Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Invest Dermatol ; 142(12): 3294-3303, 2022 12.
Article in English | MEDLINE | ID: mdl-35787400

ABSTRACT

Despite the central role of IFN-γ in vitiligo pathogenesis, systemic IFN-γ neutralization is an impractical treatment option owing to strong immunosuppression. However, most patients with vitiligo present with <20% affected body surface area, which provides an opportunity for localized treatments that avoid systemic side effects. After identifying keratinocytes as key cells that amplify IFN-γ signaling during vitiligo, we hypothesized that tethering an IFN-γ‒neutralizing antibody to keratinocytes would limit anti‒IFN-γ effects on the treated skin for the localized treatment. To that end, we developed a bispecific antibody capable of blocking IFN-γ signaling while binding to desmoglein expressed by keratinocytes. We characterized the effect of the bispecific antibody in vitro, ex vivo, and in a mouse model of vitiligo. Single-photon emission computed tomography/computed tomography biodistribution and serum assays after local footpad injection revealed that the bispecific antibody had improved skin retention, faster elimination from the blood, and less systemic IFN-γ inhibition than the nontethered version. Furthermore, the bispecific antibody conferred localized protection almost exclusively to the treated footpad during vitiligo, which was not possible by local injection of the nontethered anti‒IFN-γ antibody. Thus, keratinocyte tethering proved effective while significantly diminishing the off-tissue effects of IFN-γ blockade, offering a safer treatment strategy for localized skin diseases, including vitiligo.


Subject(s)
Biological Products , Vitiligo , Mice , Animals , Vitiligo/drug therapy , Tissue Distribution , Keratinocytes/metabolism , Skin/pathology , Biological Products/therapeutic use
2.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34325468

ABSTRACT

The STING and absent in melanoma 2 (AIM2) pathways are activated by the presence of cytosolic DNA, and STING agonists enhance immunotherapeutic responses. Here, we show that dendritic cell (DC) expression of AIM2 within human melanoma correlates with poor prognosis and, in contrast to STING, AIM2 exerts an immunosuppressive effect within the melanoma microenvironment. Vaccination with AIM2-deficient DCs improves the efficacy of both adoptive T cell therapy and anti-PD-1 immunotherapy for "cold tumors," which exhibit poor therapeutic responses. This effect did not depend on prolonged survival of vaccinated DCs, but on tumor-derived DNA that activates STING-dependent type I IFN secretion and subsequent production of CXCL10 to recruit CD8+ T cells. Additionally, loss of AIM2-dependent IL-1ß and IL-18 processing enhanced the treatment response further by limiting the recruitment of regulatory T cells. Finally, AIM2 siRNA-treated mouse DCs in vivo and human DCs in vitro enhanced similar anti-tumor immune responses. Thus, targeting AIM2 in tumor-infiltrating DCs is a promising new treatment strategy for melanoma.


Subject(s)
Cancer Vaccines/immunology , DNA-Binding Proteins/immunology , Melanoma, Experimental/immunology , Melanoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Middle Aged , Tumor Microenvironment/immunology , Young Adult
4.
Pigment Cell Melanoma Res ; 34(4): 683-695, 2021 07.
Article in English | MEDLINE | ID: mdl-33040466

ABSTRACT

Vitiligo is an autoimmune skin disease in which epidermal melanocytes are targeted for destruction by CD8+ T cells specific for melanocyte/melanoma-shared antigens. IFNγ is the central cytokine driving disease, but the role of type I IFN in vitiligo remains unclear. We investigated the functional role of type I IFN during vitiligo progression using two different mouse models: one induced with a vaccinia virus (VV) vaccine and one induced with dendritic cells to prime autoimmune T cells. Induction of vitiligo by VV in IFNaR-deficient mice led to the development of severe vitiligo compared with wild-type (WT) mice and was characterized by a significantly enhanced effector CD8+ T-cell response. Severe vitiligo in this model was a result of VV persistence, because exacerbation of disease in IFNaR-deficient mice was not observed when antigen-pulsed dendritic cells were used to induce vitiligo instead of virus. Treatment of B16F10 melanoma-inoculated mice with VV vaccine therapy also induced a significantly enhanced anti-tumor response in IFNaR-deficient mice compared with WT. These results not only help define the pathways responsible for vitiligo progression but also suggest that blockade of type I IFNs following administration of a VV vaccine may provide increased immunogenicity and efficacy for melanoma immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Genetic Vectors/metabolism , Immunotherapy , Interferon Type I/metabolism , Melanoma, Experimental/therapy , Signal Transduction , Vitiligo/therapy , Animals , B7-H1 Antigen/metabolism , Chemokine CXCL10/metabolism , Chemokine CXCL9/metabolism , Female , Hyaluronan Receptors/metabolism , Ligands , Male , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/metabolism , Receptors, CXCR3/metabolism , Vaccinia virus/genetics , Vitiligo/immunology , gp100 Melanoma Antigen/metabolism
5.
J Immunol ; 203(1): 11-19, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31209143

ABSTRACT

Vitiligo is an autoimmune skin disease mediated by autoreactive CD8+ T cells that destroy the pigment-producing cells of the epidermis, melanocytes, leading to areas of depigmentation. Patients with vitiligo require lifelong treatment to regain and maintain their pigment. Clinical observations uncovered the importance of autoimmune memory in vitiligo because cessation of treatment frequently led to relapse of disease at the site of previous lesions. A subset of memory T cells known as CD8+ resident memory T cells (TRM) are long-lived, nonmigratory memory cells that persist in most nonlymphoid tissues, including the skin. Recent reports describe the presence of CD8+ TRM in lesional vitiligo patient skin and suggest their role as active players in disease maintenance. In this review, we will discuss the role of skin CD8+ TRM in maintaining disease in vitiligo and the opportunity to target this population to induce a long-lasting reversal of disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/trends , Melanocytes/physiology , Skin/immunology , Vitiligo/immunology , Animals , Autoimmunity , Humans , Immunologic Memory , Skin Pigmentation
6.
Curr Protoc Immunol ; 124(1): e63, 2019 02.
Article in English | MEDLINE | ID: mdl-30253067

ABSTRACT

Vitiligo is an autoimmune skin disease in which the pigment-producing melanocytes are destroyed by autoreactive CD8+ T cells. As a result, patients develop disfiguring white spots on the skin. This article discusses the first mouse model of vitiligo that develops epidermal depigmentation, similar to disease in human patients. To achieve epidermal depigmentation, mice are genetically engineered to retain melanocytes in the skin epidermis. Induction of disease occurs by adoptive transfer of melanocyte-specific CD8+ T cells into recipient mice and the subsequent activation of these T cells using a viral vector. Depigmentation of the epidermis occurs within 5 to 7 weeks in a patchy pattern similar to patients with vitiligo. This article describes the methods of vitiligo induction, quantification of lesion progression and regression, processing of the skin for detailed analysis, and how to use this model to inform clinical studies. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Disease Models, Animal , Vitiligo/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Mice , Mice, Transgenic , Stem Cell Factor/immunology
7.
Sci Transl Med ; 10(450)2018 07 18.
Article in English | MEDLINE | ID: mdl-30021889

ABSTRACT

Vitiligo is an autoimmune disease of the skin mediated by CD8+ T cells that kill melanocytes and create white spots. Skin lesions in vitiligo frequently return after discontinuing conventional treatments, supporting the hypothesis that autoimmune memory is formed at these locations. We found that lesional T cells in mice and humans with vitiligo display a resident memory (TRM) phenotype, similar to those that provide rapid, localized protection against reinfection from skin and mucosal-tropic viruses. Interleukin-15 (IL-15)-deficient mice reportedly have impaired TRM formation, and IL-15 promotes TRM function ex vivo. We found that both human and mouse TRM express the CD122 subunit of the IL-15 receptor and that keratinocytes up-regulate CD215, the subunit required to display the cytokine on their surface to promote activation of T cells. Targeting IL-15 signaling with an anti-CD122 antibody reverses disease in mice with established vitiligo. Short-term treatment with anti-CD122 inhibits TRM production of interferon-γ (IFNγ), and long-term treatment depletes TRM from skin lesions. Short-term treatment with anti-CD122 can provide durable repigmentation when administered either systemically or locally in the skin. On the basis of these data, we propose that targeting CD122 may be a highly effective and even durable treatment strategy for vitiligo and other tissue-specific autoimmune diseases involving TRM.


Subject(s)
Antibodies, Blocking/therapeutic use , Interleukin-15/metabolism , Signal Transduction , Vitiligo/drug therapy , Vitiligo/immunology , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/pharmacology , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Epidermis/immunology , Humans , Immunologic Memory , Interferon-gamma/metabolism , Melanocytes/metabolism , Mice, Inbred C57BL , Phenotype , Receptors, Interleukin-15/metabolism , Vitiligo/pathology
8.
PLoS One ; 8(1): e53426, 2013.
Article in English | MEDLINE | ID: mdl-23301070

ABSTRACT

"Hairpatches" (Hpt) is a naturally occurring, autosomal semi-dominant mouse mutation. Hpt/Hpt homozygotes die in utero, while Hpt/+ heterozygotes exhibit progressive renal failure accompanied by patchy alopecia. This mutation is a model for the rare human disorder "glomerulonephritis with sparse hair and telangiectases" (OMIM 137940). Fine mapping localized the Hpt locus to a 6.7 Mb region of Chromosome 4 containing 62 known genes. Quantitative real time PCR revealed differential expression for only one gene in the interval, T-cell acute lymphocytic leukemia 1 (Tal1), which was highly upregulated in the kidney and skin of Hpt/+ mice. Southern blot analysis of Hpt mutant DNA indicated a new EcoRI site in the Tal1 gene. High throughput sequencing identified an endogenous retroviral class II intracisternal A particle insertion in Tal1 intron 4. Our data suggests that the IAP insertion in Tal1 underlies the histopathological changes in the kidney by three weeks of age, and that glomerulosclerosis is a consequence of an initial developmental defect, progressing in severity over time. The Hairpatches mouse model allows an investigation into the effects of Tal1, a transcription factor characterized by complex regulation patterns, and its effects on renal disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , DNA Transposable Elements/genetics , Kidney Diseases/metabolism , Mutation , Proto-Oncogene Proteins/genetics , Alopecia/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromosome Mapping , Crosses, Genetic , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Linkage , Kidney Diseases/virology , Mice , Proto-Oncogene Proteins/metabolism , Real-Time Polymerase Chain Reaction , T-Cell Acute Lymphocytic Leukemia Protein 1 , Time Factors , Transcription Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...