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.
JCI Insight ; 3(19)2018 10 04.
Article in English | MEDLINE | ID: mdl-30282836

ABSTRACT

Alopecia areata (AA) is an autoimmune disease in which cytotoxic T cells specifically target growing hair follicles. We used high-throughput TCR sequencing in the C3H/HeJ mouse model of AA and in human AA patients to gain insight into pathogenic T cell populations and their dynamics, which revealed clonal CD8+ T cell expansions in lesional skin. In the C3H/HeJ model, we observed interindividual sharing of TCRß chain protein sequences, which strongly supports a model of antigenic drive in AA. The overlap between the lesional TCR repertoire and a population of CD8+NKG2D+ T cells in skin-draining lymph nodes identified this subset as pathogenic effectors. In AA patients, treatment with the oral JAK inhibitor tofacitinib resulted in a decrease in clonally expanded CD8+ T cells in the scalp but also revealed that many expanded lesional T cell clones do not completely disappear from either skin or blood during treatment with tofacitinib, which may explain in part the relapse of disease after stopping treatment.


Subject(s)
Alopecia Areata/immunology , Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/genetics , Adolescent , Adult , Alopecia Areata/drug therapy , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Follow-Up Studies , Hair Follicle/cytology , Hair Follicle/immunology , High-Throughput Nucleotide Sequencing , Humans , Lymph Nodes/cytology , Male , Mice , Mice, Inbred C3H , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Pilot Projects , Piperidines/administration & dosage , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Scalp , Treatment Outcome , Young Adult
2.
J Immunol ; 197(4): 1089-99, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27412416

ABSTRACT

Alopecia areata (AA) is an autoimmune disease of the hair follicle that results in hair loss of varying severity. Recently, we showed that IFN-γ-producing NKG2D(+)CD8(+) T cells actively infiltrate the hair follicle and are responsible for its destruction in C3H/HeJ AA mice. Our transcriptional profiling of human and mouse alopecic skin showed that the IFN pathway is the dominant signaling pathway involved in AA. We showed that IFN-inducible chemokines (CXCL9/10/11) are markedly upregulated in the skin of AA lesions, and further, that the IFN-inducible chemokine receptor, CXCR3, is upregulated on alopecic effector T cells. To demonstrate whether CXCL9/10/11 chemokines were required for development of AA, we treated mice with blocking Abs to CXCR3, which prevented the development of AA in the graft model, inhibiting the accumulation of NKG2D(+)CD8(+) T cells in the skin and cutaneous lymph nodes. These data demonstrate proof of concept that interfering with the Tc1 response in AA via blockade of IFN-inducible chemokines can prevent the onset of AA. CXCR3 blockade could be approached clinically in human AA with either biologic or small-molecule inhibition, the latter being particularly intriguing as a topical therapeutic.


Subject(s)
Alopecia Areata/immunology , Receptors, CXCR3/antagonists & inhibitors , T-Lymphocytes/immunology , Animals , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Mice, Inbred C3H , Polymerase Chain Reaction , Receptors, CXCR3/biosynthesis , Skin/immunology
3.
EBioMedicine ; 2(4): 351-5, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26137574

ABSTRACT

BACKGROUND: Alopecia areata (AA) is an autoimmune disease resulting in hair loss with devastating psychosocial consequences. Despite its high prevalence, there are no FDA-approved treatments for AA. Prior studies have identified a prominent interferon signature in AA, which signals through JAK molecules. METHODS: A patient with AA was enrolled in a clinical trial to examine the efficacy of baricitinib, a JAK1/2 inhibitor, to treat concomitant CANDLE syndrome. In vivo, preclinical studies were conducted using the C3H/HeJ AA mouse model to assess the mechanism of clinical improvement by baricitinib. FINDINGS: The patient exhibited a striking improvement of his AA on baricitinib over several months. In vivo studies using the C3H/HeJ mouse model demonstrated a strong correlation between resolution of the interferon signature and clinical improvement during baricitinib treatment. INTERPRETATION: Baricitinib may be an effective treatment for AA and warrants further investigation in clinical trials.


Subject(s)
Alopecia Areata/drug therapy , Alopecia Areata/enzymology , Azetidines/therapeutic use , Janus Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Sulfonamides/therapeutic use , Animals , Disease Models, Animal , Interferons/metabolism , Male , Mice, Inbred C3H , Purines , Pyrazoles
4.
Nat Med ; 20(9): 1043-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25129481

ABSTRACT

Alopecia areata (AA) is a common autoimmune disease resulting from damage of the hair follicle by T cells. The immune pathways required for autoreactive T cell activation in AA are not defined limiting clinical development of rational targeted therapies. Genome-wide association studies (GWAS) implicated ligands for the NKG2D receptor (product of the KLRK1 gene) in disease pathogenesis. Here, we show that cytotoxic CD8(+)NKG2D(+) T cells are both necessary and sufficient for the induction of AA in mouse models of disease. Global transcriptional profiling of mouse and human AA skin revealed gene expression signatures indicative of cytotoxic T cell infiltration, an interferon-γ (IFN-γ) response and upregulation of several γ-chain (γc) cytokines known to promote the activation and survival of IFN-γ-producing CD8(+)NKG2D(+) effector T cells. Therapeutically, antibody-mediated blockade of IFN-γ, interleukin-2 (IL-2) or interleukin-15 receptor ß (IL-15Rß) prevented disease development, reducing the accumulation of CD8(+)NKG2D(+) T cells in the skin and the dermal IFN response in a mouse model of AA. Systemically administered pharmacological inhibitors of Janus kinase (JAK) family protein tyrosine kinases, downstream effectors of the IFN-γ and γc cytokine receptors, eliminated the IFN signature and prevented the development of AA, while topical administration promoted hair regrowth and reversed established disease. Notably, three patients treated with oral ruxolitinib, an inhibitor of JAK1 and JAK2, achieved near-complete hair regrowth within 5 months of treatment, suggesting the potential clinical utility of JAK inhibition in human AA.


Subject(s)
Alopecia Areata/immunology , Janus Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Alopecia Areata/drug therapy , Animals , Gene Expression Profiling , Humans , Mice , Mice, Inbred C3H , Protein Kinase Inhibitors/pharmacology , Skin/metabolism
5.
Mamm Genome ; 24(1-2): 30-43, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23096997

ABSTRACT

The chromosome 21 gene RCAN1, encoding a modulator of the calcineurin (CaN) phosphatase, is a candidate gene for contributing to cognitive disability in people with Down syndrome (DS; trisomy 21). To develop a physiologically relevant model for studying the biochemistry of RCAN1 and its contribution to DS, we generated bacterial artificial chromosome-transgenic (BAC-Tg) mouse lines containing the human RCAN1 gene with a C-terminal HA-FLAG epitope tag incorporated by recombineering. The BAC-Tg was expressed at levels only moderately higher than the native Rcan1 gene: approximately 1.5-fold in RCAN1 (BAC-Tg1) and twofold in RCAN1 (BAC-Tg2). Affinity purification of the RCAN1 protein complex from brains of these mice revealed a core complex of RCAN1 with CaN, glycogen synthase kinase 3-beta (Gsk3b), and calmodulin, with substoichiometric components, including LOC73419. The BAC-Tg mice are fully viable, but long-term synaptic potentiation is impaired in proportion to BAC-Tg dosage in hippocampal brain slices from these mice. RCAN1 can act as a tumor suppressor in some systems, but we found that the RCAN1 BAC-Tg did not reduce mammary cancer growth when present at a low copy number in Tp53;WAP-Cre mice. This work establishes a useful mouse model for investigating the biochemistry and dose-dependent functions of the RCAN1 protein in vivo.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , Mice, Transgenic , Muscle Proteins/genetics , Animals , Brain/metabolism , Calcineurin/genetics , Calcineurin/metabolism , Cells, Cultured , Chromosomes/genetics , DNA-Binding Proteins , Disease Models, Animal , Down Syndrome/genetics , Female , Gene Dosage , Genetic Loci , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/metabolism , Hippocampus/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Kaplan-Meier Estimate , Long-Term Potentiation/genetics , Male , Mice , Muscle Proteins/metabolism , Neurons/cytology , Neurons/metabolism
6.
Mamm Genome ; 18(2): 113-22, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17334655

ABSTRACT

We created transgenic mice with a bacterial artificial chromosome (BAC) containing the human COL6A1 gene. In high-copy and low-copy transgenic lines, we found correct temporal and spatial expression of COL6A1 mRNA, paralleling the expression of the murine Col6a1 gene in a panel of nine adult and four fetal organs. The only exception was the fetal lung, in which the transgene was expressed poorly compared with the endogenous gene. Expression of COL6A1 mRNA from the transgene was copy number-dependent, and the increased gene dosage correlated with increased production of collagen VI alpha 1 in skin and heart, as indicated by Western blotting and immunohistochemistry. COL6A1 maps to Chromosome 21 and this gene has been a candidate for contributing to cardiac defects and skin abnormalities in Down syndrome. The low-copy and high-copy COL6A1 transgenics were born and survived in normal Mendelian proportions, without cardiac malformations or altered skin histology. These data indicate that the major promoter and enhancer sequences regulating COL6A1 expression are present in this 167-kb BAC clone. The lack of a strong cardiac or skin phenotype in the COL6A1 BAC-transgenic mice suggests that the increased expression of this gene does not, by itself, account for these phenotypes in Down syndrome.


Subject(s)
Chromosomes, Human, Pair 21 , Collagen Type VI/genetics , Animals , Chromosome Mapping , Chromosomes, Artificial, Bacterial , DNA Probes , Heart/physiology , Humans , Introns , Mice , Mice, Transgenic , Serum Albumin/genetics , Skin Physiological Phenomena
7.
Mol Cell Biol ; 24(10): 4581-92, 2004 May.
Article in English | MEDLINE | ID: mdl-15121874

ABSTRACT

Engagement of the T-cell receptor (TCR) results in the activation of a multitude of signaling events that regulate the function of T lymphocytes. These signaling events are in turn modulated by adapter molecules, which control the final functional output through the formation of multiprotein complexes. In this report, we identified the adapter molecule Sin as a new regulator of T-cell activation. We found that the expression of Sin in transgenic T lymphocytes and Jurkat T cells inhibited interleukin-2 expression and T-cell proliferation. This inhibitory effect was specific and was due to defective phospholipase C-gamma (PLC-gamma) phosphorylation and activation. In contrast to other adapters that become phosphorylated upon TCR stimulation, Sin was constitutively phosphorylated in resting cells by the Src kinase Fyn and bound to signaling intermediates, including PLC-gamma. In stimulated cells, Sin was transiently dephosphorylated, which coincided with transient dissociation of Fyn and PLC-gamma. Downregulation of Sin expression using Sin-specific short interfering RNA oligonucleotides inhibited transcriptional activation in response to TCR stimulation. Our results suggest that endogenous Sin influences T-lymphocyte signaling by sequestering signaling substrates and regulating their availability and/or activity in resting cells, while Sin is required for targeting these intermediates to the TCR for fast signal transmission during stimulation.


Subject(s)
Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Base Sequence , DNA, Complementary/genetics , Gene Expression , Humans , Jurkat Cells , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phospholipase C gamma , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoblastoma-Like Protein p130 , Signal Transduction , Transcriptional Activation , Type C Phospholipases/antagonists & inhibitors
8.
Immunol Rev ; 192: 181-95, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12670404

ABSTRACT

Stimulation of T cells through their antigen receptor induces a multitude of signaling networks that regulate T cell activation in the form of cytokine production and T cell proliferation. Multiple signal integration sites exist along these pathways in the form of multiprotein signaling complexes, the formation of which is facilitated by adapter and scaffold molecules. In recent years a number of adapter and scaffold molecules have been described in T cells and shown to play an integral part in T cell function. Among these molecules are proteins that function as positive or negative regulators of T cell activation downstream of the activated T cell receptor (TCR). Here, we discuss the role of a small family of multiadapter proteins on T cell activation, the p130Cas family, with emphasis on one of its members, Sin (Src-interacting protein). Our results suggest that Sin inhibits thymocyte development and T cell activation and is a novel negative regulator of T lymphocyte function.


Subject(s)
Phosphoproteins/physiology , Proteins , T-Lymphocytes/immunology , Animals , Cellular Apoptosis Susceptibility Protein/metabolism , Crk-Associated Substrate Protein , Lymphocyte Activation , Mice , Models, Genetic , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , Retinoblastoma-Like Protein p130 , T-Lymphocytes/enzymology , src Homology Domains , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL