacking CCR4, wild-type animals treated with CCR4 antagonist/siRNA, as well as transgenic mice depleted of dendritic cells. Together, these results suggest an essential role for the peripheral CCL17/22:CCR4 axis in the genesis of inflammatory pain via direct communication between skin-resident dendritic cells and sensory neurons, opening therapeutic avenues for its control.


Subject(s)
Langerhans Cells/metabolism , Pain, Postoperative/etiology , Pain, Postoperative/metabolism , Receptors, CCR4/metabolism , Sensory Receptor Cells/metabolism , Action Potentials , Animals , Biomarkers , Chemokine CCL17/genetics , Chemokine CCL17/metabolism , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Langerhans Cells/immunology , Mice , Pain, Postoperative/diagnosis , Signal Transduction
7.
PLoS Pathog ; 18(1): e1010255, 2022 01.
Article in English | MEDLINE | ID: mdl-35073387

ABSTRACT

Nucleoside modified mRNA combined with Acuitas Therapeutics' lipid nanoparticles (LNPs) has been shown to support robust humoral immune responses in many preclinical animal vaccine studies and later in humans with the SARS-CoV-2 vaccination. We recently showed that this platform is highly inflammatory due to the LNPs' ionizable lipid component. The inflammatory property is key to support the development of potent humoral immune responses. However, the mechanism by which this platform drives T follicular helper (Tfh) cells and humoral immune responses remains unknown. Here we show that lack of Langerhans cells or cDC1s neither significantly affected the induction of PR8 HA and SARS-CoV-2 RBD-specific Tfh cells and humoral immune responses, nor susceptibility towards the lethal challenge of influenza and SARS-CoV-2. However, the combined deletion of these two DC subsets led to a significant decrease in the induction of PR8 HA and SARS-CoV-2 RBD-specific Tfh cell and humoral immune responses. Despite these observed defects, these mice remained protected from lethal influenza and SARS-CoV-2 challenges. We further found that IL-6, unlike neutrophils, was required to generate normal Tfh cells and antibody responses, but not for protection from influenza challenge. In summary, here we bring evidence that the mRNA-LNP platform can support the induction of protective immune responses in the absence of certain innate immune cells and cytokines.


Subject(s)
COVID-19 Vaccines/immunology , Dendritic Cells/immunology , Influenza Vaccines/immunology , Langerhans Cells/immunology , Liposomes/immunology , Vaccines, Synthetic/immunology , mRNA Vaccines/immunology , Animals , COVID-19/immunology , Mice , Nanoparticles , Orthomyxoviridae Infections/immunology , SARS-CoV-2/immunology
8.
Biochem Biophys Res Commun ; 586: 100-106, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34837833

ABSTRACT

Lipopolysaccharide (LPS) is the principal component of the outer membrane of gram-negative bacteria. The prior oral administration of LPS attenuates inflammatory responses, such as intestinal injury and atopic dermatitis, in mouse models; however, the underlying mechanism remains unclear. Here, we examined the effect of topical LPS application on allergic contact dermatitis and its mechanism of action using a murine contact hypersensitivity (CHS) model. Prolonged LPS application to the skin significantly suppressed 2,4-dinitrofluorobenzene (DNFB)-induced CHS. LPS application to the skin also reduced the phagocytosis of fluorescein isothiocyanate (FITC)-dextran by Langerhans and dendritic cells. Cutaneous cell migration into the skin-draining lymph nodes (LNs) induced by FITC painting was reduced by LPS application. During the CHS response, DNFB application induced T-cell proliferation and inflammatory cytokine production in skin-draining LNs, whereas prolonged LPS application inhibited DNFB-induced T-cell growth and interferon gamma production, indicating suppression of DNFB-induced sensitization. These results suggest that prolonged LPS application suppressed DNFB-induced sensitization and subsequently CHS response. Our findings imply that topical application of LPS may prevent allergic dermatitis such as CHS.


Subject(s)
Dermatitis, Contact/drug therapy , Immunologic Factors/pharmacology , Lipopolysaccharides/pharmacology , Lymphocytes/drug effects , Skin/drug effects , Administration, Cutaneous , Animals , Cell Line , Cell Movement/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dermatitis, Contact/etiology , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Dextrans/metabolism , Dinitrofluorobenzene/administration & dosage , Ear , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/immunology , Langerhans Cells/cytology , Langerhans Cells/drug effects , Langerhans Cells/immunology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Phagocytosis/drug effects , Primary Cell Culture , Skin/immunology , Skin/pathology
9.
Nat Immunol ; 22(12): 1538-1550, 2021 12.
Article in English | MEDLINE | ID: mdl-34795444

ABSTRACT

The signals driving the adaptation of type 2 dendritic cells (DC2s) to diverse peripheral environments remain mostly undefined. We show that differentiation of CD11blo migratory DC2s-a DC2 population unique to the dermis-required IL-13 signaling dependent on the transcription factors STAT6 and KLF4, whereas DC2s in lung and small intestine were STAT6-independent. Similarly, human DC2s in skin expressed an IL-4 and IL-13 gene signature that was not found in blood, spleen and lung DCs. In mice, IL-13 was secreted homeostatically by dermal innate lymphoid cells and was independent of microbiota, TSLP or IL-33. In the absence of IL-13 signaling, dermal DC2s were stable in number but remained CD11bhi and showed defective activation in response to allergens, with diminished ability to support the development of IL-4+GATA3+ helper T cells (TH), whereas antifungal IL-17+RORγt+ TH cells were increased. Therefore, homeostatic IL-13 fosters a noninflammatory skin environment that supports allergic sensitization.


Subject(s)
Cell Communication , Cell Differentiation , Interleukin-13/metabolism , Langerhans Cells/metabolism , Skin/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Allergens/pharmacology , Animals , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cells, Cultured , Databases, Genetic , Humans , Interleukin-13/genetics , Langerhans Cells/drug effects , Langerhans Cells/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction , Skin/cytology , Skin/drug effects , Skin/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Transcriptome
10.
Front Immunol ; 12: 732298, 2021.
Article in English | MEDLINE | ID: mdl-34745102

ABSTRACT

Immune modulating therapies and vaccines are in high demand, not least to the recent global spread of SARS-CoV2. To achieve efficient activation of the immune system, professional antigen presenting cells have proven to be key coordinators of such responses. Especially targeted approaches, actively directing antigens to specialized dendritic cells, promise to be more effective and accompanied by reduced payload due to less off-target effects. Although antibody and glycan-based targeting of receptors on dendritic cells have been employed, these are often expensive and time-consuming to manufacture or lack sufficient specificity. Thus, we applied a small-molecule ligand that specifically binds Langerin, a hallmark receptor on Langerhans cells, conjugated to a model protein antigen. Via microneedle injection, this construct was intradermally administered into intact human skin explants, selectively loading Langerhans cells in the epidermis. The ligand-mediated cellular uptake outpaces protein degradation resulting in intact antigen delivery. Due to the pivotal role of Langerhans cells in induction of immune responses, this approach of antigen-targeting of tissue-resident immune cells offers a novel way to deliver highly effective vaccines with minimally invasive administration.


Subject(s)
Antigens, CD/metabolism , Antigens/administration & dosage , Green Fluorescent Proteins/administration & dosage , Langerhans Cells/metabolism , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Animals , Antigens/immunology , Antigens/metabolism , COS Cells , Chlorocebus aethiops , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Injections, Intradermal , Langerhans Cells/immunology , Ligands , Miniaturization , Nanomedicine , Needles , Protein Binding , Protein Transport , Proteolysis , THP-1 Cells , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/metabolism
11.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504008

ABSTRACT

Chronic ultraviolet (UV) radiation exposure is the greatest risk factor for cutaneous squamous cell carcinoma (cSCC) development, and compromised immunity accelerates this risk. Having previously identified that epidermal Langerhans cells (LC) facilitate the expansion of UV-induced mutant keratinocytes (KC), we sought to more fully elucidate the immune pathways critical to cutaneous carcinogenesis and to identify potential targets of intervention. Herein, we reveal that chronic UV induces and LC enhance a local immune shift toward RORγt+ interleukin (IL)-22/IL-17A-producing cells that occurs in the presence or absence of T cells while identifying a distinct RORγt+ Sca-1+ CD103+ ICOS+ CD2+/- CCR6+ intracellular CD3+ cutaneous innate lymphoid cell type-3 (ILC3) population (uvILC3) that is associated with UV-induced mutant KC growth. We further show that mutant KC clone size is markedly reduced in the absence of RORγt+ lymphocytes or IL-22, both observed in association with expanding KC clones, and find that topical application of a RORγ/γt inhibitor during chronic UV exposure reduces local expression of IL-22 and IL-17A while markedly limiting mutant p53 KC clonal expansion. We implicate upstream Toll-like receptor signaling in driving this immune response to chronic UV exposure, as MyD88/Trif double-deficient mice also show substantially reduced p53 island number and size. These data elucidate key immune components of chronic UV-induced cutaneous carcinogenesis that might represent targets for skin cancer prevention.


Subject(s)
Interleukins/metabolism , Keratinocytes/pathology , Lymphocytes/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Skin Neoplasms/pathology , Skin/pathology , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Cells, Cultured , Immunity, Innate/immunology , Interleukins/genetics , Keratinocytes/metabolism , Keratinocytes/radiation effects , Langerhans Cells/immunology , Langerhans Cells/metabolism , Langerhans Cells/pathology , Langerhans Cells/radiation effects , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/radiation effects , Mice , Mutation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Skin/metabolism , Skin/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Interleukin-22
12.
Front Immunol ; 12: 695373, 2021.
Article in English | MEDLINE | ID: mdl-34512625

ABSTRACT

Langerhans cells (LCs) reside in the epidermis where they are poised to mount an antimicrobial response against microbial pathogens invading from the outside environment. To elucidate potential pathways by which LCs contribute to host defense, we mined published LC transcriptomes deposited in GEO and the scientific literature for genes that participate in antimicrobial responses. Overall, we identified 31 genes in LCs that encode proteins that contribute to antimicrobial activity, ten of which were cross-validated in at least two separate experiments. Seven of these ten antimicrobial genes encode chemokines, CCL1, CCL17, CCL19, CCL2, CCL22, CXCL14 and CXCL2, which mediate both antimicrobial and inflammatory responses. Of these, CCL22 was detected in seven of nine transcriptomes and by PCR in cultured LCs. Overall, the antimicrobial genes identified in LCs encode proteins with broad antibacterial activity, including against Staphylococcus aureus, which is the leading cause of skin infections. Thus, this study illustrates that LCs, consistent with their anatomical location, are programmed to mount an antimicrobial response against invading pathogens in skin.


Subject(s)
Antimicrobial Peptides/genetics , Epidermis/metabolism , Langerhans Cells/metabolism , Staphylococcal Skin Infections/genetics , Staphylococcus aureus/pathogenicity , Transcriptome , Cells, Cultured , Databases, Genetic , Epidermis/immunology , Epidermis/microbiology , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Langerhans Cells/immunology , Langerhans Cells/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/immunology
13.
Immunity ; 54(10): 2305-2320.e11, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34508661

ABSTRACT

Langerhans cells (LCs) play a pivotal role in skin homeostasis, and the heterogeneity of LCs has long been considered. In this study, we have identified two steady-state (LC1 and LC2) and two activated LC subsets in the epidermis of human skin and in LCs derived from CD34+ hemopoietic stem cells (HSC-LCs) by utilizing single-cell RNA sequencing and mass cytometry. Analysis of HSC-LCs at multiple time-points during differentiation revealed that EGR1 and Notch signaling were among the top pathways regulating the bifurcation of LC1 and LC2. LC1 were characterized as classical LCs, mainly related to innate immunity and antigen processing. LC2 were similar to monocytes or myeloid dendritic cells, involving in immune responses and leukocyte activation. LC1 remained stable under inflammatory microenvironment, whereas LC2 were prone to being activated and demonstrated elevated expression of immuno-suppressive molecules. We revealed distinct human LC subsets that require different developmental regulation and orchestrate reciprocal functions.


Subject(s)
Cell Differentiation/immunology , Langerhans Cells/cytology , Langerhans Cells/immunology , Skin/cytology , Skin/immunology , Antigen Presentation/immunology , Hematopoietic Stem Cells/immunology , Humans , Immunity, Innate/immunology
14.
Front Immunol ; 12: 665312, 2021.
Article in English | MEDLINE | ID: mdl-34211464

ABSTRACT

Langerhans cells (LCs) reside in the epidermis as a dense network of immune system sentinels, coordinating both immunogenic and tolerogenic immune responses. To determine molecular switches directing induction of LC immune activation, we performed mathematical modelling of gene regulatory networks identified by single cell RNA sequencing of LCs exposed to TNF-alpha, a key pro-inflammatory signal produced by the skin. Our approach delineated three programmes of LC phenotypic activation (immunogenic, tolerogenic or ambivalent), and confirmed that TNF-alpha enhanced LC immunogenic programming. Through regulon analysis followed by mutual information modelling, we identified IRF1 as the key transcription factor for the regulation of immunogenicity in LCs. Application of a mathematical toggle switch model, coupling IRF1 with tolerance-inducing transcription factors, determined the key set of transcription factors regulating the switch between tolerance and immunogenicity, and correctly predicted LC behaviour in LCs derived from different body sites. Our findings provide a mechanistic explanation of how combinatorial interactions between different transcription factors can coordinate specific transcriptional programmes in human LCs, interpreting the microenvironmental context of the local tissue microenvironments.


Subject(s)
Interferon Regulatory Factors/metabolism , Langerhans Cells/immunology , Langerhans Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Epidermis/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Humans , Interferon Regulatory Factors/genetics , Signal Transduction , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics
15.
Anticancer Res ; 41(7): 3439-3448, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34230139

ABSTRACT

BACKGROUND/AIM: The role of immune cells and PD-L1 in cutaneous squamous carcinogenesis is unclear. This study examines T-cell populations, Langerhans cells (LCs) and PD-L1 in invasive squamous cell carcinoma (inSCC), adjacent precursors and normal skin (NS) to investigate their participation in tumorigenesis. MATERIALS AND METHODS: Cases of cutaneous inSCC with adjacent precursors (n=125) were selected. In situ SCC (isSCC) and actinic keratosis (AK) were observed in 53 and 123 cases, respectively, whereas NS was present in 123 lesions. Immunohistochemistry was performed for CD3, CD8, Foxp3, CD1a and PD-L1. RESULTS: T-cells, LCs and PD-L1 gradually increase during the evolution from AK to isSCC and inSCC, with statistical significance between all lesions, except for CD3+ and CD8+ cells between isSCC and inSCC. Epithelial PD-L1 expression correlates with tumor diameter and thickness. CONCLUSION: The progressive increase of T-cells, LCs and PD-L1 in cutaneous squamous carcinogenesis provides rationale for immunotherapy and identification of predictive biomarkers.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Langerhans Cells/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , T-Lymphocyte Subsets/metabolism , Adult , Aged , Aged, 80 and over , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/pathology , Female , Humans , Immunohistochemistry/methods , Langerhans Cells/immunology , Langerhans Cells/pathology , Male , Middle Aged , Skin Neoplasms/pathology , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/immunology
16.
PLoS Pathog ; 17(7): e1009749, 2021 07.
Article in English | MEDLINE | ID: mdl-34324611

ABSTRACT

The main avenue for the development of an HIV-1 vaccine remains the induction of protective antibodies. A rationale approach is to target antigen to specific receptors on dendritic cells (DC) via fused monoclonal antibodies (mAb). In mouse and non-human primate models, targeting of skin Langerhans cells (LC) with anti-Langerin mAbs fused with HIV-1 Gag antigen drives antigen-specific humoral responses. The development of these immunization strategies in humans requires a better understanding of early immune events driven by human LC. We therefore produced anti-Langerin mAbs fused with the HIV-1 gp140z Envelope (αLC.Env). First, we show that primary skin human LC and in vitro differentiated LC induce differentiation and expansion of naïve CD4+ T cells into T follicular helper (Tfh) cells. Second, when human LC are pre-treated with αLC.Env, differentiated Tfh cells significantly promote the production of specific IgG by B cells. Strikingly, HIV-Env-specific Ig are secreted by HIV-specific memory B cells. Consistently, we found that receptors and cytokines involved in Tfh differentiation and B cell functions are upregulated by LC during their maturation and after targeting Langerin. Finally, we show that subcutaneous immunization of mice by αLC.Env induces germinal center (GC) reaction in draining lymph nodes with higher numbers of Tfh cells, Env-specific B cells, as well as specific IgG serum levels compared to mice immunized with the non-targeting Env antigen. Altogether, we provide evidence that human LC properly targeted may be licensed to efficiently induce Tfh cell and B cell responses in GC.


Subject(s)
AIDS Vaccines/immunology , Antigens, CD/immunology , HIV-1/immunology , Immunity, Humoral/immunology , Langerhans Cells/immunology , Lectins, C-Type/immunology , Mannose-Binding Lectins/immunology , Animals , Humans , Lymphocyte Activation/immunology , Mice , env Gene Products, Human Immunodeficiency Virus/immunology
17.
EMBO J ; 40(16): e106540, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34121210

ABSTRACT

Dendritic cells (DC) subsets, like Langerhans cells (LC), are immune cells involved in pathogen sensing. They express specific antimicrobial cellular factors that are able to restrict infection and limit further pathogen transmission. Here, we identify the alarmin S100A9 as a novel intracellular antiretroviral factor expressed in human monocyte-derived and skin-derived LC. The intracellular expression of S100A9 is decreased upon LC maturation and inversely correlates with enhanced susceptibility to HIV-1 infection of LC. Furthermore, silencing of S100A9 in primary human LC relieves HIV-1 restriction while ectopic expression of S100A9 in various cell lines promotes intrinsic resistance to both HIV-1 and MLV infection by acting on reverse transcription. Mechanistically, the intracellular expression of S100A9 alters viral capsid uncoating and reverse transcription. S100A9 also shows potent inhibitory effect against HIV-1 and MMLV reverse transcriptase (RTase) activity in vitro in a divalent cation-dependent manner. Our findings uncover an unexpected intracellular function of the human alarmin S100A9 in regulating antiretroviral immunity in Langerhans cells.


Subject(s)
Alarmins/genetics , Calgranulin B/genetics , HIV-1/physiology , Langerhans Cells/virology , Moloney murine leukemia virus/physiology , Retroviridae Infections/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cricetulus , HIV-1/genetics , Host-Pathogen Interactions , Humans , Langerhans Cells/immunology , Leukemia, Experimental/prevention & control , Mice , Moloney murine leukemia virus/genetics , Reverse Transcription , Transforming Growth Factor beta/immunology , Tumor Virus Infections/prevention & control , Virus Replication
19.
Appl Immunohistochem Mol Morphol ; 29(8): 606-612, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33958525

ABSTRACT

Langerhans cells (LCs) play important roles in cell-mediated immune reactions, as well as in the pathogenesis of periapical lesions. The aim of this study is to evaluate the role of LCs in the proliferative epithelium of radicular cysts (RCs) and the release of the proinflammatory cytokine tumor necrosis factor α (TNF-α) associated with epithelial thickness. Thirty cases of RCs and 30 cases of residual RCs were randomly selected. Morphologic analysis was performed to evaluate the association between the inflammatory infiltrate, cystic epithelial thickness and lesion size, in addition to immunohistochemical assessment of CD1a, CD68, and TNF-α. The highest macrophage percentages and TNF-α scores were found in RCs (P=0.038 and 0.017, respectively). The largest number of LCs was observed in RCs (P=0.021), especially those exhibiting atrophic epithelium (P=0.05). In addition, LCs were positively correlated with the number of macrophages in both RCs and residual RCs (P=0.033 and 0.002, respectively). In contrast to LCs, the largest number of macrophages was detected in cases with an intense inflammatory infiltrate (P=0.022). In addition, the highest TNF-α scores were associated with an intense inflammatory infiltrate (P=0.024) when analyzed in the capsule of RCs (P=0.017). In conclusion, LCs participate in defense mechanisms and were present in all cases evaluated. Along with macrophages, these cells release proinflammatory cytokines such as TNF-α, which is responsible for inducing the continued proliferation of cystic epithelium.


Subject(s)
Langerhans Cells , Macrophages , Radicular Cyst , Adult , Aged , Antigens, CD/immunology , Antigens, CD1/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Epithelium/immunology , Epithelium/pathology , Female , Humans , Langerhans Cells/immunology , Langerhans Cells/pathology , Macrophages/immunology , Macrophages/pathology , Male , Middle Aged , Radicular Cyst/immunology , Radicular Cyst/pathology , Tumor Necrosis Factor-alpha/immunology
20.
J Dermatol Sci ; 102(3): 177-183, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33994063

ABSTRACT

BACKGROUND: Alopecia areata (AA) is an autoimmune disease resulting in non-scarring hair loss. Animal models are useful means to identify candidates for therapeutic agents. The C3H/HeJ mouse AA model induced via transferring cultured lymphoid cells isolated from AA-affected mice is widely used for AA research. However, this conventional method requires the continuous breeding of AA mice. OBJECTIVE: We aimed to establish a new method to generate AA model using the transfer of cryopreserved cells, which allows the rapid induction of a large number of AA mice when needed. METHODS: We cryopreserved lymph node cells soon after isolation from AA-affected mice and injected thawed-cultured cells into recipient mice. H&E staining, immunohistochemical staining, quantitative real-time PCR and ELISA were conducted to identify pathological characteristics. Flow cytometry was performed to reveal the profile of transferred cells. RESULTS: More than 90 % of recipient mice developed AA-like hair loss and showed inflammatory cell infiltration around anagen hair follicles, markedly increased mRNA expressions of interferon-γ, CXCL11, and granzyme B, and elevated interferon-α protein levels in the skin compared with naïve mice. Higher percentages of effector memory T cells and dendritic cells in transferred cells resulted in a higher incidence of AA. CONCLUSION: This is the first report to establish a method for generating AA mice using cryopreserved lymphocytes. These AA mice have similar pathological characteristics to AA mice generated with the conventional method and AA patients. This convenient and reproducible method is expected to be valuable for AA study.


Subject(s)
Alopecia Areata/immunology , Hair Follicle/pathology , Lymph Nodes/cytology , Lymphocyte Transfusion/methods , Alopecia Areata/diagnosis , Alopecia Areata/pathology , Animals , Cells, Cultured , Cryopreservation , Disease Models, Animal , Female , Hair Follicle/immunology , Humans , Injections, Intradermal , Langerhans Cells/immunology , Lymphocytes/immunology , Mice , Mice, Inbred C3H , Primary Cell Culture , Reproducibility of Results , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...