53 results on '"Daniel H. Kaplan"'
Search Results
2. Skin codelivery of contact sensitizers and neurokinin-1 receptor antagonists integrated in microneedle arrays suppresses allergic contact dermatitis
- Author
-
Mohna Bandyopadhyay, Adrian E. Morelli, Stephen C. Balmert, Nicole L. Ward, Geza Erdos, Tina L. Sumpter, Emrullah Korkmaz, Daniel H. Kaplan, Martin H. Oberbarnscheidt, Olga Tkacheva, William J. Shufesky, Louis D. Falo, and Adriana T. Larregina
- Subjects
Mice ,Neurokinin-1 Receptor Antagonists ,Immunology ,Dermatitis, Allergic Contact ,Immunology and Allergy ,Animals ,Receptors, Neurokinin-1 ,Substance P ,Haptens ,Article - Abstract
BACKGROUND: Allergic contact dermatitis (CD) is a chronic inflammatory skin disease caused by type-1 biased adaptive immunity for which there is an unmet need for antigen (Ag)-specific immunotherapies. Exposure to skin sensitizers stimulates secretion of the proinflammatory neuropeptides substance P (SP) and hemokinin 1 (HK1), which signal via the neurokinin-1 receptor (NK1R) to promote the innate and adaptive immune responses of CD. Accordingly, mice lacking the NK1R develop impaired CD. Nonetheless, the role and therapeutic opportunities of targeting the NK1R in CD remain to be elucidated. OBJECTIVE: To develop an Ag-specific immunosuppressive approach to treat CD by skin co-delivery of hapten and NK1R antagonists integrated in dissolvable microneedle arrays (MNA). METHODS: In vivo mouse models of contact hypersensitivity and ex-vivo models of human skin were used to delineate the effects and mechanisms of NK1R-signaling and the immunosuppressive effects of the contact sensitizer-NK1R-antagonists-MNA in CD. RESULTS: We demonstrate in mice that CD requires NK1R signaling by SP and HK1. Specific deletion of the NK1R in keratinocytes and dendritic cells, but not in mast cells prevented CD. Skin co-delivery of hapten- or Ag-NK1R-antagonist-MNA inhibited neuropeptide-mediated skin inflammation in mouse and human skin, promoted deletion of Ag-specific effector T cells and increased regulatory T cells, which prevented CD onset and relapses locally and systemically in an Ag-specific manner. CONCLUSIONS: Our findings demonstrate that immune-regulation by engineering localized skin neuroimmune networks can be used to treat cutaneous diseases that like CD, are caused by type-1 immunity.
- Published
- 2021
3. Adenosine Triphosphate Released by Candida albicans Is Associated with Reduced Skin Infectivity
- Author
-
Selene Mogavero, Christophe d'Enfert, Tara N. Edwards, Marie-Elisabeth Bougnoux, Alicia R. Mathers, Shiqun Zhang, Daniel H. Kaplan, Bernhard Hube, Judith Berman, University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE), Leibniz Institute for Natural Product Research and Infection Biology (Hans Knoell Institute), Tel Aviv University (TAU), Biologie et Pathogénicité fongiques (BPF), Institut National de la Recherche Agronomique (INRA)-Institut Pasteur [Paris] (IP), Unité de Parasitologie-Mycologie, Service de Microbiologie [Hôpital Necker-Enfants-Malades, Paris], Assistance Publique - Hôpitaux de Paris, and National Institute for Health grant R01AR067187 (DHK)European Research Council Advanced Award (340087 RAPLODAPT to JB).National Institutes for Health 1S10OD011925-01
- Subjects
Dermatology ,Candidiasis, Cutaneous ,Biochemistry ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Adenosine Triphosphate ,Candida albicans ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Skin ,Colony-forming unit ,Infectivity ,0303 health sciences ,[QFIN]Quantitative Finance [q-fin] ,biology ,Host Microbial Interactions ,Virulence ,[SDE.IE]Environmental Sciences/Environmental Engineering ,030306 microbiology ,Cell Biology ,biology.organism_classification ,Immunity, Innate ,chemistry ,Adenosine triphosphate - Abstract
International audience
- Published
- 2021
- Full Text
- View/download PDF
4. Gut Helicobacter presentation by multiple dendritic cell subsets enables context-specific regulatory T cell generation
- Author
-
Shannon Young, Vivek Durai, Jiani N. Chai, Ronald N. Germain, Harikesh S. Wong, Jaeu Yi, Chyi-Song Hsieh, Daniel H. Kaplan, Emilie V. Russler-Germain, Kenneth M. Murphy, Teresa L. Ai, and Katherine Nutsch
- Subjects
0301 basic medicine ,Mouse ,QH301-705.5 ,Regulatory T cell ,dendritic cell ,Science ,Cell ,chemical and pharmacologic phenomena ,Mice, Transgenic ,regulatory t cell ,Biology ,T-Lymphocytes, Regulatory ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immunology and Inflammation ,Antigen ,Cell Movement ,Helicobacter ,Genetic model ,medicine ,microbiota ,Animals ,Biology (General) ,Mice, Knockout ,tolerance ,General Immunology and Microbiology ,colon ,General Neuroscience ,T-cell receptor ,hemic and immune systems ,Cell Differentiation ,General Medicine ,Dendritic cell ,Dendritic Cells ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,T cell differentiation ,Medicine ,Lymph Nodes ,Ex vivo ,030215 immunology ,Research Article - Abstract
Generation of tolerogenic peripheral regulatory T (pTreg) cells is commonly thought to involve CD103+ gut dendritic cells (DCs), yet their role in commensal-reactive pTreg development is unclear. Using two Helicobacter-specific T cell receptor (TCR) transgenic mouse lines, we found that both CD103+ and CD103– migratory, but not resident, DCs from the colon-draining mesenteric lymph node presented Helicobacter antigens to T cells ex vivo. Loss of most CD103+ migratory DCs in vivo using murine genetic models did not affect the frequency of Helicobacter-specific pTreg cell generation or induce compensatory tolerogenic changes in the remaining CD103– DCs. By contrast, activation in a Th1-promoting niche in vivo blocked Helicobacter-specific pTreg generation. Thus, these data suggest a model where DC-mediated effector T cell differentiation is ‘dominant’, necessitating that all DC subsets presenting antigen are permissive for pTreg cell induction to maintain gut tolerance.
- Published
- 2021
5. An IL-17F.S65L knockin mouse reveals similarities and differences in IL-17F function in oral candidiasis: A new tool to understand IL-17F
- Author
-
Chunsheng Zhou, Sarah L. Gaffen, Leticia Monin, Rami Bechara, Rachael A. Gordon, Tara N. Edwards, Felix E. Y. Aggor, Sebastien Gingras, and Daniel H. Kaplan
- Subjects
medicine.medical_treatment ,Immunology ,Mutant ,Mutation, Missense ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Oropharyngeal Candidiasis ,Article ,Mice ,Candida albicans ,medicine ,Immunology and Allergy ,Animals ,Gene Knock-In Techniques ,Chronic mucocutaneous candidiasis ,Phenocopy ,Mutation ,Interleukin-17 ,Candidiasis ,biology.organism_classification ,medicine.disease ,Cytokine ,IL17A ,Mouth Diseases - Abstract
Oropharyngeal candidiasis (OPC) is an opportunistic infection of the oral mucosa caused by the commensal fungus Candida albicans. IL-17R signaling is essential to prevent OPC in mice and humans, but the individual roles of its ligands, IL-17A, IL-17F, and IL-17AF, are less clear. A homozygous IL-17F deficiency in mice does not cause OPC susceptibility, whereas mice lacking IL-17A are moderately susceptible. In humans, a rare heterozygous mutation in IL-17F (IL-17F.S65L) was identified that causes chronic mucocutaneous candidiasis, suggesting the existence of essential antifungal pathways mediated by IL-17F and/or IL-17AF. To investigate the role of IL-17F and IL-17AF in more detail, we exploited this “experiment of nature” by creating a mouse line bearing the homologous mutation in IL-17F (Ser65Leu) by CRISPR/Cas9. Unlike Il17f−/− mice that are resistant to OPC, Il17fS65L/S65L mice showed increased oral fungal burdens similar to Il17a−/− mice. In contrast to humans, however, disease was only evident in homozygous, not heterozygous, mutant mice. The mutation was linked to modestly impaired CXC chemokine expression and neutrophil recruitment to the infected tongue but not to alterations in oral antimicrobial peptide expression. These findings suggest mechanisms by which the enigmatic cytokine IL-17F contributes to host defense against fungi. Moreover, because these mice do not phenocopy Il17f−/− mice, they may provide a valuable tool to interrogate IL-17F and IL-17AF function in vivo in other settings.
- Published
- 2020
6. Suppression of Th1 Priming by TLR2 Agonists during Cutaneous Immunization Is Mediated by Recruited CCR2+ Monocytes
- Author
-
Ulrich D. Kadolsky, Daniel H. Kaplan, Alison J. Johnson, Tony W. Ng, Andrew J. Yates, Steven A. Porcelli, Grégoire Lauvau, Shajo Kunnath-Velayudhan, G. H. Gossel, William R. Jacobs, John Chan, Christopher T. Johndrow, and Michael F. Goldberg
- Subjects
0301 basic medicine ,Agonist ,Receptors, CCR2 ,medicine.drug_class ,Immunology ,Priming (immunology) ,Mice, Transgenic ,Biology ,Lymphocyte Activation ,Article ,Monocytes ,Immune tolerance ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cell Movement ,Immune Tolerance ,medicine ,Animals ,Immunology and Allergy ,Receptor ,Cells, Cultured ,Skin ,Mice, Inbred BALB C ,Innate immune system ,Drug Administration Routes ,Vaccination ,Pattern recognition receptor ,Th1 Cells ,Mycobacterium bovis ,Toll-Like Receptor 2 ,Mice, Inbred C57BL ,Repressor Proteins ,TLR2 ,Basic-Leucine Zipper Transcription Factors ,030104 developmental biology ,Female ,Immunization ,030215 immunology - Abstract
Effective subunit vaccines require the incorporation of adjuvants that stimulate cells of the innate immune system to generate protective adaptive immune responses. Pattern recognition receptor agonists are a growing class of potential adjuvants that can shape the character of the immune response to subunit vaccines by directing the polarization of CD4 T cell differentiation to various functional subsets. In the current study, we applied a high-throughput in vitro screen to assess murine CD4 T cell polarization by a panel of pattern recognition receptor agonists. This identified lipopeptides with TLR2 agonist activity as exceptional Th1-polarizing adjuvants. In vivo, we demonstrated that i.v. administration of TLR2 agonists with Ag in mice replicated the findings from in vitro screening by promoting strong Th1 polarization. In contrast, TLR2 agonists inhibited priming of Th1 responses when administered cutaneously in mice. This route-specific suppression was associated with infiltrating CCR2+ cells in the skin-draining lymph nodes and was not uniquely dependent on any of the well characterized subsets of dendritic cells known to reside in the skin. We further demonstrated that priming of CD4 T cells to generate Th1 effectors following immunization with the Mycobacterium bovis bacillus Calmette–Guérin (BCG) strain, a lipoprotein-rich bacterium recognized by TLR2, was dependent on the immunization route, with significantly greater Th1 responses with i.v. compared with intradermal administration of BCG. A more complete understanding of route-dependent TLR2 responses may be critical for informed design of novel subunit vaccines and for improvement of BCG and other vaccines based on live-attenuated organisms.
- Published
- 2018
- Full Text
- View/download PDF
7. Antigen-Presenting Cells in the Skin
- Author
-
Sakeen W. Kashem, Daniel H. Kaplan, and Muzlifah Haniffa
- Subjects
0301 basic medicine ,Cell type ,T-Lymphocytes ,T cell ,Immunology ,Antigen presentation ,Antigen-Presenting Cells ,Inflammation ,Human skin ,Biology ,Lymphocyte Activation ,Monocytes ,Mice ,03 medical and health sciences ,Immune system ,Cell Movement ,medicine ,Animals ,Homeostasis ,Humans ,Immunology and Allergy ,Antigen-presenting cell ,Skin ,Antigen Presentation ,Macrophages ,Dendritic Cells ,030104 developmental biology ,Lymphatic system ,medicine.anatomical_structure ,Langerhans Cells ,medicine.symptom - Abstract
Professional antigen-presenting cells (APCs) in the skin include dendritic cells, monocytes, and macrophages. They are highly dynamic, with the capacity to enter skin from the peripheral circulation, patrol within tissue, and migrate through lymphatics to draining lymph nodes. Skin APCs are endowed with antigen-sensing, -processing, and -presenting machinery and play key roles in initiating, modulating, and resolving cutaneous inflammation. Skin APCs are a highly heterogeneous population with functionally specialized subsets that are developmentally imprinted and modulated by local tissue microenvironmental and inflammatory cues. This review explores recent advances that have allowed for a more accurate taxonomy of APC subsets found in both mouse and human skin. It also examines the functional specificity of individual APC subsets and their collaboration with other immune cell types that together promote adaptive T cell and regional cutaneous immune responses during homeostasis, inflammation, and disease.
- Published
- 2017
- Full Text
- View/download PDF
8. Nonpeptidergic neurons suppress mast cells via glutamate to maintain skin homeostasis
- Author
-
Amanda C. Poholek, Natalie Rittenhouse, Tara N. Edwards, Tina L. Sumpter, Jianing Wu, Brian M. Davis, Shiqun Zhang, Virendra K. Chaudhri, Kathryn M. Albers, Paul Yifan Zhou, Harinder Singh, Yi Yang, Daniel H. Kaplan, Elizabeth G. Schmitz, Jonathan A. Cohen, Toshiro Hirai, Benjamin D. McNeil, and H. Richard Koerber
- Subjects
Langerhans cell ,Integrin beta Chains ,Glutamic Acid ,Kainate receptor ,Inflammation ,Dermatitis ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Receptors, G-Protein-Coupled ,Glutamate receptor binding ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Diphtheria Toxin ,Mast Cells ,Cells, Cultured ,030304 developmental biology ,Skin ,Mice, Knockout ,Neurons ,0303 health sciences ,Degranulation ,Glutamate receptor ,Mast cell ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Langerhans Cells ,beta-Alanine ,Female ,medicine.symptom ,Free nerve ending ,030217 neurology & neurosurgery - Abstract
Cutaneous mast cells mediate numerous skin inflammatory processes and have anatomical and functional associations with sensory afferent neurons. We reveal that epidermal nerve endings from a subset of sensory nonpeptidergic neurons expressing MrgprD are reduced by the absence of Langerhans cells. Loss of epidermal innervation or ablation of MrgprD-expressing neurons increased expression of a mast cell gene module, including the activating receptor, Mrgprb2, resulting in increased mast cell degranulation and cutaneous inflammation in multiple disease models. Agonism of MrgprD-expressing neurons reduced expression of module genes and suppressed mast cell responses. MrgprD-expressing neurons released glutamate which was increased by MrgprD agonism. Inhibiting glutamate release or glutamate receptor binding yielded hyperresponsive mast cells with a genomic state similar to that in mice lacking MrgprD-expressing neurons. These data demonstrate that MrgprD-expressing neurons suppress mast cell hyperresponsiveness and skin inflammation via glutamate release, thereby revealing an unexpected neuroimmune mechanism maintaining cutaneous immune homeostasis.
- Published
- 2019
9. Corrigendum to 'Keratinocyte-derived TGFβ is not required to maintain skin immune homeostasis' [J. Dermatol. Sci. 94 (2) (2019) 290–297]
- Author
-
J.A. Shaik, Toshiro Hirai, Daniel H. Kaplan, Yukari Zenke, and Yi Yang
- Subjects
Keratinocytes ,Mice, Knockout ,Autoimmunity ,Dermatology ,Biology ,Biochemistry ,Article ,Cell biology ,Transforming Growth Factor beta1 ,Gene Knockout Techniques ,Mice ,Tamoxifen ,medicine.anatomical_structure ,Models, Animal ,medicine ,Animals ,Immune homeostasis ,Keratinocyte ,Molecular Biology ,Skin - Abstract
Transforming growth factor beta 1 (TGFβ) is known to be a regulator of autoimmunity. Loss of TGFβ leads to severe multi-organ autoimmunity in mice. In skin, role of TGFβ in suppressing autoimmunity is unclear.Determine whether Keratinocyte (KC)-derived TGFβ is required for skin immune homeostasis.We generated K14-CreERKC was the major source of TGFβ in epidermis. Topical tamoxifen application led to efficient TGFβ1 deletion. The expected acanthosis was observed but no inflammatory infiltrate or altered numbers of resident immune cells were evident. Similarly, Itgb6KC-derived TGFβ and epidermal TGFβ activation are not required to suppress skin autoimmunity in steady state.
- Published
- 2019
10. Langerhans Cells Spy on Keratinocytes
- Author
-
Daniel H. Kaplan and Jacinto S. De La Cruz Diaz
- Subjects
0301 basic medicine ,Keratinocytes ,Cell signaling ,Mice, Transgenic ,Dermatology ,Cell Communication ,Biochemistry ,Article ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Cell Movement ,In vivo ,Animals ,RNA, Messenger ,Molecular Biology ,Messenger RNA ,Nanotubes ,integumentary system ,Chemistry ,Mechanism (biology) ,RNA ,Biological Transport ,Dendrites ,Cell Biology ,Cell biology ,030104 developmental biology ,Cellular Microenvironment ,030220 oncology & carcinogenesis ,Langerhans Cells ,Keratins ,Epidermis ,Intracellular - Abstract
The immune functions of epithelia-resident dendritic cells are influenced by epithelial-derived cytokines. Here we identified a communication form between tissue-resident dendritic cells and niche cells that allows direct intracellular material exchange between the parties. We show that many keratinocyte (KC)-specific molecules such as keratins and adhesion molecules could be detected in the epidermal-resident Langerhans cells (LCs) as mRNA and protein. Furthermore, KC-derived Cre led to genetic recombination in the LCs. We also found that LCs containing KC-derived material were more prone to migration. The KC-specific signatures were transferred from KCs to LCs through an exosome-independent mechanism that likely involved nanotubes/dendrites. The transfer of material between epithelial cells and epithelia-associated dendritic cells was not limited to mice or to KC-to-LC transfer. Taken together, these data suggest that the epithelial environment might have a long-term effect on dendritic cell biology and that genetic tools that specifically target epithelial cells also affect tissue-resident immune cells.
- Published
- 2019
11. Langerhans Cells Sense Staphylococcus aureus Wall Teichoic Acid through Langerin To Induce Inflammatory Responses
- Author
-
Christoph Rademacher, Daniel H. Kaplan, Felix F. Fuchsberger, Nina M. van Sorge, Teunis B. H. Geijtenbeek, Andreas Peschel, Nienke H. van Teijlingen, Christopher Weidenmaier, Jacinto S. De La Cruz Diaz, Rob van Dalen, Jos A. G. van Strijp, Matevž Rumpret, Jonas Hanske, Graduate School, Experimental Immunology, Infectious diseases, and AII - Infectious diseases
- Subjects
Staphylococcus ,medicine.disease_cause ,Langerhans cell ,chemistry.chemical_compound ,Mice ,Cells, Cultured ,Skin ,0303 health sciences ,Teichoic acid ,biology ,integumentary system ,atopic dermatitis ,Interleukin-17 ,Pattern recognition receptor ,hemic and immune systems ,Atopic dermatitis ,Staphylococcal Infections ,QR1-502 ,medicine.anatomical_structure ,Staphylococcus aureus ,Antigens, Surface ,Cytokines ,Research Article ,Langerin ,glycosylation ,wall teichoic acid ,chemical and pharmacologic phenomena ,Microbiology ,Proinflammatory cytokine ,Host-Microbe Biology ,Acetylglucosamine ,03 medical and health sciences ,Immune system ,Antigens, CD ,Virology ,medicine ,Journal Article ,Animals ,Humans ,Lectins, C-Type ,030304 developmental biology ,Inflammation ,030306 microbiology ,medicine.disease ,Mice, Inbred C57BL ,Teichoic Acids ,Mannose-Binding Lectins ,chemistry ,langerin ,Langerhans Cells ,biology.protein - Abstract
The bacterium Staphylococcus aureus is an important cause of skin infections and is also associated with the occurrence and severity of eczema. Langerhans cells (LCs), a specific subset of skin immune cells, participate in the immune response to S. aureus, but it is yet unclear how LCs recognize S. aureus. Therefore, we investigated the molecular mechanism underlying the interaction between LCs and S. aureus. We identified that wall teichoic acid, an abundant polymer on the S. aureus surface, is recognized by langerin, a receptor unique to LCs. This interaction allows LCs to discriminate S. aureus from other related staphylococcal species and initiates a proinflammatory response similar to that observed in patients with eczema. Our data therefore provide important new insights into the relationship between S. aureus, LCs, and eczema., Staphylococcus aureus is a major cause of skin and soft tissue infections and aggravator of the inflammatory skin disease atopic dermatitis (AD [eczema]). Epicutaneous exposure to S. aureus induces Th17 responses through skin Langerhans cells (LCs), which paradoxically contribute to host defense but also to AD pathogenesis. The molecular mechanisms underlying the interaction between S. aureus and LCs are poorly understood. Here we demonstrate that human LCs directly interact with S. aureus through the pattern recognition receptor langerin (CD207). Human, but not mouse, langerin interacts with S. aureus through the conserved β-N-acetylglucosamine (GlcNAc) modifications on wall teichoic acid (WTA), thereby discriminating S. aureus from other staphylococcal species. Importantly, the specific S. aureus WTA glycoprofile strongly influences the level of proinflammatory cytokines that are produced by in vitro-generated LCs. Finally, in a murine epicutaneous infection model, S. aureus strongly upregulated transcripts of Cxcl1, Il6, and Il17, which required the presence of both human langerin and WTA β-GlcNAc. Our findings provide molecular insight into the unique proinflammatory capacities of S. aureus in relation to skin inflammation.
- Published
- 2019
12. Keratinocyte-mediated activation of the cytokine TGFβ maintains skin-recirculating memory CD8(+) T cells
- Author
-
Lalit K. Beura, Daniel H. Kaplan, Yukari Zenke, Laurent Bartholin, David Masopust, Yi Yang, and Toshiro Hirai
- Subjects
0301 basic medicine ,Male ,Keratinocytes ,Integrins ,medicine.medical_treatment ,Immunology ,Integrin ,Vaccinia virus ,Skin infection ,Biology ,CD8-Positive T-Lymphocytes ,Virus ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigens, Neoplasm ,Transforming Growth Factor beta ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Skin ,integumentary system ,Cell Differentiation ,medicine.disease ,Cell biology ,Enzyme Activation ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Cytokine ,030220 oncology & carcinogenesis ,biology.protein ,Cytokines ,Female ,Keratinocyte ,Immunologic Memory ,CD8 ,Transforming growth factor - Abstract
Regulated activation of the cytokine TGFβ by integrins α(v)β(6) and α(v)β(8) expressed on keratinocytes is required for residence of epidermal-resident memory T cells, but whether skin-derived signals also affect recirculating memory cells in the skin remains unclear. Here, we show that after resolution of skin vaccinia virus (VV) infection, antigen-specific circulating memory CD8(+) T cells migrated into skin. In mice lacking α(v)β(6) and α(v)β(8) integrins (Itgb6(−/−)Itgb8(fl/fl)-K14-cre), the absence of epidermal activated TGFβ resulted in a gradual loss of E- or P-selectin-binding central and peripheral memory populations, that was rescued when skin entry was inhibited. Skin recirculating memory cells were required for optimal host defense against skin VV infection. These data demonstrate that skin migration can persist after resolution of local skin infection and that the cytokine environment within this nonlymphoid tissue shapes the differentiation state and persistence of the central and peripheral memory T cell pool.
- Published
- 2019
13. Competition for Active TGFβ Cytokine Allows for Selective Retention of Antigen-Specific Tissue- Resident Memory T Cells in the Epidermal Niche
- Author
-
Dario A. A. Vignali, David W. Griggs, Daniel H. Kaplan, Haiyue Li, Creg J. Workman, Yi Yang, Toshiro Hirai, Yukari Zenke, David Masopust, Breanna Anh Thu Nguyen, Harinder Singh, Laurent Bartholin, Jacinto S. De La Cruz Diaz, Virendra K. Chaudhri, and Paul Yifan Zhou
- Subjects
Keratinocytes ,0301 basic medicine ,Receptors, Antigen, T-Cell, alpha-beta ,medicine.medical_treatment ,Immunology ,T-Cell Antigen Receptor Specificity ,CD8-Positive T-Lymphocytes ,Biology ,Binding, Competitive ,T-Lymphocytes, Regulatory ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Transforming Growth Factor beta ,medicine ,Bystander effect ,Animals ,Immunology and Allergy ,Autocrine signalling ,Lymph node ,integumentary system ,Epidermis (botany) ,Bystander Effect ,Clone Cells ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,Cytokine ,medicine.anatomical_structure ,Cellular Microenvironment ,Organ Specificity ,030220 oncology & carcinogenesis ,Epidermis ,Immunologic Memory ,CD8 ,Signal Transduction ,Transforming growth factor - Abstract
Summary Following antigen-driven expansion in lymph node, transforming growth factor-β (TGFβ) is required for differentiation of skin-recruited CD8+ T cell effectors into epidermal resident memory T (Trm) cells and their epidermal persistence. We found that the source of TGFβ -supporting Trm cells was autocrine. In addition, antigen-specific Trm cells that encountered cognate antigen in the skin, and bystander Trm cells that did not, both displayed long-term persistence in the epidermis under steady-state conditions. However, when the active-TGFβ was limited or when new T cell clones were recruited into the epidermis, antigen-specific Trm cells were more efficiently retained than bystander Trm cells. Genetically enforced TGFβR signaling allowed bystander Trm cells to persist in the epidermis as efficiently as antigen-specific Trm cells in both contexts. Thus, competition between T cells for active TGFβ represents an unappreciated selective pressure that promotes the accumulation and persistence of antigen-specific Trm cells in the epidermal niche.
- Published
- 2021
- Full Text
- View/download PDF
14. Stromal cells control the epithelial residence of DCs and memory T cells by regulated activation of TGF-β
- Author
-
Brian Chicoine, David Masopust, Sakeen W. Kashem, Laurent Bartholin, Sathi Wijeyesinghe, Alesia Kaplan, Nathan E. Welty, Alina G. Bridges, Emily A. Thompson, Botond Z. Igyártó, Warren D. Shlomchik, Brian Astry, Daniel H. Kaplan, Aleh Bobr, Lalit K. Beura, Catherine C. Matte, Dean Sheppard, Javed Mohammed, Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de Recherche en Cancérologie de Lyon ( CRCL ), Université Claude Bernard Lyon 1 ( UCBL ), and Université de Lyon-Université de Lyon-Centre Léon Bérard [Lyon]-Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS )
- Subjects
Keratinocytes ,0301 basic medicine ,Integrins ,T-Lymphocytes ,Fluorescent Antibody Technique ,Growth ,CD8-Positive T-Lymphocytes ,Polymerase Chain Reaction ,Epithelium ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,Mice ,0302 clinical medicine ,Cell Movement ,T-Lymphocyte Subsets ,Transforming Growth Factor beta ,Intestine, Small ,Immunology and Allergy ,Intestinal Mucosa ,Skin ,Mice, Knockout ,integumentary system ,medicine.diagnostic_test ,Flow Cytometry ,Cell biology ,Medicine ,France ,Stromal cell ,Cells ,Immunology ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Dermatology ,Biology ,Article ,Flow cytometry ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Immune system ,Antigen ,Antigens, Neoplasm ,medicine ,Animals ,Humans ,Immunity, Mucosal ,Epidermis (botany) ,microbiology ,Dendritic Cells ,Transforming growth factor beta ,030104 developmental biology ,Epidermal Cells ,Mink ,Langerhans Cells ,Immune System ,biology.protein ,pathology ,Epidermis ,Stromal Cells ,Laboratories ,CD8 ,030215 immunology ,Transforming growth factor - Abstract
International audience; Cells of the immune system that reside in barrier epithelia provide a first line of defense against pathogens. Langerhans cells (LCs) and CD8+ tissue-resident memory T cells (TRM cells) require active transforming growth factor-beta1 (TGF-beta) for epidermal residence. Here we found that integrins alphavbeta6 and alphavbeta8 were expressed in non-overlapping patterns by keratinocytes (KCs) and maintained the epidermal residence of LCs and TRM cells by activating latent TGF-beta. Similarly, the residence of dendritic cells and TRM cells in the small intestine epithelium also required alphavbeta6. Treatment of the skin with ultraviolet irradiation decreased integrin expression on KCs and reduced the availability of active TGF-beta, which resulted in LC migration. Our data demonstrated that regulated activation of TGF-beta by stromal cells was able to directly control epithelial residence of cells of the immune system through a novel mechanism of intercellular communication
- Published
- 2016
- Full Text
- View/download PDF
15. Oral epithelial IL-22/STAT3 signaling licenses IL-17-mediated immunity to oral mucosal candidiasis
- Author
-
Daniel H. Kaplan, Partha S. Biswas, Felix E. Y. Aggor, Michail S. Lionakis, Natasha Whibley, Wei Shan, Jay K. Kolls, Vincent M. Bruno, Rachel D. Bailey, Bianca M. Coleman, Amol C. Shetty, Scott K. Durum, Carrie McCracken, Sarah L. Gaffen, Julian R. Naglik, Giraldina Trevejo-Nunez, and Timothy J. Break
- Subjects
0301 basic medicine ,Male ,STAT3 Transcription Factor ,Immunology ,Biology ,Oropharyngeal Candidiasis ,Article ,Interleukin 22 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immunity ,Candidiasis, Oral ,Candida albicans ,medicine ,Animals ,Immunology and Allergy ,Oral mucosa ,STAT3 ,Mice, Knockout ,Interleukins ,Interleukin-17 ,Mouth Mucosa ,Epithelial Cells ,General Medicine ,biology.organism_classification ,Epithelium ,Mice, Inbred C57BL ,stomatognathic diseases ,030104 developmental biology ,medicine.anatomical_structure ,STAT protein ,Cancer research ,biology.protein ,Female ,030215 immunology ,Signal Transduction - Abstract
Oropharyngeal candidiasis (OPC) is an opportunistic infection of the oral mucosa caused by the commensal fungus Candida albicans. IL-17 and IL-22 both mediate antifungal immunity yet activate distinct downstream signaling pathways. While much is known about IL-17-dependent immunity in OPC, the activities of IL-22 are less well delineated. We show that induction of Il22 is independent of Dectin-1, CARD9 and aryl hydrocarbon receptor (AhR) and is driven by IL-23 and the C. albicans pore forming peptide candidalysin. Despite similar induction requirements and cellular sources, IL-22 and IL-17 function non-redundantly during OPC and exert opposing roles in neutrophil recruitment. The IL-22 and IL-17 receptors are required in anatomically distinct locations; loss of IL-22RA1 in the oral basal epithelial layer (BEL) but not the suprabasal epithelial layer (SEL) causes susceptibility to OPC, whereas IL-17RA is needed in the SEL. Our data reveal that IL-22 is a major activator of STAT3 in the BEL during OPC. Moreover, loss of STAT3 in the BEL but not the SEL renders mice susceptible to OPC. Transcriptional profiling of RNASeq data linked IL-22/STAT3 to oral epithelial cell proliferation and survival, but also, unexpectedly, to driving an IL-17 gene signature. We show that IL-22 acts on the BEL to replenish the IL-17RA-expressing SEL, thereby restoring the ability of the oral epithelium to respond to IL-17. Consequently, IL-22 signaling in BEL ‘licenses’ IL-17R signaling in the oral epithelium, revealing spatially distinct yet cooperative activities of IL-22 and IL-17 in oral candidiasis. This work also suggests that oral thrush in Jobs’ syndrome patients may be caused by STAT3 impairments in the oral epithelium, not just Th17 cells.
- Published
- 2020
- Full Text
- View/download PDF
16. Isolation of Murine Skin Resident and Migratory Dendritic Cells via Enzymatic Digestion
- Author
-
Daniel H. Kaplan and Sakeen W. Kashem
- Subjects
0301 basic medicine ,Immunology ,chemical and pharmacologic phenomena ,Cell Separation ,Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Dermis ,Antigen ,Cell Movement ,medicine ,Animals ,Antigen-presenting cell ,Lymph node ,Skin ,Innate immune system ,Epidermis (botany) ,Dendritic Cells ,Dendritic cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Female ,Lymph Nodes ,Lymph ,030215 immunology - Abstract
Dendritic cells (DCs) are a highly specialized subset of professional antigen-presenting cells (APCs) that reside in peripheral and lymphoid tissues. DCs capture antigen in the periphery and migrate to the lymph node where they prime naive T cells. In addition, DCs have been recently appreciated to have function in innate immunity within tissues. In the skin, heterogeneous populations of DCs reside within the epidermis and the dermis. Analysis of the cutaneous DC subsets is complicated by requirements of distinct enzymatic digestion protocols for isolation of APCs from distinct anatomical compartments of the skin. Here, specific approaches for isolation of DCs from the epidermis, dermis, and the skin-draining lymph nodes of mice are described. © 2018 by John Wiley & Sons, Inc.
- Published
- 2018
- Full Text
- View/download PDF
17. Effector T
- Author
-
Maria Carolina, Amezcua Vesely, Paris, Pallis, Piotr, Bielecki, Jun Siong, Low, Jun, Zhao, Christian C D, Harman, Lina, Kroehling, Ruaidhrí, Jackson, Will, Bailis, Paula, Licona-Limón, Hao, Xu, Norifumi, Iijima, Padmini S, Pillai, Daniel H, Kaplan, Casey T, Weaver, Yuval, Kluger, Monika S, Kowalczyk, Akiko, Iwasaki, Joao P, Pereira, Enric, Esplugues, Nicola, Gagliani, and Richard A, Flavell
- Subjects
CD4-Positive T-Lymphocytes ,Interleukin-17 ,Article ,Klebsiella Infections ,Mice, Inbred C57BL ,Disease Models, Animal ,Klebsiella pneumoniae ,Mice ,Animals ,Th17 Cells ,Diphtheria Toxin ,Female ,Immunologic Memory ,Lung - Abstract
Adaptive immunity provides life-long protection by generating central and effector memory T cells and the most recently described tissue resident memory (T(RM)) cells. However, the cellular origin of CD4 T(RM) cells, and their contribution to host defense remains elusive. Using IL-17A tracking-fate-mouse models we found that a significant fraction of lung CD4 T(RM) cells derive from IL-17A producing effector (T(H)17) cells following immunization with heat-killed Klebsiella pneumonia (Kp). These exT(H)17 T(RM) cells are maintained in the lung by IL-7, produced by lymphatic endothelial cells. During a memory response, neither antibodies, γδ T cells, nor circulatory T cells are sufficient for the rapid host defense required to eliminate Kp. Conversely, using parabiosis and depletion studies, we demonstrated that exT(H)17 T(RM) cells play an important role in bacterial clearance. Thus, we delineate the origin and function of airway CD4 T(RM) cells during bacterial infection, offering novel strategies for targeted vaccine design.
- Published
- 2018
18. Peripheral tissues reprogram CD8+ T cells for pathogenicity during graft-versus-host disease
- Author
-
Séverine Ciré, Terry K. Means, Matthew Collin, Stephen Henderson, Sophie Ward, Vincent Plagnol, Clare L. Bennett, Simone Dertschnig, Cara Lomas, Thomas Conlan, Sven Blobner, Ivana R. Ferrer, Ronjon Chakraverty, Daniel H. Kaplan, Claire Tkacz, Laura Jardine, Pedro Santos e Sousa, and Heather C. West
- Subjects
Male ,0301 basic medicine ,lcsh:Medicine ,Graft vs Host Disease ,Mice ,Cytotoxic T cell ,Cells, Cultured ,Bone Marrow Transplantation ,Skin ,Receptors, Notch ,Effector ,Hematopoietic Stem Cell Transplantation ,Stem cell transplantation ,General Medicine ,Cellular Reprogramming ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Multigene Family ,Antigens, Surface ,Female ,Stem cell ,Research Article ,T cell ,Primary Cell Culture ,Immunology ,Mice, Transgenic ,Biology ,Proinflammatory cytokine ,03 medical and health sciences ,Immune system ,Antigens, CD ,medicine ,Animals ,Humans ,Transplantation, Homologous ,Lectins, C-Type ,Molecular pathology ,Transplantation Chimera ,Transplantation ,lcsh:R ,T cell development ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,Mannose-Binding Lectins ,030104 developmental biology ,Graft-versus-host disease ,Gene Expression Regulation ,Langerhans Cells ,T-Lymphocytes, Cytotoxic - Abstract
Graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic stem cell transplantation induced by the influx of donor-derived effector T cells (TE) into peripheral tissues. Current treatment strategies rely on targeting systemic T cells; however, the precise location and nature of instructions that program TE to become pathogenic and trigger injury are unknown. We therefore used weighted gene coexpression network analysis to construct an unbiased spatial map of TE differentiation during the evolution of GVHD and identified wide variation in effector programs in mice and humans according to location. Idiosyncrasy of effector programming in affected organs did not result from variation in T cell receptor repertoire or the selection of optimally activated TE. Instead, TE were reprogrammed by tissue-autonomous mechanisms in target organs for site-specific proinflammatory functions that were highly divergent from those primed in lymph nodes. In the skin, we combined the correlation-based network with a module-based differential expression analysis and showed that Langerhans cells provided in situ instructions for a Notch-dependent T cell gene cluster critical for triggering local injury. Thus, the principal determinant of TE pathogenicity in GVHD is the final destination, highlighting the need for target organ–specific approaches to block immunopathology while avoiding global immune suppression., During the evolution of graft-versus-host disease, effector CD8+ T cells are re-programmed by tissue-autonomous mechanisms to develop idiosyncratic pathogenic functions in target organs.
- Published
- 2018
- Full Text
- View/download PDF
19. Selective programming of CCR10+ innate lymphoid cells in skin-draining lymph nodes for cutaneous homeostatic regulation
- Author
-
Na Xiong, Luming Zhao, Jie Yang, Gary H. Perdew, Daniel H. Kaplan, and Shaomin Hu
- Subjects
0301 basic medicine ,Chemokine ,Adoptive cell transfer ,Immunology ,Priming (immunology) ,Receptors, CCR10 ,Article ,03 medical and health sciences ,Chemokine receptor ,Mice ,Immunology and Allergy ,Animals ,Homeostasis ,CCR10 ,Lymphocytes ,skin and connective tissue diseases ,Transcription factor ,Skin ,Mice, Knockout ,biology ,integumentary system ,Innate lymphoid cell ,FOXN1 ,Flow Cytometry ,Adoptive Transfer ,Immunity, Innate ,Cell biology ,body regions ,Mice, Inbred C57BL ,030104 developmental biology ,biology.protein ,Lymph Nodes - Abstract
Innate lymphoid cells (ILCs) 'preferentially' localize into barrier tissues, where they function in tissue protection but can also contribute to inflammatory diseases. The mechanisms that regulate the establishment of ILCs in barrier tissues are poorly understood. Here we found that under steady-state conditions, ILCs in skin-draining lymph nodes (sLNs) were continuously activated to acquire regulatory properties and high expression of the chemokine receptor CCR10 for localization into the skin. CCR10(+) ILCs promoted the homeostasis of skin-resident T cells and, reciprocally, their establishment in the skin required T cell-regulated homeostatic environments. CD207(+) dendritic cells expressing the transcription factor Foxn1 were required for the proper generation of CCR10(+) ILCs. These observations reveal mechanisms that underlie the specific programming and priming of skin-homing CCR10(+) ILCs in the sLNs.
- Published
- 2015
20. Generation of Th17 cells in response to intranasal infection requires TGF-β1 from dendritic cells and IL-6 from CD301b + dendritic cells
- Author
-
Marc K. Jenkins, P. Patrick Cleary, Jonathan L. Linehan, Sakeen W. Kashem, Daniel H. Kaplan, and Thamotharampillai Dileepan
- Subjects
Immunity, Cellular ,Multidisciplinary ,CD40 ,biology ,Follicular dendritic cells ,Interleukin-6 ,Streptococcus pyogenes ,Antigen presentation ,chemical and pharmacologic phenomena ,Dendritic Cells ,Dendritic cell ,Biological Sciences ,Transforming Growth Factor beta1 ,Mice ,Streptococcal Infections ,Nose Diseases ,Immunology ,biology.protein ,Lymph node stromal cell ,Interleukin 12 ,Animals ,Th17 Cells ,Cytotoxic T cell ,Lectins, C-Type ,Antigen-presenting cell - Abstract
Intranasal (i.n.) infections preferentially generate Th17 cells. We explored the basis for this anatomic preference by tracking polyclonal CD4(+) T cells specific for an MHC class II-bound peptide from the mucosal pathogen Streptococcus pyogenes. S. pyogenes MHC class II-bound peptide-specific CD4(+) T cells were first activated in the cervical lymph nodes following i.n. inoculation and then differentiated into Th17 cells. S. pyogenes-induced Th17 formation depended on TGF-β1 from dendritic cells and IL-6 from a CD301b(+) dendritic cell subset located in the cervical lymph nodes but not the spleen. Thus, the tendency of i.n. infection to induce Th17 cells is related to cytokine production by specialized dendritic cells that drain this site.
- Published
- 2015
- Full Text
- View/download PDF
21. The NF‐κB regulator Bcl‐3 modulates inflammation during contact hypersensitivity reactions in radioresistant cells
- Author
-
Wanhu Tang, Estefania Claudio, Sun Saret, Nimisha Rikhi, Ulrich Siebenlist, Daniel H. Kaplan, Hongshan Wang, Hye lin Ha, and Ilaria Tassi
- Subjects
Keratinocytes ,Chemokine ,Transcription, Genetic ,Neutrophils ,Chemokine CXCL2 ,Immunology ,Inflammation ,CD8-Positive T-Lymphocytes ,Chemokine CXCL9 ,Radiation Tolerance ,Article ,Oxazolone ,Mice ,chemistry.chemical_compound ,Immune system ,NF-kappa B p52 Subunit ,B-Cell Lymphoma 3 Protein ,Proto-Oncogene Proteins ,medicine ,Animals ,Immunology and Allergy ,CXCL10 ,Mice, Knockout ,biology ,NF-kappa B p50 Subunit ,NF-κB ,Cell biology ,Chemokine CXCL10 ,Mice, Inbred C57BL ,CXCL2 ,chemistry ,Dermatitis, Allergic Contact ,biology.protein ,Inflammation Mediators ,medicine.symptom ,CD8 ,Transcription Factors - Abstract
Bcl-3 is an atypical member of the IκB family. Bcl-3 functions as a cofactor of p50/NF-κB1 or p52/NF-κB2 homodimers in nuclei, where it modulates NF-κB-regulated transcription in a context-dependent way. Bcl-3 has tumorigenic potential, is critical in host defense of pathogens, and has been reported to ameliorate or exacerbate inflammation, depending on disease model. However, cell-specific functions of Bcl-3 remain largely unknown. Here, we explored the role of Bcl-3 in a contact hypersensitivity (CHS) mouse model, which depends on the interplay between keratinocytes and immune cells. Bcl-3-deficient mice exhibited an exacerbated and prolonged CHS response to oxazolone. Increased inflammation correlated with higher production of chemokines CXCL2, CXCL9 and CXCL10, and consequently increased recruitment of neutrophils and CD8+ T cells. Bone marrow chimera experiments indicated that the ability of Bcl-3 to reduce the CHS response depended on Bcl-3 activity in radioresistant cells. Specific ablation of Bcl-3 in keratinocytes resulted in increased production of CXCL9 and CXCL10 and sustained recruitment of specifically CD8+ T cells. These findings identify Bcl-3 as a critical player during the later stage of the CHS reaction to limit inflammation via actions in radioresistant cells, including keratinocytes.
- Published
- 2015
- Full Text
- View/download PDF
22. Targeting Antigens through Blood Dendritic Cell Antigen 2 on Plasmacytoid Dendritic Cells Promotes Immunologic Tolerance
- Author
-
Kevin E. Draves, Chang Hung Chen, Natalia V. Giltiay, Daniel H. Kaplan, Edward A. Clark, Mark J. Shlomchik, Martha Hayden-Ledbetter, and Craig P. Chappell
- Subjects
media_common.quotation_subject ,T cell ,Plasma Cells ,Immunology ,Mice, Transgenic ,Biology ,T-Lymphocytes, Regulatory ,Article ,Immune tolerance ,Mice ,Antigen ,Immune Tolerance ,medicine ,Animals ,Humans ,Immunology and Allergy ,Lectins, C-Type ,Receptors, Immunologic ,Antigen-presenting cell ,Internalization ,Immunologic Tolerance ,media_common ,Membrane Glycoproteins ,FOXP3 ,hemic and immune systems ,Dendritic Cells ,Dendritic cell ,Cell biology ,medicine.anatomical_structure - Abstract
The C-type lectin receptor blood dendritic cell Ag 2 (BDCA2) is expressed exclusively on human plasmacytoid DCs (pDCs) and plays a role in Ag capture, internalization, and presentation to T cells. We used transgenic mice that express human BDCA2 and anti-BDCA2 mAbs to deliver Ags directly to BDCA2 on pDCs in vivo. Targeting Ag to pDCs in this manner resulted in significant suppression of Ag-specific CD4+ T cell and Ab responses upon secondary exposure to Ag in the presence of adjuvant. Suppression of Ab responses required both a decrease in effector CD4+ T cells and preservation of Foxp3+ regulatory T cells (Tregs). Reduction in Treg numbers following Ag delivery to BDCA2 restored both CD4+ T cell activation and Ab responses, demonstrating that Tregs were required for the observed tolerance. Our results demonstrate that Ag delivery to pDCs through BDCA2 is an effective method to induce immunological tolerance, which may be useful for treating autoimmune diseases or to inhibit unwanted Ab responses.
- Published
- 2014
- Full Text
- View/download PDF
23. BRAF V600E and Pten deletion in mice produces a histiocytic disorder with features of Langerhans cell histiocytosis
- Author
-
Yechaan Joo, Bhumi Patel, Daniel H. Kaplan, David S. Nelson, Barrett J. Rollins, Roderick T. Bronson, Kristen E. Stevenson, Mark J. Shlomchik, Ryan L. Marano, and Sara Y. Tian
- Subjects
Male ,0301 basic medicine ,Physiology ,Artificial Gene Amplification and Extension ,CD8-Positive T-Lymphocytes ,Polymerase Chain Reaction ,White Blood Cells ,Mice ,0302 clinical medicine ,Langerhans cell histiocytosis ,Animal Cells ,Immune Physiology ,Intestine, Small ,Medicine and Health Sciences ,Promoter Regions, Genetic ,Sequence Deletion ,Multidisciplinary ,biology ,Thymus ,Histiocytosis ,Phenotype ,medicine.anatomical_structure ,Lymphatic system ,030220 oncology & carcinogenesis ,Antigens, Surface ,Medicine ,Small Intestine ,Cellular Types ,Anatomy ,Research Article ,Proto-Oncogene Proteins B-raf ,Langerin ,Precursor Cells ,Science ,Immune Cells ,Immunology ,Antigen-Presenting Cells ,Spleen ,Research and Analysis Methods ,Lymphatic System ,03 medical and health sciences ,medicine ,Animals ,Humans ,PTEN ,Molecular Biology Techniques ,Molecular Biology ,Histiocyte ,Cell Proliferation ,Blood Cells ,PTEN Phosphohydrolase ,Biology and Life Sciences ,Histiocytes ,Cell Biology ,Dendritic Cells ,medicine.disease ,Gastrointestinal Tract ,Histiocytosis, Langerhans-Cell ,030104 developmental biology ,Immune System ,Langerhans Cells ,Cancer research ,biology.protein ,Lymph Nodes ,Digestive System ,CD8 - Abstract
Langerhans cell histiocytosis (LCH) is characterized by the accumulation of Langerin (CD207)-expressing histiocytes. Mutational activation of mitogen-activated protein kinase pathway genes, in particular BRAF, drives most cases. To test whether activated BRAF is sufficient for the development of LCH, we engineered mice to express BRAF V600E under the control of the human Langerin promoter. These mice have shortened survivals, smaller lymphoid organs, absent Leydig cells, and fewer epidermal LCs than controls, but do not accumulate histiocytes. To test whether the absence of histiocyte proliferation could be due to oncogene-induced senescence, we engineered homozygous Pten loss in the same cells that expressed BRAF V600E. Like mice with intact Pten, these mice have shortened survivals, smaller thymi, and absent Leydig cells. However, loss of Pten also leads to the accumulation of CD207+ histiocytes in spleen, thymus, and some lymph nodes. While many CD207+ histiocytes in the thymus are CD8-, reminiscent of LCH cells, the CD207+ histiocytes in the spleen and lymph nodes are CD8+. These mice also accumulate large numbers of CD207- cells in the lamina propria (LP) of the small intestine. Both the lymphoid and LP phenotypes are likely due to human Langerin promoter-driven BRAF V600E expression in resident CD8+ dendritic cells in the former and LP dendritic cells in the latter and confirm that Pten loss is required to overcome inhibitory pathways induced by BRAF V600E expression. The complex phenotype of these mice is a consequence of the multiple murine cell types in which the human Langerin promoter is active.
- Published
- 2019
- Full Text
- View/download PDF
24. Intestinal lamina propria dendritic cells maintain T cell homeostasis but do not affect commensalism
- Author
-
Christopher Staley, Michael J. Sadowsky, Nathan E. Welty, Nico Ghilardi, Daniel H. Kaplan, and Botond Z. Igyártó
- Subjects
Langerin ,Transgene ,Immunology ,Cell ,Mice, Transgenic ,chemical and pharmacologic phenomena ,T-Lymphocytes, Regulatory ,Article ,Immunophenotyping ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Intestinal mucosa ,Antigens, CD ,T-Lymphocyte Subsets ,medicine ,Animals ,Homeostasis ,Humans ,Immunology and Allergy ,Diphtheria Toxin ,Lectins, C-Type ,Intestinal Mucosa ,030304 developmental biology ,0303 health sciences ,CD11b Antigen ,biology ,hemic and immune systems ,Dendritic Cells ,3. Good health ,Cell biology ,Mannose-Binding Lectins ,Phenotype ,medicine.anatomical_structure ,biology.protein ,Metagenome ,Th17 Cells ,Integrin alpha Chains ,030215 immunology ,Homing (hematopoietic) - Abstract
Targeted deletion of CD103+CD11b+ LP DCs results in reduced LP Th17 cells at steady state, but has no impact on Citrobacter infection or the composition of the intestinal microbiota., Dendritic cells (DCs) in the intestinal lamina propria (LP) are composed of two CD103+ subsets that differ in CD11b expression. We report here that Langerin is expressed by human LP DCs and that transgenic human langerin drives expression in CD103+CD11b+ LP DCs in mice. This subset was ablated in huLangerin-DTA mice, resulting in reduced LP Th17 cells without affecting Th1 or T reg cells. Notably, cognate DC–T cell interactions were not required for Th17 development, as this response was intact in huLangerin-Cre I-Aβfl/fl mice. In contrast, responses to intestinal infection or flagellin administration were unaffected by the absence of CD103+CD11b+ DCs. huLangerin-DTA x BatF3−/− mice lacked both CD103+ LP DC subsets, resulting in defective gut homing and fewer LP T reg cells. Despite these defects in LP DCs and resident T cells, we did not observe alterations of intestinal microbial communities. Thus, CD103+ LP DC subsets control T cell homeostasis through both nonredundant and overlapping mechanisms.
- Published
- 2013
- Full Text
- View/download PDF
25. Antigen presentation by Langerhans cells
- Author
-
Daniel H. Kaplan and Botond Z. Igyártó
- Subjects
CD4-Positive T-Lymphocytes ,Immunology ,Antigen presentation ,CD1 ,Adaptive Immunity ,Lymphocyte Activation ,Article ,Animals, Genetically Modified ,Mice ,Antigen ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Antigen-presenting cell ,Histiocyte ,Skin ,Antigen Presentation ,CD40 ,biology ,Interleukin-17 ,Cell Differentiation ,Acquired immune system ,Cell biology ,Cellular Microenvironment ,Langerhans Cells ,Host-Pathogen Interactions ,biology.protein - Abstract
Langerhans cells and other skin-resident dendritic cells (DC) are required for the development of cutaneous adaptive immune responses. In vivo experiments using mice with selective DC-subset deficiencies and ex vivo experiments using isolated DC suggests that each subset makes a unique contribution to the adaptive response. This review focuses on the functional outcome of antigen presentation by Langerhans cells. Special attention is given to their ability to promote CD4 T cell differentiation in a variety of inflammatory contexts and whether this subset has the capacity to cross-prime CD8 T cells.
- Published
- 2013
- Full Text
- View/download PDF
26. Neoantigen Expression in Steady-State Langerhans Cells Induces CTL Tolerance
- Author
-
Helen, Strandt, Douglas Florindo, Pinheiro, Daniel H, Kaplan, Dagmar, Wirth, Iris Karina, Gratz, Peter, Hammerl, Josef, Thalhamer, and Angelika, Stoecklinger
- Subjects
Antigen Presentation ,integumentary system ,Ovalbumin ,hemic and immune systems ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Lymphocyte Activation ,Autoantigens ,T-Lymphocytes, Regulatory ,Mice, Inbred C57BL ,Immune Regulation ,Mice ,Tamoxifen ,Gene Expression Regulation ,Langerhans Cells ,Models, Animal ,Immune Tolerance ,Animals ,Immunologic Memory ,Cells, Cultured ,Skin ,T-Lymphocytes, Cytotoxic - Abstract
The skin hosts a variety of dendritic cells (DCs), which act as professional APC to control cutaneous immunity. Langerhans cells (LCs) are the only DC subset in the healthy epidermis. However, due to the complexity of the skin DC network, their relative contribution to either immune activation or immune tolerance is still not entirely understood. To specifically study the function of LCs in vivo, without altering the DC subset composition in the skin, we have generated transgenic mouse models for tamoxifen-inducible de novo expression of Ags in LCs but no other langerin+ DCs. Therefore, this system allows for LC-restricted Ag presentation to T cells. Presentation of nonsecreted OVA (GFPOVA) by steady-state LCs resulted in transient activation of endogenous CTL in transgenic mice. However, when these mice were challenged with OVA by gene gun immunization in the contraction phase of the primary CTL response they did not respond with a recall of CTL memory but, instead, with robust Ag-specific CTL tolerance. We found regulatory T cells (Tregs) enriched in the skin of tolerized mice, and depletion of Tregs or adoptive experiments revealed that Tregs were critically involved in CTL tolerance. By contrast, when OVA was presented by activated LCs, a recallable CTL memory response developed in transgenic mice. Thus, neoantigen presentation by epidermal LCs results in either robust CTL tolerance or CTL memory, and this decision-making depends on the activation state of the presenting LCs.
- Published
- 2016
27. Generation of a NK1R-CreER knockin mouse strain to study cells involved in Neurokinin 1 Receptor signaling
- Author
-
Huizhen, Huang, Marissa S, Kuzirian, Xiaoyun, Cai, Lindsey M, Snyder, Jonathan, Cohen, Daniel H, Kaplan, and Sarah E, Ross
- Subjects
Neurons ,Mice ,Gene Expression Regulation ,Integrases ,Spinal Cord ,Animals ,Pain ,Gene Knock-In Techniques ,Receptors, Neurokinin-1 ,Substance P ,Article ,Signal Transduction - Abstract
The Neurokinin 1 Receptor (NK1R), which binds Substance P, is expressed in discrete populations of neurons throughout the nervous system, where it has numerous roles including the modulation of pain and affective behaviors. Here, we report the generation of a NK1R-CreER knockin allele, in which CreERT2 replaces the coding sequence of the TACR1 gene (encoding NK1R) in order to gain genetic access to these cells. We find that the NK1R-CreER allele mediates recombination in many regions of the nervous system that are important in pain and anxiety including the amygdala, hypothalamus, frontal cortex, raphe nucleus, and dorsal horn of the spinal cord. Other cell types that are labeled by this allele include amacrine cells in the retina and fibroblasts in the skin. Thus, the NK1R-CreER mouse line is a valuable new tool for conditional gene manipulation enabling the visualization and manipulation of cells that express NK1R.
- Published
- 2016
28. Langerhans Cells Require MyD88-Dependent Signals for Candida albicans Response but Not for Contact Hypersensitivity or Migration
- Author
-
Daniel H. Kaplan, Dominik Schenten, Daniela Ortner, Krystal M. Haley, Sakeen W. Kashem, Botond Z. Igyártó, and Aleh Bobr
- Subjects
Antigens, Fungal ,Immunology ,Dermatitis, Contact ,Lymphocyte Activation ,Article ,Microbiology ,Mice ,Antigen ,Cell Movement ,Candida albicans ,medicine ,Animals ,Dermatomycoses ,Immunology and Allergy ,Sensitization ,Mice, Knockout ,Epidermis (botany) ,biology ,Interleukins ,Candidiasis ,Fungal genetics ,biology.organism_classification ,Acquired immune system ,Corpus albicans ,Cell biology ,medicine.anatomical_structure ,Langerhans Cells ,Myeloid Differentiation Factor 88 ,Th17 Cells ,Haptens ,Hapten ,Signal Transduction - Abstract
Langerhans cells (LC) are a subset of skin-resident dendritic cells (DC) that reside in the epidermis as immature DC, where they acquire Ag. A key step in the life cycle of LC is their activation into mature DC in response to various stimuli, including epicutaneous sensitization with hapten and skin infection with Candida albicans. Mature LC migrate to the skin-draining LN, where they present Ag to CD4 T cells and modulate the adaptive immune response. LC migration is thought to require the direct action of IL-1β and IL-18 on LC. In addition, TLR ligands are present in C. albicans, and hapten sensitization produces endogenous TLR ligands. Both could contribute to LC activation. We generated Langerin-Cre MyD88fl mice in which LC are insensitive to IL-1 family members and most TLR ligands. LC migration in the steady state, after hapten sensitization and postinfection with C. albicans, was unaffected. Contact hypersensitivity in Langerin-Cre MyD88fl mice was similarly unaffected. Interestingly, in response to C. albicans infection, these mice displayed reduced proliferation of Ag-specific CD4 T cells and defective Th17 subset differentiation. Surface expression of costimulatory molecules was intact on LC, but expression of IL-1β, IL-6, and IL-23 was reduced. Thus, sensitivity to MyD88-dependent signals is not required for LC migration, but is required for the full activation and function of LC in the setting of fungal infection.
- Published
- 2012
- Full Text
- View/download PDF
29. Langerhans Cells Facilitate Epithelial DNA Damage and Squamous Cell Carcinoma
- Author
-
Daniel H. Kaplan, Jason H. Neustadter, Lining Cai, Badri Modi, Robert E. Tigelaar, Elisa Binda, Scott J. Roberts, Peter P. Fu, Adrian Hayday, Michael Girardi, Mark J. Shlomchik, Bernice Y. Kwong, Anjela Galan, Renata B. Filler, John D. Overton, Swapna Reddy, and Julia M. Lewis
- Subjects
Keratinocytes ,endocrine system ,Skin Neoplasms ,DNA damage ,9,10-Dimethyl-1,2-benzanthracene ,T-Lymphocytes ,CD1 ,DMBA ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Article ,Mice ,Immune system ,polycyclic compounds ,Cytochrome P-450 CYP1A1 ,medicine ,Animals ,Humans ,HRAS ,skin and connective tissue diseases ,neoplasms ,Cells, Cultured ,Carcinogen ,Multidisciplinary ,integumentary system ,Dendritic cell ,Cell Transformation, Neoplastic ,Genes, ras ,Biochemistry ,Langerhans Cells ,Cytochrome P-450 CYP1B1 ,Carcinogens ,Carcinoma, Squamous Cell ,Cancer research ,Aryl Hydrocarbon Hydroxylases ,Carcinogenesis ,DNA Damage - Abstract
The Dark Side of Langerhans Cells Several immune cell populations reside in the skin and are thought to provide a protective barrier against infections and to act as sentinels against malignant transformation. However, studies in mice that lack Langerhans cells, a subset of dendritic cells, have suggested that these cells may actually promote tumorigenesis. Using a mouse model of squamous cell carcinoma, Modi et al. (p. 104 ) now reveal how Langerhans cells may promote the transformation of skin epithelial cells. In response to the carcinogen 7,12-dimethylbenz[α]anthracene (DMBA), Langerhans cells increased their expression of the cytochrome P-450 enzyme CYP1B1, which can metabolize DMBA to the mutagenic DMBA- trans -3,4-diol. Thus, besides their functions in regulating the adaptive immune response, Langerhans cells may participate in the metabolism of environmental carcinogens.
- Published
- 2012
- Full Text
- View/download PDF
30. Kappa Opioid Receptor Distribution and Function in Primary Afferents
- Author
-
Elizabeth I. Sypek, Robert L. Friedman, Sarah E. Ross, Michael C. Chiang, Emanuel Loeza-Alcocer, Frank Porreca, H. Richard Koerber, Brian M. Davis, Margaret C. Wright, Jenna R. Gale, Xiaoyun Cai, Toshiro Hirai, Yu Omori, Vidhya Nagarajan, Grégory Scherrer, Stephanie Fulton, Yeon Sun Lee, Tayler D. Sheahan, Kimberly A. Meerschaert, Daniel H. Kaplan, Peter C. Adelman, Lindsey M. Snyder, Zeyu Hu, Melissa Giraldo Duque, Junichi Hachisuka, Huizhen Huang, and Michael S. Gold
- Subjects
0301 basic medicine ,Pain ,Mice, Transgenic ,Sensory system ,Dynorphin ,Biology ,Neurotransmission ,Somatosensory system ,κ-opioid receptor ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Pain Management ,Neurons, Afferent ,Neurons ,Neurogenic inflammation ,Receptors, Opioid, kappa ,General Neuroscience ,Nociceptors ,Axons ,030104 developmental biology ,Nociceptor ,Neuroscience ,030217 neurology & neurosurgery ,Nalfurafine ,Signal Transduction ,medicine.drug - Abstract
Primary afferents are known to be inhibited by kappa opioid receptor (KOR) signaling. However, the specific types of somatosensory neurons that express KOR remain unclear. Here, using a newly developed KOR-cre knockin allele, viral tracing, single-cell RT-PCR, and ex vivo recordings, we show that KOR is expressed in several populations of primary afferents: a subset of peptidergic sensory neurons, as well as low-threshold mechanoreceptors that form lanceolate or circumferential endings around hair follicles. We find that KOR acts centrally to inhibit excitatory neurotransmission from KOR-cre afferents in laminae I and III, and this effect is likely due to KOR-mediated inhibition of Ca2+ influx, which we observed in sensory neurons from both mouse and human. In the periphery, KOR signaling inhibits neurogenic inflammation and nociceptor sensitization by inflammatory mediators. Finally, peripherally restricted KOR agonists selectively reduce pain and itch behaviors, as well as mechanical hypersensitivity associated with a surgical incision. These experiments provide a rationale for the use of peripherally restricted KOR agonists for therapeutic treatment.
- Published
- 2018
- Full Text
- View/download PDF
31. Insights into Langerhans cell function from Langerhans cell ablation models
- Author
-
Björn E. Clausen, Daniel H. Kaplan, and Adrien Kissenpfennig
- Subjects
Langerhans cell ,Langerin ,Green Fluorescent Proteins ,Immunology ,Population ,Antigen presentation ,Mice, Transgenic ,Dermatitis, Contact ,Mice ,Immune system ,medicine ,Animals ,Immunology and Allergy ,Skin immunity ,Lectins, C-Type ,education ,Skin ,Antigen Presentation ,education.field_of_study ,Epidermis (botany) ,biology ,integumentary system ,Dendritic Cells ,Dendritic cell ,Cell biology ,Mannose-Binding Lectins ,medicine.anatomical_structure ,Langerhans Cells ,Antigens, Surface ,Models, Animal ,biology.protein - Abstract
Langerhans cells (LC) are the principal dendritic cell (DC) population in the epidermis of the skin. Owing to their prominent position at the environmental barrier, LC have long been considered to be prototypic sentinel DC. More recently, the precise role of LC in the initiation and control of cutaneous immune responses has become debatable. To elucidate their contribution to immune regulation in the skin, our laboratories have generated genetically modified mice in which LC can be followed in situ by expression of enhanced green fluorescent protein and can be either inducibly or constitutively depleted in vivo. This review highlights the similarities and differences between these mouse models, discusses the discovery and functional significance of Langerin(+) dermal DC, and examines some recent data that help to shed light on LC function.
- Published
- 2008
- Full Text
- View/download PDF
32. Autocrine/paracrine TGFβ1 is required for the development of epidermal Langerhans cells
- Author
-
Matthew C. Jenison, Richard A. Flavell, Mark J. Shlomchik, Daniel H. Kaplan, Ming O. Li, and Warren D. Shlomchik
- Subjects
Langerhans cell ,Langerin ,Immunology ,Cre recombinase ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Transforming Growth Factor beta1 ,Mice ,03 medical and health sciences ,Paracrine signalling ,Chimera (genetics) ,0302 clinical medicine ,Antigens, CD ,Genes, Reporter ,Skin Physiological Phenomena ,medicine ,Animals ,Humans ,Immunology and Allergy ,Lectins, C-Type ,Autocrine signalling ,030304 developmental biology ,Recombination, Genetic ,0303 health sciences ,Integrases ,integumentary system ,biology ,Brief Definitive Report ,hemic and immune systems ,Cell Biology ,Molecular biology ,Cell biology ,Transplantation ,Haematopoiesis ,Mannose-Binding Lectins ,medicine.anatomical_structure ,Langerhans Cells ,biology.protein ,Brief Definitive Reports ,Epidermis ,030215 immunology - Abstract
Langerhans cells (LCs) are bone marrow (BM)-derived epidermal dendritic cells (DCs) that develop from precursors found in the dermis. Epidermal LCs are absent in transforming growth factor (TGF) beta1-deficient mice. It is not clear whether TGFbeta1 acts directly on LC precursors to promote maturation or whether it acts on accessory cells, which in turn affect LC precursors. In addition, the physiologic source of TGFbeta1 is uncertain because BM chimera experiments showed that neither hematopoietic nor nonhematopoietic-derived TGFbeta1 is required for LC development. To address these issues, we created mice transgenic for a bacterial artificial chromosome (BAC) containing the gene for human Langerin into which Cre recombinase had been inserted by homologous recombination (Langerin-Cre). These mice express Cre selectively in LCs, and they were bred to floxed TGFbetaRII and TGFbeta1 mice, thereby generating mice with LCs that either cannot respond to or generate TGFbeta1, respectively. Langerin-Cre TGFbetaRII mice had substantially reduced numbers of epidermal LCs, demonstrating that TGFbeta1 acts directly on LCs in vivo. Interestingly, Langerin-Cre TGFbeta1 mice also had very few LCs both in the steady state and after BM transplantation. Thus, TGFbeta1 derived from LCs acts directly on LCs through an autocrine/paracrine loop, and it is required for LC development and/or survival.
- Published
- 2007
- Full Text
- View/download PDF
33. Candida albicans morphology and dendritic cell subsets determine T helper cell differentiation
- Author
-
Maryam Gerami-Nejad, Akiko Iwasaki, Daniel H. Kaplan, Gordon D. Brown, Yosuke Kumamoto, Gerard Zurawski, Elizabeth S. Jarrett, Judith Berman, Botond Z. Igyártó, Sakeen W. Kashem, Rebecca A. Drummond, Javed Mohammed, and Sandra Zurawski
- Subjects
Cellular differentiation ,Cell ,Immunology ,Biology ,Lymphocyte Activation ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immunity ,Candida albicans ,medicine ,T-helper cell differentiation ,Immunology and Allergy ,Animals ,Lectins, C-Type ,030304 developmental biology ,Skin ,Mice, Knockout ,0303 health sciences ,Interleukin-6 ,Candidiasis, Chronic Mucocutaneous ,Cell Differentiation ,T helper cell ,Dendritic cell ,Dendritic Cells ,Th1 Cells ,biology.organism_classification ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Repressor Proteins ,medicine.anatomical_structure ,Infectious Diseases ,Basic-Leucine Zipper Transcription Factors ,Langerhans Cells ,Th17 Cells ,Dimorphic fungus ,030215 immunology - Abstract
Summary Candida albicans is a dimorphic fungus responsible for chronic mucocutaneous and systemic infections. Mucocutaneous immunity to C. albicans requires T helper 17 (Th17) cell differentiation that is thought to depend on recognition of filamentous C. albicans . Systemic immunity is considered T cell independent. Using a murine skin infection model, we compared T helper cell responses to yeast and filamentous C. albicans . We found that only yeast induced Th17 cell responses through a mechanism that required Dectin-1-mediated expression of interleukin-6 (IL-6) by Langerhans cells. Filamentous forms induced Th1 without Th17 cell responses due to the absence of Dectin-1 ligation. Notably, Th17 cell responses provided protection against cutaneous infection while Th1 cell responses provided protection against systemic infection. Thus, C. albicans morphology drives distinct T helper cell responses that provide tissue-specific protection. These findings provide insight into compartmentalization of Th cell responses and C. albicans pathogenesis and have critical implications for vaccine strategies.
- Published
- 2015
34. Epidermal Langerhans Cell-Deficient Mice Develop Enhanced Contact Hypersensitivity
- Author
-
Warren D. Shlomchik, Daniel H. Kaplan, Mathew C. Jenison, Mark J. Shlomchik, and S Saeland
- Subjects
Adoptive cell transfer ,Chromosomes, Artificial, Bacterial ,Langerin ,Population ,Immunology ,Oligonucleotides ,Priming (immunology) ,Fluorescent Antibody Technique ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Dermatitis, Contact ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Antigens, CD ,Skin immunity ,Animals ,Immunology and Allergy ,Diphtheria Toxin ,Lectins, C-Type ,Regulatory Elements, Transcriptional ,education ,030304 developmental biology ,Diphtheria toxin ,0303 health sciences ,education.field_of_study ,biology ,integumentary system ,hemic and immune systems ,Dendritic cell ,Flow Cytometry ,Adoptive Transfer ,Immunohistochemistry ,3. Good health ,Cell biology ,Mannose-Binding Lectins ,Infectious Diseases ,Langerhans Cells ,biology.protein ,030215 immunology - Abstract
SummaryEpidermal Langerhans cells (LCs), a distinct skin-resident dendritic cell population, acquire antigen in the skin and migrate to draining lymph nodes where they are thought to initiate adaptive immune responses. To examine the functional requirement of LCs in skin immunity, we generated BAC transgenic mice in which the regulatory elements from human Langerin were used to drive expression of diphtheria toxin. The resulting mice have a constitutive and durable absence of epidermal LCs but are otherwise intact. Unexpectedly, we found that contact hypersensitivity (CHS) was amplified rather than abrogated in the absence of LCs. Moreover, we showed that LCs act during the priming and not the effector phase. Thus, LCs not only were dispensable for CHS, but they served to regulate the response, a previously unappreciated function.
- Published
- 2005
- Full Text
- View/download PDF
35. Cutting edge: Failure of antigen-specific CD4+ T cell recruitment to the kidney during systemic candidiasis
- Author
-
Delyth M. Reid, Daniel H. Kaplan, Sing Sing Way, Carol Wallace, Gordon D. Brown, and Rebecca A. Drummond
- Subjects
CD4-Positive T-Lymphocytes ,Immunology ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Kidney ,Mice ,Antigen ,Immunity ,Cell Movement ,Candida albicans ,medicine ,Immunology and Allergy ,Animals ,Antigens ,Cells, Cultured ,Innate immune system ,biology ,Candidiasis ,biology.organism_classification ,medicine.disease ,Corpus albicans ,Immunity, Innate ,3. Good health ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cytoprotection ,Liposomes ,Th17 Cells ,Female ,Cutting Edge ,Systemic candidiasis ,CD8 - Abstract
Candida albicans is the leading cause of systemic candidiasis, a fungal disease associated with high mortality and poor treatment options. The kidney is the target organ during infection and whose control is largely dependent on innate immunity, because lymphocytes appear redundant for protection. In this article, we show that this apparent redundancy stems from a failure of Ag-specific CD4+ T cells to migrate into infected kidneys. In contrast, Ag-specific CD8+ T cells are recruited normally. Using Ag-loaded immunoliposomes to artificially reverse this defective migration, we show that recruited Ag-specific CD4+ T cells polarize toward a Th17 phenotype in the kidney and are protective during fungal infection. Therefore, our data explain the redundancy of CD4+ T cells for defense against systemic infection with C. albicans and have important implications for our understanding of antifungal immunity and the control of renal infections.
- Published
- 2014
36. Immune mediated shaping of microflora community composition depends on barrier site
- Author
-
Daniel H. Kaplan, Michael J. Sadowsky, Felix Scholz, and Brian D. Badgley
- Subjects
Mouse ,lcsh:Medicine ,Adaptive Immunity ,Monocytes ,Mice ,0302 clinical medicine ,Intestinal mucosa ,Intestinal Mucosa ,lcsh:Science ,Skin ,0303 health sciences ,Multidisciplinary ,biology ,Ecology ,Microbiota ,Animal Models ,Acquired immune system ,Innate Immunity ,Host-Pathogen Interaction ,medicine.anatomical_structure ,Community Ecology ,Medicine ,Research Article ,Langerhans cell ,Colon ,Immune Cells ,Immunology ,Dermatology ,Microbiology ,Immune Activation ,03 medical and health sciences ,Immune system ,Model Organisms ,medicine ,Animals ,Microbiome ,Biology ,030304 developmental biology ,Homeodomain Proteins ,Mouth ,Epidermis (botany) ,Host (biology) ,lcsh:R ,Immunity ,biology.organism_classification ,Immunity, Innate ,Mice, Inbred C57BL ,Adaptor Proteins, Vesicular Transport ,Immune System ,Langerhans Cells ,Myeloid Differentiation Factor 88 ,lcsh:Q ,Bacteria ,030215 immunology - Abstract
Barrier surfaces, such as the intestinal lining and the skin, are colonized by a diverse community of commensal microorganisms. Although commensal microorganisms clearly impact the host immune system, whether the immune system also shapes the commensal community is poorly understood. We used 16S rDNA deep sequencing to test whether mice with specific immune defects have an altered commensal microflora. Initially, skin swabs were obtained from wild-type and Langerhans Cell (LC) deficient mice. Despite the intimate contacts that LC make with the upper epidermis, no significant differences were observed in microbial community composition. Similarly, the skin of MyD88/TRIF(-/-), Rag1(-/-) and heterozygous littermate controls showed no alteration in their commensal communities. Next we examined mouth swabs and feces. We did not find a difference in the MyD88/TRIF(-/-) mice. However, we did observe a significant shift in the microbial composition in the feces and mouths of Rag1(-/-) mice. Thus, we conclude that the adaptive immune system modulates the microbial composition at mucosal surfaces in the steady-state but LC, adaptive immunity, and MyD88-dependent innate responses do not affect the skin microbiome revealing a major distinction between barrier sites.
- Published
- 2014
37. Targeted Disruption of the Stat1 Gene in Mice Reveals Unexpected Physiologic Specificity in the JAK–STAT Signaling Pathway
- Author
-
Marco A Meraz, Karen Carver-Moore, J. Michael White, Scott J. Rodig, Raymond N. DuBois, Kathleen C. F. Sheehan, Erika A. Bach, Michel Aguet, Ross G. Clark, Dayle Campbell, Anand S. Dighe, Robert D. Schreiber, Daniel H. Kaplan, Andrew C. Greenlund, and Joan K. Riley
- Subjects
Lipopolysaccharides ,Male ,Molecular Sequence Data ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Vesicular stomatitis Indiana virus ,Interferon-gamma ,Mice ,Cytopathogenic Effect, Viral ,Animals ,Humans ,STAT1 ,STAT2 ,Mice, Knockout ,Base Sequence ,Biochemistry, Genetics and Molecular Biology(all) ,Gene targeting ,JAK-STAT signaling pathway ,Interferon-alpha ,In vitro ,Cell biology ,Rats ,DNA-Binding Proteins ,Mice, Inbred C57BL ,STAT1 Transcription Factor ,Interferon-Stimulated Gene Factor 3 ,Gene Targeting ,STAT protein ,biology.protein ,Trans-Activators ,Cytokines ,Female ,Signal transduction ,DNA Probes ,Signal Transduction - Abstract
The JAK-STAT signaling pathway has been implicated in mediating biological responses induced by many cytokines. However, cytokines that promote distinct cellular responses often activate identical STAT proteins, thereby raising the question of how specificity is manifest within this signaling pathway. Here we report the generation and characterization of mice deficient in STAT1. STAT1-deficient mice show no overt developmental abnormalities, but display a complete lack of responsiveness to either IFN alpha or IFN gamma and are highly sensitive to infection by microbial pathogens and viruses. In contrast, these mice respond normally to several other cytokines that activate STAT1 in vitro. These observations document that STAT1 plays an obligate and dedicated role in mediating IFN-dependent biologic responses and reveal an unexpected level of physiologic specificity for STAT1 action.
- Published
- 1996
- Full Text
- View/download PDF
38. Noncovalent assembly of anti-dendritic cell antibodies and antigens for evoking immune responses in vitro and in vivo
- Author
-
Felix Scholz, Anne-Laure Flamar, Sandra Zurawski, Ling Ni, Gerard Zurawski, Xiao Hua Li, Eynav Klechevsky, Daniel H. Kaplan, Ingrid Gayet, John P. Quinn, Jacques Banchereau, and Sangkon Oh
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,Recombinant Fusion Proteins ,Immunology ,Hemagglutinin (influenza) ,Dockerin ,Mice, Transgenic ,Antigen-Antibody Complex ,CD8-Positive T-Lymphocytes ,Antibodies ,Article ,Mice ,Antigen ,medicine ,Immunology and Allergy ,Animals ,Humans ,Molecular Targeted Therapy ,Antigens ,B cell ,Cells, Cultured ,Mice, Inbred BALB C ,biology ,fungi ,Cell Differentiation ,Dendritic cell ,Dendritic Cells ,Molecular biology ,Fusion protein ,Cell biology ,medicine.anatomical_structure ,HEK293 Cells ,biology.protein ,Rab - Abstract
Targeting antigens directly to DCs through anti-DC receptor antibody fused to antigen proteins is a promising approach to vaccine development. However, not all antigens can be expressed as a recombinant antibody directly fused to a protein antigen. Here, we show that non-covalent assembly of antibody - antigen complexes, mediated by interaction between dockerin and cohesin domains from cellulose-degrading bacteria, can greatly expand the range of antigens for this DC-targeting vaccine technology. Recombinant antibodies with a dockerin domain fused to the antibody heavy chain C-terminus are efficiently secreted by mammalian cells, while many antigens not secreted as antibody fusion proteins are readily expressed as cohesin directly fused to antigen either via secretion from mammalian cells, or as soluble cytoplasmic E. coli products. These form very stable and homogeneous complexes with antibody fused to dockerin. In vitro, these complexes can efficiently bind to human DC receptors followed by presentation to antigen-specific CD4+ and CD8+ T cells. Low doses of the HA1 subunit of Influenza hemagglutinin conjugated through this means to anti-Langerin antibodies elicited Flu HA1-specific antibody and T cell responses in mice. Thus, the non-covalent assembly of antibody and antigen through dockerin and cohesin interaction provides a useful modular strategy for developing and testing prototype vaccines for eliciting antigen-specific T and B cell responses, particularly when direct antibody fusions to antigen cannot be expressed.
- Published
- 2012
39. Autocrine/paracrine TGF-β1 inhibits Langerhans cell migration
- Author
-
Krystal M. Haley, Ming O. Li, Richard A. Flavell, Daniel H. Kaplan, Botond Z. Igyártó, and Aleh Bobr
- Subjects
Multidisciplinary ,Langerhans cell ,Epidermis (botany) ,integumentary system ,Transgene ,Mice, Transgenic ,Biology ,Biological Sciences ,Proinflammatory cytokine ,Cell biology ,Cell Line ,Transforming Growth Factor beta1 ,Paracrine signalling ,Mice ,medicine.anatomical_structure ,Cell Movement ,Langerhans Cells ,Immunology ,medicine ,Animals ,Homeostasis ,Signal transduction ,Receptor ,Autocrine signalling ,Signal Transduction - Abstract
Langerhans cells (LCs) are skin-resident dendritic cells (DC) located in the epidermis that migrate to skin-draining lymph nodes during the steady state and in response to inflammatory stimuli. TGF-β1 is a critical immune regulator that is highly expressed by LCs. The ability to test the functional importance of LC-derived TGF-β1 is complicated by the requirement of TGF-β1 for LC development and by the absence of LCs in mice with an LC-specific ablation of TGF-β1 or its receptor. To overcome these problems, we have engineered transgenic huLangerin-CreER T2 mice that allow for inducible LC-specific excision. Highly efficient and LC-specific expression was confirmed in mice bred onto a YFP Cre reporter strain. We next generated huLangerin-CreER T2 × TGF-βRII fl and huLangerin-CreER T2 × TGF-β1 fl mice. Excision of the TGFβRII or TGFβ1 genes induced mass migration of LCs to the regional lymph node. Expression of costimulatory markers and inflammatory cytokines was unaffected, consistent with homeostatic migration. In addition, levels of p-SMAD2/3 were decreased in LCs from wild-type mice before inflammation-induced migration. We conclude that TGF-β1 acts directly on LCs in an autocrine/paracrine manner to inhibit steady-state and inflammation-induced migration. This is a readily targetable pathway with potential therapeutic implications for skin disease.
- Published
- 2012
40. Characterization of Langerin-expressing dendritic cell subsets in the normal cornea
- Author
-
Hiroki Ueno, Daniel H. Kaplan, Daniel R. Saban, Takaaki Hattori, Reza Dana, Sunil K. Chauhan, and Hyun Soo Lee
- Subjects
Male ,Langerin ,genetic structures ,Corneal Stroma ,Population ,CD11c ,chemical and pharmacologic phenomena ,Major histocompatibility complex ,Polymerase Chain Reaction ,Cornea ,Mice ,medicine ,Animals ,Lectins, C-Type ,RNA, Messenger ,education ,Corneal epithelium ,education.field_of_study ,biology ,integumentary system ,hemic and immune systems ,Dendritic cell ,Dendritic Cells ,Articles ,Flow Cytometry ,Immunohistochemistry ,eye diseases ,CD8A ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Mannose-Binding Lectins ,Gene Expression Regulation ,Microscopy, Fluorescence ,Immunology ,Antigens, Surface ,biology.protein ,sense organs - Abstract
Langerin is a c-type lectin expressed by specific dendritic cell (DC) populations, and it recognizes glycosylated patterns on pathogens such as mycobacteria. It was initially thought that Langerin is expressed only by Langerhans cells (LCs), a unique population of DCs originally identified in the epidermis that have Birbeck granules.1,2 Recently, however, other DC (CD11c+) populations distinct from LCs were also found to express Langerin.3–7 Such (non-LC) Langerin+ DCs include Langerin+ DCs in the dermis,3–5 a subset of CD8a+ DCs in lymphoid tissues,6,7 and distinct populations in tissues of the lung, gut, kidney, and liver.8 Collectively, (non-LC) Langerin+ DCs are mostly CD11blowCD103+, albeit CD11b+CD103low have also been described.8,9 Furthermore, (non-LC) Langerin+ DCs (often referred to elsewhere as CD103+ DCs) have recently become recognized for their proficiency at antigen cross-presentation through major histocompatibility complex (MHC) class I molecules and, thus, are thought to be critical in host defense against viral infection.10 In the skin, (non-LC) dermal Langerin+ DCs differ from LCs in their capacity to prime specific T-cell subsets.11,12 In addition, (non-LC) dermal Langerin+ DCs are crucial in mounting contact hypersensitivity (CHS), whereas LCs in the epidermis have been shown to play a tolerogenic role and thereby to counteract (non-LC) Langerin+ DCs in CHS.13–15 Thus, it is now well recognized that LCs and (non-LC) Langerin+ DCs are crucial in both mounting and regulating immunologic processes. Despite the importance of these cells, characterization of CD11c+ Langerin+ populations in the cornea, an immune-privileged tissue, is poorly defined. It is agreed that the normal cornea is endowed with a significant population of DCs,16–22 though their frequencies and anatomic location within the cornea are a subject of debate. DCs previously described in the stroma18,20 of normal cornea include CD11c+CD11b+ DCs20 and a population of previously implicated plasmacytoid DCs.21 DCs have also been described in the epithelium of the normal cornea17–19; they are CD11c+CD11blow17,19 and are often collectively referred to as LCs. However, it has recently been shown that only a fraction of corneal epithelial DCs actually express Birbeck granules17 or Langerin.22 Furthermore, whether these are all LCs, or whether there are (non-LC) Langerin+ DCs in the cornea, is completely unknown. In the present study, we found by qRT-PCR, immunohistochemistry, and FACS analysis that both the epithelium and the stroma of the normal murine cornea have CD11c+ Langerin+ populations. Furthermore, using genetically modified mice—including muLangerin-eGFP, huLangerin-DTA, and huLangerin-Cre YFP-flox mice13—previously established to study Langerin+ CD11c+ cells in the skin, we found that the CD11c+ Langerin+ population in the corneal epithelium consists mostly of LCs, whereas those in the corneal stroma were mostly (non-LC) Langerin+ DCs. Thus, such characterizations of CD11c+ Langerin+ subsets in the cornea will permit future work in addressing their role in the immunopathogenesis of clinical conditions such as autoimmune dry eye, allergic conjunctivitis, and corneal transplant rejection.
- Published
- 2011
41. Langerhans cells are not required for epidermal V gamma 3 T cell homeostasis and function
- Author
-
Daniel H. Kaplan, Jean Plum, Bart Vandekerckhove, Tom Taghon, Björn E. Clausen, Sylvie Taveirne, Tina Van Den Broeck, Georges Leclercq, Clare L. Bennett, Veerle De Colvenaer, Els Van Ammel, Immunology, and University of Groningen
- Subjects
EXPRESSION ,skin ,T-Lymphocytes ,Cellular differentiation ,medicine.medical_treatment ,T cell ,BONE-MARROW ,Immunology ,Population ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Cell Separation ,Biology ,DENDRITIC CELLS ,DEFICIENT MICE ,GAMMA-GENES ,Mice ,Interleukin 21 ,NATURAL-KILLER-CELLS ,medicine ,Animals ,Homeostasis ,Immunology and Allergy ,Cytotoxic T cell ,education ,CONTACT HYPERSENSITIVITY ,LC deficiency ,education.field_of_study ,Epidermis (botany) ,RECEPTOR ,integumentary system ,Cell Development, Differtiation, and Trafficking ,Langerin DTR mice ,Cell Differentiation ,Receptors, Antigen, T-Cell, gamma-delta ,hemic and immune systems ,Cell Biology ,IN-VITRO ,Flow Cytometry ,Immunohistochemistry ,In vitro ,Cell biology ,Cytokine ,medicine.anatomical_structure ,Epidermal Cells ,Langerhans Cells ,Epidermis ,Langerin DTA mice - Abstract
This study tested the hypothesis that V gamma 3 TCR-bearing T cells are influenced by LCs. V gamma 3 T cells and LCs are located in the epidermis of mice. V gamma 3 T cells represent the main T cell population in the skin epithelium and play a crucial role in maintaining the skin integrity, whereas LCs are professional APCs. Although V gamma 3 T cells and LCs form an interdigitating network in the epidermis, not much is known about their reciprocal influence and/or interdependence. We used two different LC-deficient mouse models, in which LCs are constitutively or inducibly depleted, to investigate the role of LCs in maturation, homeostasis, and function of V gamma 3 T cells. We show that V gamma 3 T cell numbers are unaltered by LC deficiency, and V gamma 3 T cells isolated from LC-deficient mice are phenotypically and upon in vitro stimulation, functionally indistinguishable from V gamma 3 T cells isolated from WT mice based on their cytotoxic potential and cytokine production. Additionally, in vivo skin-wounding experiments show no major difference in response of V gamma 3 T cells to wounding in the absence or presence of LCs. These observations indicate that V gamma 3 T cells develop and function independently of LCs. J. Leukoc. Biol. 90: 61-68; 2011.
- Published
- 2011
42. Langerhans cells are not required for graft-versus-host disease
- Author
-
Jennifer M. McNiff, Catherine Matte-Martone, Hongmei Li, Hung Sheng Tan, Mark J. Shlomchik, Srividhya Venkatesan, Warren D. Shlomchik, Anthony J. Demetris, Daniel H. Kaplan, and Kody L. Johnson
- Subjects
Langerhans cell ,Langerin ,Immunology ,Fluorescent Antibody Technique ,Graft vs Host Disease ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Cell Separation ,Biochemistry ,Mice ,medicine ,Animals ,Humans ,Antigen-presenting cell ,Diphtheria toxin ,Transplantation ,biology ,integumentary system ,Cell Biology ,Hematology ,medicine.disease ,Flow Cytometry ,Graft-versus-host disease ,medicine.anatomical_structure ,surgical procedures, operative ,Langerhans Cells ,biology.protein ,Bone marrow ,Stem cell ,CD8 - Abstract
Graft-versus-host disease (GVHD) is initiated and maintained by antigen-presenting cells (APCs) that prime alloreactive donor T cells. APCs are therefore attractive targets for GVHD prevention and treatment. APCs are diverse in phenotype and function, making understanding how APC subsets contribute to GVHD necessary for the development of APC-targeted therapies. Langerhans cells (LCs) have been shown to be sufficient to initiate skin GVHD in a major histocompatibility complex–mismatched model; however, their role when other host APC subsets are intact is unknown. To address this question, we used mice genetically engineered to be deficient in LCs by virtue of expression of diphtheria toxin A under the control of a BAC (bacterial artificial chromosome) transgenic hu-man Langerin locus. Neither CD8- nor CD4-mediated GVHD was diminished in recipients lacking LCs. Similarly, CD8- and CD4-mediated GVHD, including that in the skin, was unaffected if bone marrow came from donors that could not generate LCs, even though donor LCs engrafted in control mice. Engraftment of donor LCs after irradiation in wild-type hosts required donor T cells, with immunofluorescence revealing patches of donor and residual host LCs. Surprisingly, donor LC engraftment in Langerin-diphtheria toxin A (DTA) transgenic hosts was independent of donor T cells, suggesting that a Langerin+ cell regulates repopulation of the LC compartment.
- Published
- 2010
43. Protective T cell immunity in mice following protein-TLR7/8 agonist-conjugate immunization requires aggregation, type I IFN, and multiple DC subsets
- Author
-
Daniel H. Kaplan, Mark C. Udey, Wolfgang Kastenmüller, Maria R. Becker, Björn E. Clausen, Barbara J. Flynn, Ulrike Wille-Reece, Robert A. Seder, Ross W. B. Lindsay, Ronald N. Germain, Patricia A. Darrah, Kathrin Kastenmüller, Lauren Trager, Botond Z. Igyártó, and Immunology
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,medicine.medical_treatment ,Priming (immunology) ,Receptors, Cell Surface ,Biology ,CD8-Positive T-Lymphocytes ,Proinflammatory cytokine ,Minor Histocompatibility Antigens ,Mice ,Antigen ,SDG 3 - Good Health and Well-being ,Conjugate vaccine ,Antigens, CD ,Cell Movement ,medicine ,Animals ,Lectins, C-Type ,Antigens ,Mice, Knockout ,Mice, Inbred BALB C ,Membrane Glycoproteins ,General Medicine ,TLR7 ,Dendritic Cells ,Th1 Cells ,CD11c Antigen ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Toll-Like Receptor 7 ,Toll-Like Receptor 8 ,Immunology ,Interferon Type I ,Adjuvant ,CD8 ,Research Article - Abstract
The success of a non-live vaccine requires improved formulation and adjuvant selection to generate robust T cell immunity following immunization. Here, using protein linked to a TLR7/8 agonist (conjugate vaccine), we investigated the functional properties of vaccine formulation, the cytokines, and the DC subsets required to induce protective multifunctional T cell immunity in vivo. The conjugate vaccine required aggregation of the protein to elicit potent Th1 CD4(+) and CD8(+) T cell responses. Remarkably, the conjugate vaccine, through aggregation of the protein and activation of TLR7 in vivo, led to an influx of migratory DCs to the LN and increased antigen uptake by several resident and migratory DC subsets, with the latter effect strongly influenced by vaccine-induced type I IFN. Ex vivo migratory CD8(-)DEC205(+)CD103(-)CD326(-) langerin-negative dermal DCs were as potent in cross-presenting antigen to naive CD8(+) T cells as CD11c(+)CD8(+) DCs. Moreover, these cells also influenced Th1 CD4(+) T cell priming. In summary, we propose a model in which broad-based T cell-mediated responses upon vaccination can be maximized by codelivery of aggregated protein and TLR7/8 agonist, which together promote optimal antigen acquisition and presentation by multiple DC subsets in the context of critical proinflammatory cytokines.
- Published
- 2010
44. Acute ablation of Langerhans cells enhances skin immune responses
- Author
-
Kristin A. Hogquist, Daniel H. Kaplan, Botond Z. Igyártó, Irlanda Olvera-Gomez, Aleh Bobr, and Krystal M. Haley
- Subjects
Langerin ,Heparin-binding EGF-like growth factor ,Transgene ,Immunology ,Fluorescent Antibody Technique ,Mice, Transgenic ,Cell Separation ,Dermatitis, Contact ,Article ,Mice ,Immune system ,Antigen ,In vivo ,Antigens, CD ,Immunology and Allergy ,Animals ,Humans ,Lectins, C-Type ,Gene Knock-In Techniques ,Receptor ,Skin ,Diphtheria toxin ,biology ,integumentary system ,Flow Cytometry ,Cell biology ,Mice, Inbred C57BL ,Mannose-Binding Lectins ,Langerhans Cells ,biology.protein ,Intercellular Signaling Peptides and Proteins ,Heparin-binding EGF-like Growth Factor - Abstract
Understanding the function of Langerhans cells (LCs) in vivo has been complicated by conflicting results from LC-deficient mice. Human Langerin-DTA mice constitutively lack LCs and develop exaggerated contact hypersensitivity (CHS) responses. Murine Langerin-diptheria toxin receptor (DTR) mice allow for the inducible elimination of LCs and Langerin+ dermal dendritic cells (dDCs) after administration of diphtheria toxin, which results in reduced CHS. When Langerin+ dDCs have partially repopulated the skin but LCs are still absent, CHS returns to normal. Thus, LCs appear to be suppressive in human Langerin-DTA mice and redundant in murine Langerin-DTR mice. To determine whether inducible versus constitutive LC ablation explains these results, we engineered human Langerin-DTR mice in which diphtheria toxin ablates LCs without affecting Langerin+ dDCs. The inducible ablation of LCs in human Langerin-DTR mice resulted in increased CHS. Thus, LC-mediated suppression does not require their absence during ontogeny or during the steady-state and is consistent with a model in which LCs actively suppress Ag-specific CHS responses.
- Published
- 2010
45. Langerhans cells suppress contact hypersensitivity responses via cognate CD4 interaction and langerhans cell-derived IL-10
- Author
-
Botond Z. Igyártó, Mark J. Shlomchik, Axel Roers, Daniel J. Campbell, Daniel H. Kaplan, Jan C. Dudda, Pandelakis A. Koni, Matthew C. Jenison, and Werner Müller
- Subjects
CD4-Positive T-Lymphocytes ,Langerhans cell ,T cell ,Immunology ,CD1 ,Inflammation ,Dermatitis, Contact ,T-Lymphocytes, Regulatory ,Article ,Mice ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Sensitization ,Skin ,Homeodomain Proteins ,MHC class II ,biology ,integumentary system ,Histocompatibility Antigens Class II ,Mice, Mutant Strains ,Cell biology ,Interleukin-10 ,Interleukin 10 ,medicine.anatomical_structure ,Langerhans Cells ,biology.protein ,Cytokines ,Dinitrofluorobenzene ,medicine.symptom - Abstract
Mice lacking epidermal Langerhans cells (LC) develop exaggerated contact-hypersensitivity (CHS) responses due to the absence of LC during sensitization/initiation. Examination of T cell responses reveals that the absence of LC leads to increased numbers of hapten-specific CD4 and CD8 T cells but does not alter cytokine expression or development of T regulatory cells. CHS responses and Ag-specific T cells are increased in mice in which MHC class II is ablated specifically in LC suggesting that direct cognate interaction between LC and CD4 cells is required for suppression. LC-derived IL-10 is also required for optimal inhibition of CHS. Both LC-derived IL-10-mediated suppression and full LC activation require LC expression of MHC class II. These data support a model in which cognate interaction of LC with CD4 T cells enables LC to inhibit expansion of Ag-specific responses via elaboration of IL-10.
- Published
- 2009
46. Langerhans cell deficiency impairs Ixodes scapularis suppression of Th1 responses in mice
- Author
-
Diana L. Vesely, Mark J. Shlomchik, Daniel H. Kaplan, Durland Fish, and Linda K. Bockenstedt
- Subjects
Tick infestation ,Langerhans cell ,Immunology ,Tick ,Microbiology ,Immune tolerance ,Mice ,Immune system ,Th2 Cells ,parasitic diseases ,medicine ,Immune Tolerance ,Animals ,Borrelia burgdorferi ,Saliva ,Interleukin 4 ,Mice, Knockout ,Host Response and Inflammation ,biology ,integumentary system ,Ixodes ,Feeding Behavior ,Th1 Cells ,medicine.disease ,biology.organism_classification ,bacterial infections and mycoses ,Infectious Diseases ,medicine.anatomical_structure ,Ixodes scapularis ,Langerhans Cells ,Parasitology ,Lymph Nodes ,Spleen - Abstract
Ixodes scapularis ticks transmit a number of human pathogens, including the Lyme disease spirochete Borrelia burgdorferi. I. scapularis suppresses host immunity in the skin to promote feeding and systemically skew T-helper (Th)-cell differentiation toward Th2 cells in secondary lymphoid organs. Although components of tick saliva are known to influence Th-cell polarization, the mechanism whereby tick feeding in the skin modulates regional and systemic Th-cell responses is unknown. In this study, the role of the epidermal Langerhans cell (LC) subset of skin dendritic cells in tick-mediated Th1/Th2-cell immunomodulation was assessed. Mice deficient in LCs (Langerin-DTA mice) exhibited enhanced lymph node (LN) concanavalin A (ConA)-induced Th1 responses after tick infestation in comparison to results for uninfested Langerin-DTA or wild-type (WT) mice, whereas effects on Th2-cell production of interleukin 4 were more variable. Nonetheless, the altered T-cell response did not impact tick feeding or refeeding. Gamma interferon production by ConA-stimulated LN cells of both WT and LC-deficient mice was enhanced by as much as fourfold after B. burgdorferi -infected-tick feeding, indicating that immunomodulatory effects of tick saliva were not able to attenuate the Th1 immune responses induced by this pathogen. Taken together, these findings show a requirement for LCs in the tick-mediated attenuation of Th1 responses in regional lymph nodes but not in the spleens of mice and show that the presence of a pathogen can overcome the Th1-inhibitory effects of tick feeding on the host.
- Published
- 2009
47. Langerhans cells are not required for efficient skin graft rejection
- Author
-
Daniel H. Kaplan, Martin H. Oberbarnscheidt, Daniel L. Mueller, Fadi G. Lakkis, Jagdeep S. Obhrai, Mark J. Shlomchik, Na Zhang, and Warren D. Shlomchik
- Subjects
Graft Rejection ,Male ,Pathology ,medicine.medical_specialty ,H-Y Antigen ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Dermatology ,Biology ,Biochemistry ,Article ,Mice ,Antigen ,Donor graft ,medicine ,Animals ,Hypersensitivity, Delayed ,Lectins, C-Type ,Molecular Biology ,Graft acceptance ,Sex Characteristics ,Graft rejection ,integumentary system ,Extramural ,hemic and immune systems ,Cell Biology ,Skin Transplantation ,Tumor Necrosis Factor Receptor Superfamily, Member 7 ,Mice, Inbred C57BL ,surgical procedures, operative ,Mannose-Binding Lectins ,Allograft rejection ,Langerhans Cells ,Immunology ,Antigens, Surface ,Models, Animal ,Female ,Tumor necrosis factor receptor ,Donor skin - Abstract
The mechanism of skin allograft rejection has been thought to require presentation of graft antigen by resident epidermal Langerhans cells (LCs). We have previously engineered mice that have a selective and constitutive absence of epidermal LCs. By using donor skin from these LC-deficient mice, we show that LCs are not required for rejection of major (FVB → B6) or minor (H-Y, male → female on B6 background) antigen-mismatched skin grafts. On the FVB background, where H-Y mismatched grafts are normally maintained indefinitely, grafts lacking LCs are efficiently rejected. Thus, LCs in the donor graft are required for long-term skin engraftment, which supports a regulatory role for LCs in skin graft acceptance. JID JOURNAL CLUB ARTICLE: For questions, answers, and open discussion about this article please go to http://network.nature.com/group/jidclub
- Published
- 2008
48. Identification of a novel population of Langerin+ dendritic cells
- Author
-
Laura S. Bursch, Botond Z. Igyártó, Liangchun Wang, Adrien Kissenpfennig, Kristin A. Hogquist, Daniel H. Kaplan, Bernard Malissen, Center for Immunology - University of Minnesota, University of Minnesota [Twin Cities] (UMN), University of Minnesota System-University of Minnesota System, Centre d'Immunologie de Marseille - Luminy (CIML), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), and Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
MESH: Lymph Nodes ,Mice ,0302 clinical medicine ,Immunology and Allergy ,MESH: Animals ,Skin ,0303 health sciences ,education.field_of_study ,biology ,MESH: Dendritic Cells ,integumentary system ,MESH: Bone Marrow Cells ,hemic and immune systems ,Dermis ,Articles ,Cell biology ,medicine.anatomical_structure ,Phenotype ,Antigens, Surface ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,medicine.symptom ,Langerin ,MESH: Antigens, Surface ,Population ,Immunology ,Inflammation ,chemical and pharmacologic phenomena ,Bone Marrow Cells ,MESH: Phenotype ,Models, Biological ,Article ,03 medical and health sciences ,Immune system ,Antigen ,MESH: Skin ,MESH: Mice, Inbred C57BL ,MESH: Mannose-Binding Lectins ,medicine ,Animals ,Lectins, C-Type ,education ,MESH: Mice ,030304 developmental biology ,Diphtheria toxin ,MESH: Immune System ,Epidermis (botany) ,MESH: Models, Biological ,Dendritic Cells ,MESH: Dermis ,Mice, Inbred C57BL ,Mannose-Binding Lectins ,Immune System ,biology.protein ,Lymph Nodes ,MESH: Epidermis ,Epidermis ,MESH: Lectins, C-Type ,030215 immunology - Abstract
International audience; Langerhans cells (LCs) are antigen-presenting cells that reside in the epidermis of the skin and traffic to lymph nodes (LNs). The general role of these cells in skin immune responses is not clear because distinct models of LC depletion resulted in opposite conclusions about their role in contact hypersensitivity (CHS) responses. While comparing these models, we discovered a novel population of LCs that resides in the dermis and does not represent migrating epidermal LCs, as previously thought. Unlike epidermal LCs, dermal Langerin(+) dendritic cells (DCs) were radiosensitive and displayed a distinct cell surface phenotype. Dermal Langerin(+) DCs migrate from the skin to the LNs after inflammation and in the steady state, and represent the majority of Langerin(+) DCs in skin draining LNs. Both epidermal and dermal Langerin(+) DCs were depleted by treatment with diphtheria toxin in Lang-DTREGFP knock-in mice. In contrast, transgenic hLang-DTA mice lack epidermal LCs, but have normal numbers of dermal Langerin(+) DCs. CHS responses were abrogated upon depletion of both epidermal and dermal LCs, but were unaffected in the absence of only epidermal LCs. This suggests that dermal LCs can mediate CHS and provides an explanation for previous differences observed in the two-model systems.
- Published
- 2007
- Full Text
- View/download PDF
49. Acute upregulation of an NKG2D ligand promotes rapid reorganization of a local immune compartment with pleiotropic effects on carcinogenesis
- Author
-
Adrian Hayday, Michael Girardi, Daniel H. Kaplan, William L. Schpero, Bernice Y. Kwong, Renata B. Filler, Jessica Strid, Julia M. Lewis, and Scott J. Roberts
- Subjects
Skin Neoplasms ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,Immunology ,Nkg2d ligands ,chemical and pharmacologic phenomena ,Mice, Inbred Strains ,Biology ,medicine.disease_cause ,Ligands ,Mice ,Immune system ,Downregulation and upregulation ,medicine ,Immunology and Allergy ,Animals ,Receptors, Immunologic ,Immunologic Surveillance ,Histocompatibility Antigens Class I ,hemic and immune systems ,Receptors, Antigen, T-Cell, gamma-delta ,Compartment (chemistry) ,biochemical phenomena, metabolism, and nutrition ,biological factors ,Cell biology ,Up-Regulation ,Cell Transformation, Neoplastic ,NK Cell Lectin-Like Receptor Subfamily K ,Langerhans Cells ,Receptors, Natural Killer Cell ,Epidermis ,Carcinogenesis - Abstract
The self-encoded ligands MICA (human) and Rae-1 (mouse) for the cytotoxic lymphocyte activating receptor NKG2D are highly expressed in carcinomas and inflammatory lesions and have been linked to immunosurveillance and graft rejection. However, whether NKG2D ligands have an intrinsic ability to acutely regulate tissue-associated immune compartments is not known. Here we show that epidermis-specific upregulation of Rae-1 induced rapid, coincident and reversible changes in the organization of tissue-resident V(gamma)5V(delta)1 TCRgammadelta+ intraepithelial T cells and Langerhans cells, swiftly followed by epithelial infiltration by unconventional alphabeta T cells. Whereas local V(gamma)5V(delta)1+ T cells limited carcinogenesis, Langerhans cells unexpectedly promoted it. These results provide unique insight into the early phases of tissue immunosurveillance and indicate that acute changes in NKG2D ligands may alone initiate a rapid, multifaceted immunosurveillance response in vivo.
- Published
- 2007
50. Identification of an interferon-gamma receptor alpha chain sequence required for JAK-1 binding
- Author
-
Daniel H. Kaplan, Andrew C. Greenlund, J. William Tanner, Robert D. Schreiber, and Andrey S. Shaw
- Subjects
Proline ,Molecular Sequence Data ,Biology ,Biochemistry ,Tropomyosin receptor kinase C ,Mice ,Antigens, CD ,Tumor Cells, Cultured ,Animals ,Humans ,5-HT5A receptor ,Amino Acid Sequence ,Interleukin-7 receptor ,Molecular Biology ,Receptors, Interferon ,Alanine ,Janus kinase 1 ,Cell Biology ,Janus Kinase 1 ,Protein-Tyrosine Kinases ,Molecular biology ,Interleukin-21 receptor ,Signal transduction ,Tyrosine kinase ,Protein Binding - Abstract
We have shown previously that a four-amino acid block residing at positions 266-269 (LPKS) in the intracellular domain of the human interferon-gamma (IFN-gamma) receptor alpha chain is critical for IFN-gamma-dependent tyrosine kinase activation and biologic response induction. Herein we show that this sequence is required for the constitutive attachment of the tyrosine kinase JAK-1. Using a vaccinia expression system, a receptor alpha chain-specific monoclonal antibody coprecipitated JAK-1 from cells coexpressing JAK-1 and either (a) wild type IFN-gamma receptor alpha chain, (b) a receptor alpha chain truncation mutant containing only the first 59 intracellular domain amino acids, or (c) a receptor mutant containing alanine substitutions for the functionally irrelevant residues 272-275. In contrast, JAK-1 was not coprecipitated when coexpressed with a receptor alpha chain mutant containing alanine substitutions for the functionally critical residues 266-269 (LPKS). Mutagenesis of the LPKS sequence revealed that Pro-267 is the only residue obligatorily required for receptor function. In addition, Pro-267 is required for JAK-1 binding. These results thus identify a site in the IFN-gamma receptor alpha chain required for constitutive JAK-1 association and establish that this association is critical for IFN-gamma signal transduction.
- Published
- 1996
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.