Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Type of study
Publication year range
1.
Cancer Immunol Res ; 6(8): 900-909, 2018 08.
Article in English | MEDLINE | ID: mdl-29895574

ABSTRACT

Cutaneous T-cell lymphoma (CTCL) develops from clonally expanded CD4+ T cells in a background of chronic inflammation. Although dendritic cells (DCs) stimulate T cells and are present in skin, cutaneous T cells in CTCL do not respond with effective antitumor immunity. We evaluated primary T-cell and DC émigrés from epidermal and dermal explant cultures of skin biopsies from CTCL patients (n = 37) and healthy donors (n = 5). Compared with healthy skin, CD4+ CTCL populations contained more T cells expressing PD-1, CTLA-4, and LAG-3. CD8+ CTCL populations contained more T cells expressing CTLA-4 and LAG-3. CTCL populations also contained more T cells expressing the inducible T-cell costimulator (ICOS), a marker of T-cell activation. DC émigrés from healthy or CTCL skin biopsies expressed PD-L1, indicating that maturation during migration resulted in PD-L1 expression irrespective of disease. Most T cells did not express PD-L1. Using skin samples from 49 additional CTCL patients for an unsupervised analysis of genome-wide mRNA expression profiles corroborated that advanced T3/T4-stage samples expressed more checkpoint inhibition mRNA compared with T1/T2 stage patients or healthy controls. Exhaustion of activated T cells is therefore a hallmark of both CD4+ and CD8+ T cells isolated from the lesional skin of patients with CTCL, with increasing expression as the disease progresses. These results justify identification of antigens driving T-cell exhaustion and the evaluation of immune checkpoint inhibition to reverse T-cell exhaustion earlier in the treatment of CTCL. Cancer Immunol Res; 6(8); 900-9. ©2018 AACR.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphoma, T-Cell, Cutaneous/immunology , Skin Neoplasms/immunology , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Epidermis/immunology , Female , Gene Expression Profiling/methods , Genes, cdc/genetics , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Lymphoma, T-Cell, Cutaneous/genetics , Lymphoma, T-Cell, Cutaneous/pathology , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Cells, Cultured , Young Adult
2.
Cancer Immunol Res ; 5(1): 52-60, 2017 01.
Article in English | MEDLINE | ID: mdl-27923824

ABSTRACT

Janus kinase (JAK) inhibitors have achieved positive responses in myeloproliferative neoplasms, but at the expense of decreased natural killer (NK) cell numbers and compromised function. Selective JAK2 inhibition may also have a role in preventing and treating graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Although JAK inhibitors can impair monocyte-derived dendritic cell (moDC) activation and function and suppress effector T-cell responses, the effects on NK cells and the relevant mechanisms remain undefined. Using common γc cytokines and distinct human dendritic cell (DC) subtypes, we compared the effects of a JAK2-specific (TG101348) with a less selective JAK1/2 (ruxolitinib) inhibitor on NK-cell activation and function. Ruxolitinib treatment completely blocked IL2, IL15, and DC-mediated STAT5 phosphorylation, along with the capacity of NK cells to secrete IFNγ or lyse NK cell-sensitive targets. Only NK-cell proliferation stimulated by moDCs resisted ruxolitinib treatment. In contrast, TG101348 treatment of stimulated NK cells resulted in far less functional compromise. TG101348 completely inhibited only soluble IL15-mediated STAT5 phosphorylation, which Langerhans-type DCs (LCs), presenting membrane-bound IL15 in trans, could salvage. These results demonstrate that ruxolitinib's nonselective inhibition of JAK1/2 results in profound NK-cell dysfunction by blocking downstream pSTAT5, hence providing a persuasive rationale for the development of selective JAK2 inhibitors for immunotherapeutic applications. Cancer Immunol Res; 5(1); 52-60. ©2016 AACR.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Protein Kinase Inhibitors/pharmacology , Cell Line , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Humans , Immunophenotyping , Interferon-gamma/metabolism , Lymphocyte Count , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Phosphorylation , Receptors, Natural Killer Cell/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects
3.
Cancer Immunol Res ; 4(1): 61-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26464015

ABSTRACT

Multiple myeloma is the most common indication for high-dose chemotherapy and autologous stem cell transplantation (ASCT), and lenalidomide maintenance after transplant is now standard. Although lenalidomide doubles progression-free survival, almost all patients eventually relapse. Posttransplant immunotherapy to improve outcomes after ASCT therefore has great merit but first requires delineation of the dynamics of immune reconstitution. We evaluated lymphocyte composition and function after ASCT to guide optimal timing of immunotherapy and to identify potential markers of relapse. Regulatory T cells (Treg) decline as CD8(+) T cells expand during early lymphocyte recovery after ASCT, markedly reducing the Treg:CD8(+) effector T-cell ratio. These CD8(+) T cells can respond to autologous dendritic cells presenting tumor antigen in vitro as early as day +12 after transplant, becoming antigen-specific cytolytic T-lymphocyte effectors and thereby demonstrating preservation of cellular reactivity. CD4(+) and CD8(+) T cells express the negative regulatory molecules, CTLA-4, PD-1, LAG-3, and TIM-3, before and after ASCT. A subpopulation of exhausted/senescent CD8(+) T cells, however, downregulates CD28 and upregulates CD57 and PD-1, characterizing immune impairment and relapse after ASCT. Relapsing patients have higher numbers of these cells at +3 months after transplant, but before detection of clinical disease, indicating their applicability in identifying patients at higher risk of relapse. PD-1 blockade also revives the proliferation and cytokine secretion of the hyporesponsive, exhausted/senescent CD8(+) T cells in vitro. Collectively, these results identify T-cell exhaustion/senescence as a distinguishing feature of relapse and support early introduction of immunotherapy to stimulate antitumor immunity after ASCT.


Subject(s)
Multiple Myeloma/immunology , Neoplasm Recurrence, Local/immunology , Stem Cell Transplantation , Adult , Aged , Autografts , CD4-CD8 Ratio , Female , Humans , Immunotherapy , Male , Middle Aged , Multiple Myeloma/therapy , Neoplasm Recurrence, Local/prevention & control , T-Lymphocytes, Regulatory/immunology
4.
Cancer Immunol Res ; 2(10): 1000-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25023628

ABSTRACT

A functionally responsive natural killer (NK)-cell repertoire requires the acquisition of inhibitory NKG2A and killer immunoglobulin-like receptors (KIR) through pathways that remain undefined. Functional donor NK cells expressing KIRs for non-self class I MHC ligands contribute to a positive outcome after allogeneic hematopoietic stem cell transplantation (alloHSCT) by targeting HLA-matched recipient leukemic cells. Insofar as circulating donor conventional dendritic cells (DC) reconstitute with comparable kinetics with donor NK cells after alloHSCT, we used hyporesponsive KIRnegNKG2Aneg precursor cells to evaluate how specific DC subtypes generate a functionally active NK-cell repertoire. Both monocyte-derived DCs (moDC) and Langerhans-type DCs (LC) induce KIRnegNKG2Aneg precursor cells to express the inhibitory receptors NKG2A and KIR, without requiring cell proliferation. Poly(I:C)-matured moDCs significantly augmented the expression of NKG2A, but not KIR, in an IL12p70-dependent manner. Although all DC-stimulated KIRnegNKG2Aneg cells were able to acquire cytolytic activity against class I MHC-negative targets, the ability to secrete IFNγ was restricted to cells that were stimulated by IL12p70-producing, poly(I:C)-matured moDCs. This critical ability of poly(I:C)-matured moDCs to provide IL12p70 to developing KIRnegNKG2Aneg precursors results in a dom4inant, multifunctional, NKG2Apos NK-cell population that is capable of both cytolysis and IFNγ production. Poly(I:C)-matured moDCs are, therefore, the most effective conventional DC subtype for generating a functionally competent NK-cell repertoire by an IL12p70-dependent mechanism.


Subject(s)
Dendritic Cells/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Cell Differentiation/immunology , Coculture Techniques , Cytokines/biosynthesis , Humans , Immune Tolerance/immunology , Immunocompetence/immunology , Immunophenotyping , Interferon-gamma/biosynthesis , Poly I-C/immunology , Receptors, KIR/metabolism
5.
PLoS One ; 8(9): e76292, 2013.
Article in English | MEDLINE | ID: mdl-24086722

ABSTRACT

NK cells are large granular lymphocytes that form a critical component of the innate immune system, whose functions include the killing of cells expressing stress-induced molecules. It is increasingly accepted that despite being considered prototypical effector cells, NK cells require signals to reach their full cytotoxic potential. We previously showed that IL-15 is capable of arming CD8 effector T cells to kill independently of their TCR via NKG2D in a cPLA2-dependent process. As NK cells also express NKG2D, we wanted to investigate whether this pathway functioned in an analogous manner and if resting NK cells could be primed to the effector phase by IL-15. Furthermore, to establish relevance to human disease we studied a possible role for this pathway in the pathogenesis of psoriatic arthritis, since there are aspects of this disease that suggest a potential effector role for the innate immune system. We found that PsA patients had upregulated IL-15 and MIC in their affected synovial tissues, and that this unique inflammatory environment enabled NK cell activation and killing via NKG2D and cPLA2. Moreover, we were able to reproduce the phenotype of joint NK cells from blood NK cells by incubating them with IL-15. Altogether, these findings suggest a destructive role for NK cells when activated by environmental stress signals during the pathogenesis of PsA and demonstrate that IL-15 is capable of priming resting NK cells in tissues to the effector phase.


Subject(s)
Arthritis, Psoriatic/immunology , Gene Expression Regulation/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phospholipases A2, Cytosolic/metabolism , Signal Transduction/immunology , Arthritis, Psoriatic/metabolism , Cytokines/genetics , Cytokines/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Interleukin-15/metabolism , Killer Cells, Natural/metabolism , RNA, Small Interfering/genetics , Transfection
6.
Blood ; 118(19): 5330-9, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21917753

ABSTRACT

Janus kinase-2 (JAK2) conveys receptor-binding signals by several inflammatory cytokines, including IL-6, via phosphorylation of signal transducer and activator of transcription 3 (STAT3). We demonstrate that selective JAK2 inhibition by TG101348 during initial encounters between human T cells and allogeneic monocyte-derived dendritic cells induces durable, profound, and specific T-cell tolerance upon reexposure to the same alloantigens. Subsequent responses by nonalloreactive T cells to stimulation de novo by a pathogenic nominal antigen remain intact. TG101348 also suppresses primed T-cell responses when present only during alloantigen restimulation. TG101348 ablates IL-6/JAK2-mediated phosphorylation of STAT3, but has no off-target effects on IL-2 or IL-15/JAK3/pSTAT5-dependent signaling, which sustain the responses of regulatory T cells (Tregs) and other effector T cells. JAK2 inhibition preserves Treg numbers and thereby enhances the ratio of CD4(+) Tregs to CD8(+)CD25(+) effector T cells in favor of Tregs. JAK2 inhibition also reduces the production of IL-6 and TNF-α in allogeneic MLRs, impairing the activation of central and effector memory T cells as well as the expansion of responder Th1 and Th17 cells. While we have reported the limitations of isolated IL-6R-α inhibition on dendritic cell-stimulated alloreactivity, we demonstrate here that JAK2 represents a relevant biologic target for controlling GVHD or allograft rejection without broader immune impairment.


Subject(s)
Dendritic Cells/enzymology , Dendritic Cells/immunology , Immune Tolerance , Janus Kinase 2/antagonists & inhibitors , T-Lymphocytes/immunology , Cell Differentiation , Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/drug effects , Humans , Immune Tolerance/drug effects , Immunologic Memory , In Vitro Techniques , Isoantigens , Janus Kinase 2/immunology , Lymphocyte Culture Test, Mixed , Pyrrolidines/pharmacology , Signal Transduction/immunology , Sulfonamides/pharmacology , T-Lymphocytes, Regulatory/immunology
7.
Nature ; 456(7221): 534-8, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-19037317

ABSTRACT

Major histocompatibility complex (MHC) class II alleles HLA-DQ8 and the mouse homologue I-A(g7) lacking a canonical aspartic acid residue at position beta57 are associated with coeliac disease and type I diabetes. However, the role of this single polymorphism in disease initiation and progression remains poorly understood. The lack of Asp 57 creates a positively charged P9 pocket, which confers a preference for negatively charged peptides. Gluten lacks such peptides, but tissue transglutaminase (TG2) introduces negatively charged residues at defined positions into gluten T-cell epitopes by deamidating specific glutamine residues on the basis of their spacing to proline residues. The commonly accepted model, proposing that HLA-DQ8 simply favours binding of negatively charged peptides, does not take into account the fact that TG2 requires inflammation for activation and that T-cell responses against native gluten peptides are found, particularly in children. Here we show that beta57 polymorphism promotes the recruitment of T-cell receptors bearing a negative signature charge in the complementary determining region 3beta (CDR3beta) during the response against native gluten peptides presented by HLA-DQ8 in coeliac disease. These T cells showed a crossreactive and heteroclitic (stronger) response to deamidated gluten peptides. Furthermore, gluten peptide deamidation extended the T-cell-receptor repertoire by relieving the requirement for a charged residue in CDR3beta. Thus, the lack of a negative charge at position beta57 in MHC class II was met by negatively charged residues in the T-cell receptor or in the peptide, the combination of which might explain the role of HLA-DQ8 in amplifying the T-cell response against dietary gluten.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Celiac Disease/genetics , Celiac Disease/immunology , Glutens/immunology , HLA-DQ Antigens/genetics , Polymorphism, Genetic/genetics , Amides/chemistry , Animals , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Cross Reactions , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Gliadin/chemistry , Gliadin/immunology , Glutens/chemistry , HLA-DQ Antigens/chemistry , HLA-DQ Antigens/immunology , Humans , Hybridomas/immunology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Static Electricity
8.
J Exp Med ; 203(5): 1343-55, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16682498

ABSTRACT

Celiac disease is an intestinal inflammatory disorder induced by dietary gluten in genetically susceptible individuals. The mechanisms underlying the massive expansion of interferon gamma-producing intraepithelial cytotoxic T lymphocytes (CTLs) and the destruction of the epithelial cells lining the small intestine of celiac patients have remained elusive. We report massive oligoclonal expansions of intraepithelial CTLs that exhibit a profound genetic reprogramming of natural killer (NK) functions. These CTLs aberrantly expressed cytolytic NK lineage receptors, such as NKG2C, NKp44, and NKp46, which associate with adaptor molecules bearing immunoreceptor tyrosine-based activation motifs and induce ZAP-70 phosphorylation, cytokine secretion, and proliferation independently of T cell receptor signaling. This NK transformation of CTLs may underlie both the self-perpetuating, gluten-independent tissue damage and the uncontrolled CTL expansion leading to malignant lymphomas in severe forms of celiac disease. Because similar changes were detected in a subset of CTLs from cytomegalovirus-seropositive patients, we suggest that a stepwise transformation of CTLs into NK-like cells may underlie immunopathology in various chronic infectious and inflammatory diseases.


Subject(s)
Celiac Disease/immunology , Cell Differentiation/immunology , Cell Proliferation , Killer Cells, Natural/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , Base Sequence , Celiac Disease/complications , Celiac Disease/genetics , Celiac Disease/pathology , Chronic Disease , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Gene Expression Profiling , Gene Expression Regulation/immunology , Humans , Interferon-gamma/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/pathology , Killer Cells, Natural/pathology , Lymphoma/etiology , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , Molecular Sequence Data , Phosphorylation , Protein Processing, Post-Translational/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Cytotoxic/pathology , ZAP-70 Protein-Tyrosine Kinase/immunology
9.
J Exp Med ; 203(5): 1197-207, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16651387

ABSTRACT

Mouse and human natural killer T (NKT) cells recognize a restricted set of glycosphingolipids presented by CD1d molecules, including self iGb3 and microbial alpha-glycuronosylceramides. The importance of the canonical Valpha14-Jalpha18 TCR alpha chain for antigen recognition by NKT cells is well recognized, but the mechanisms underlying the Vbeta8, Vbeta7, and Vbeta2 bias in mouse have not been explored. To study the influences of thymic selection and the constraints of pairing with Valpha14-Jalpha18, we have created a population of mature T cells expressing Valpha14-Jalpha18 TCR alpha chain in CD1d-deficient mice and studied its recognition properties in vitro and in vivo. Transgenic cells expressed a diverse Vbeta repertoire but their recognition of endogenous ligands and synthetic iGb3 was restricted to the same biased Vbeta repertoire as expressed in natural NKT cells. In contrast, alpha-GalCer, a synthetic homologue of microbial alpha-glycuronosylceramides, was recognized by a broader set of Vbeta chains, including the biased NKT set but also Vbeta6, Vbeta9, Vbeta10, and Vbeta14. These surprising findings demonstrate that, whereas Vbeta8, Vbeta7, and Vbeta2 represent the optimal solution for recognition of endogenous ligand, many Vbeta chains that are potentially useful for the recognition of foreign lipids fail to be selected in the NKT cell repertoire.


Subject(s)
Antigen Presentation/immunology , Antigens, CD1/immunology , Glycosphingolipids/immunology , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD1/genetics , Antigens, CD1d , Autoantigens/immunology , Bacteria/immunology , Cells, Cultured , Humans , Ligands , Mice , Mice, Inbred NOD , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymus Gland/immunology
10.
J Immunol ; 172(3): 1935-44, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734779

ABSTRACT

Psoriatic arthritis is an interesting MHC class I allele associated autoimmune disease where injury is likely mediated exclusively by T cells. We used TCR beta-chain nucleotide sequencing to gain insight into the adaptive immune events responsible for this injury and determine whether the numerous oligoclonal expansions of this disease represent extreme determinant spreading among driving clones that recognize autoantigen or were non-Ag-driven, inflammation-related expansions. Because methotrexate suppresses but does not eliminate this inflammation, we hypothesized that clones persisting during methotrexate treatment would likely drive the inflammation. Seventy-six percent of the T cell clones in active tissue were polyclonal and unexpanded, accounting for 31% of transcripts. They were decreased greatly by methotrexate. Strikingly, most expanded clones in the inflamed joint did not persist during methotrexate treatment, were found only in inflammatory sites, exhibited no structural homology to one another, and were either CD4 or CD8 in lineage, suggesting they were non-autoantigen-driven, inflammation-related expansions. Only 12% of the expanded clones could be grouped into clonal sets distinguished by structurally homologous CDR3 beta-chain amino acid motifs suggesting Ag drive. These were exclusively CD8 in lineage, persisted during methotrexate administration, and were present in both joint fluid and blood implying they were candidate driver clones that recognized an autoantigen. However, a major set of putative driver clones exhibited a previously described EBV-specific beta-chain motif, emphasizing that the dominant feature of the disease was activation of multiple clones apparently lacking specificity for an inciting autoantigen.


Subject(s)
Arthritis, Psoriatic/immunology , Arthritis, Psoriatic/pathology , Autoantigens/immunology , Cell Movement/drug effects , Lymphocyte Activation/immunology , Methotrexate/administration & dosage , Sequence Analysis, DNA/methods , T-Lymphocyte Subsets/pathology , Amino Acid Sequence , Arthritis, Psoriatic/blood , Arthritis, Psoriatic/drug therapy , Autoantigens/metabolism , Base Sequence , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Lineage/genetics , Cell Lineage/immunology , Clone Cells , Complementarity Determining Regions/analysis , Complementarity Determining Regions/genetics , Humans , Lymphocyte Activation/genetics , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/genetics , Synovial Fluid/cytology , Synovial Fluid/drug effects , Synovial Fluid/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...