Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Cell Res ; 30(6): 465-474, 2020 06.
Article in English | MEDLINE | ID: mdl-32367041

ABSTRACT

Naturally arising regulatory CD4+ T (Treg) cells, which specifically express the transcription factor FoxP3 in the nucleus and CD25 and CTLA-4 on the cell surface, are a T-cell subpopulation specialized for immune suppression, playing a key role in maintaining immunological self-tolerance and homeostasis. FoxP3 is required for Treg function, especially for its suppressive activity. However, FoxP3 expression per se is not necessary for Treg cell lineage commitment in the thymus and insufficient for full Treg-type gene expression in mature Treg cells. It is Treg-specific epigenetic changes such as CpG demethylation and histone modification that can confer a stable and heritable pattern of Treg type gene expression on developing Treg cells in a FoxP3-independent manner. Anomalies in the formation of Treg-specific epigenome, in particular, Treg-specific super-enhancers, which largely include Treg-specific DNA demethylated regions, are indeed able to cause autoimmune diseases in rodents. Furthermore, in humans, single nucleotide polymorphisms in Treg-specific DNA demethylated regions associated with Treg signature genes, such as IL2RA (CD25) and CTLA4, can affect the development and function of naïve Treg cells rather than effector T cells. Such genetic variations are therefore causative of polygenic common autoimmune diseases including type 1 diabetes and rheumatoid arthritis via affecting endogenous natural Treg cells. These findings on the transcription factor network with FoxP3 at a key position as well as Treg-specific epigenetic landscape facilitate our understanding of Treg cell development and function, and can be exploited to prepare functionally stable FoxP3-expressing Treg cells from antigen-specific conventional T cells to treat autoimmune diseases.


Subject(s)
Autoimmune Diseases/pathology , CTLA-4 Antigen , Forkhead Transcription Factors , Interleukin-2 Receptor alpha Subunit , T-Lymphocytes, Regulatory , Animals , CTLA-4 Antigen/genetics , CTLA-4 Antigen/physiology , DNA Methylation , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Gene Expression Regulation , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/physiology , Polymorphism, Single Nucleotide , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
2.
PLoS One ; 12(8): e0182532, 2017.
Article in English | MEDLINE | ID: mdl-28792982

ABSTRACT

The recruitment and retention of Natural Killer (NK) cells in the liver are thought to play an important role during hepatotropic infections and liver cirrhosis. The aims of this study were to determine differences between liver-derived and peripheral blood-derived NK cells in the context of liver inflammation and cirrhosis. We conducted a prospective dual-center cross-sectional study in patients undergoing liver transplantation or tumor-free liver resections, in which both liver tissue and peripheral blood samples were obtained from each consenting study participants. Intrahepatic lymphocytes and PBMCs were stained, fixed and analyzed by flow cytometry. Our results showed that, within cirrhotic liver samples, intrahepatic NK cells were particularly enriched for CD49a+ NK cells when compared to tumor-free liver resection samples. CD49a+ liver-derived NK cells included populations of cells expressing CD25, CD34 and CXCR3. Moreover, CD49a+CD25+ liver-derived NK cells exhibited high proliferative capacity in vitro in response to low doses of IL-2. Our study identified a specific subset of CD49a+CD25+ NK cells in cirrhotic livers bearing functional features of proliferation.


Subject(s)
Cell Proliferation/physiology , Integrin alpha1/physiology , Interleukin-2 Receptor alpha Subunit/physiology , Killer Cells, Natural/physiology , Liver/cytology , Adult , Aged , Antigens, CD34/physiology , Cross-Sectional Studies , Female , Flow Cytometry , Humans , Killer Cells, Natural/immunology , Liver/immunology , Liver/physiology , Liver Transplantation , Male , Middle Aged , Prospective Studies , Receptors, CXCR3/physiology
3.
J Immunol ; 197(6): 2208-18, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27511734

ABSTRACT

CD4(+)Foxp3(+) regulatory T cells (Tregs) are key immune suppressors that regulate immunity in diverse tissues. The tissue and/or inflammatory signals that influence the magnitude of the Treg response remain unclear. To define signals that promote Treg accumulation, we developed a simple system of skin inflammation using defined Ags and adjuvants that induce distinct cytokine milieus: OVA protein in CFA, aluminum salts (Alum), and Schistosoma mansoni eggs (Sm Egg). Polyclonal and Ag-specific Treg accumulation in the skin differed significantly between adjuvants. CFA and Alum led to robust Treg accumulation, with >50% of all skin CD4(+) T cells being Foxp3(+) In contrast, Tregs accumulated poorly in the Sm Egg-inflamed skin. Surprisingly, we found no evidence of inflammation-specific changes to the Treg gene program between adjuvant-inflamed skin types, suggesting a lack of selective recruitment or adaptation to the inflammatory milieu. Instead, Treg accumulation patterns were linked to differences in CD80/CD86 expression by APC and the regulation of CD25 expression, specifically in the inflamed skin. Inflammatory cues alone, without cognate Ag, differentially supported CD25 upregulation (CFA and Alum > Sm Egg). Only in inflammatory milieus that upregulated CD25 did the provision of Ag enhance local Treg proliferation. Reduced IL-33 in the Sm Egg-inflamed environment was shown to contribute to the failure to upregulate CD25. Thus, the magnitude of the Treg response in inflamed tissues is controlled at two interdependent levels: inflammatory signals that support the upregulation of the important Treg survival factor CD25 and Ag signals that drive local expansion.


Subject(s)
Dermatitis/immunology , Interleukin-2 Receptor alpha Subunit/physiology , Lymphocyte Activation , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , B7-1 Antigen/analysis , B7-2 Antigen/analysis , Female , Inflammation/immunology , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Lymphocyte Count , Mice , Mice, Inbred BALB C
4.
Leukemia ; 30(5): 1033-43, 2016 05.
Article in English | MEDLINE | ID: mdl-26707936

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is a high-risk subset of acute leukemia, characterized by frequent activation of Notch1 or AKT signaling, where new therapeutic approaches are needed. We showed previously that cyclin-dependent kinase 6 (CDK6) is required for thymic lymphoblastic lymphoma induced by activated AKT. Here, we show CDK6 is required for initiation and maintenance of Notch-induced T-ALL. In a mouse retroviral model, hematopoietic stem/progenitor cells lacking CDK6 protein or expressing kinase-inactive (K43M) CDK6 are resistant to induction of T-ALL by activated Notch, whereas those expressing INK4-insensitive (R31C) CDK6 are permissive. Pharmacologic inhibition of CDK6 kinase induces CD25 and RUNX1 expression, cell cycle arrest and apoptosis in mouse and human T-ALL. Ablation of Cd25 in a K43M background restores Notch-induced T leukemogenesis, with disease that is resistant to CDK6 inhibitors in vivo. These data support a model whereby CDK6-mediated suppression of CD25 is required for initiation of T-ALL by activated Notch1, and CD25 induction mediates the therapeutic response to CDK6 inhibition in established T-ALL. These results both validate CDK6 as a molecular target for therapy of this subset of T-ALL and suggest that CD25 expression could serve as a biomarker for responsiveness of T-ALL to CDK4/6 inhibitor therapy.


Subject(s)
Cyclin-Dependent Kinase 6/physiology , Interleukin-2 Receptor alpha Subunit/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Receptor, Notch1/physiology , Animals , Apoptosis/drug effects , Carcinogenesis/metabolism , Cell Cycle Checkpoints/drug effects , Core Binding Factor Alpha 2 Subunit/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Receptor, Notch1/metabolism
5.
Immunity ; 43(4): 690-702, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26410627

ABSTRACT

The differentiation of CD4(+) helper T cell subsets with diverse effector functions is accompanied by changes in metabolism required to meet their bioenergetic demands. We find that follicular B helper T (Tfh) cells exhibited less proliferation, glycolysis, and mitochondrial respiration, accompanied by reduced mTOR kinase activity compared to T helper 1 (Th1) cells in response to acute viral infection. IL-2-mediated activation of the Akt kinase and mTORc1 signaling was both necessary and sufficient to shift differentiation away from Tfh cells, instead promoting that of Th1 cells. These findings were not the result of generalized signaling attenuation in Tfh cells, because they retained the ability to flux calcium and activate NFAT-transcription-factor-dependent cytokine production. These data identify the interleukin-2 (IL-2)-mTORc1 axis as a critical orchestrator of the reciprocal balance between Tfh and Th1 cell fates and their respective metabolic activities after acute viral infection.


Subject(s)
Interleukin-2/physiology , Multiprotein Complexes/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases/physiology , Animals , Apoptosis , Calcium Signaling , Cell Cycle , Cell Division , Enzyme Activation , Glucose/metabolism , Glycolysis , Interleukin-2 Receptor alpha Subunit/physiology , Lymphocytic choriomeningitis virus/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , NFATC Transcription Factors/physiology , Oxygen Consumption , Positive Regulatory Domain I-Binding Factor 1 , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
6.
J Neuroimmunol ; 281: 51-60, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25867468

ABSTRACT

Regulatory T cells were investigated in early-onset (EO) and late-onset (LO) myasthenia gravis patients with anti-acetylcholine receptor antibody (AChR-MG). Alterations in PD-1 and PD-L1 on CD4(+)CD25(++) (Treg) and responder T cells (Tresp, CD4(+)CD25(-)) were observed in LOMG patients. GITR was decreased on CD4(+)CD25(++) of all patients. Decrease of FOXP3 was associated with lower phosphorylation of STAT5.1,25-dihydroxyvitamin D3 (VitD3) increased suppression in co-culture with a stronger effect in patients by acting possibly both on cell groups. Changes in surface molecules and intracellular pathways contribute to the defects of Treg in non-thymomatous AChR-MG and VitD3 can have modulatory effects.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Calcitriol/pharmacology , Interleukin-2 Receptor alpha Subunit/physiology , Myasthenia Gravis/metabolism , Phenotype , STAT5 Transcription Factor/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Cells, Cultured , Coculture Techniques , Female , Humans , Male , Middle Aged , Signal Transduction/drug effects , Signal Transduction/physiology , Young Adult
7.
Stem Cells ; 33(3): 870-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25524029

ABSTRACT

The successful suppression of human immunodeficiency virus (HIV) in the "Berlin Patient" has highlighted the ability of HIV-resistant hematopoietic stem cells to offer a potential functional cure for HIV-infected patients. HIV stem cell gene therapy can mimic this result by genetically modifying a patient's own cells with anti-HIV genes. Previous attempts of HIV gene therapy have been hampered by a low percentage of transplanted HIV-resistant cells which has led to minimal clinical efficacy. In our current study, we have evaluated the in vitro and in vivo safety and efficacy of a truncated/mutated form of human CD25 preselective anti-HIV lentiviral vector in human hematopoietic stem cells. This preselective vector allows us to purify vector-transduced cells prior to transplantation so an increased percentage of gene-modified cells can be delivered. Here, we demonstrate the safety of this strategy with successful engraftment and multilineage hematopoiesis of transduced cells in a humanized NOD-RAG1-/-IL-2rγ-/- knockout mouse model. Efficacy was also demonstrated with significant protection from HIV-1 infection including maintenance of human CD4+ cell levels and a decrease in HIV-1 plasma viremia. Collectively, these results establish the utility of this HIV stem cell gene therapy strategy and bring it closer to providing a functional cure for HIV-infected patients.


Subject(s)
Genetic Therapy/methods , HIV Infections/therapy , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Interleukin-2 Receptor alpha Subunit/physiology , Animals , Gene Expression , Genetic Vectors/genetics , HIV Infections/genetics , HIV Infections/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Lentivirus/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Transduction, Genetic/methods
8.
Anticancer Res ; 34(8): 4489-95, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25075091

ABSTRACT

ZFAT (zinc-finger gene in AITD susceptibility region), originally identified as a candidate susceptibility gene for autoimmune thyroid disease, has been reported to be involved in various cellular processes and several common diseases including multiple sclerosis. Recent studies revealed that mouse Zfat is a novel critical regulator for both thymocyte differentiation and peripheral T-cell homeostasis. Zfat deficiency at early thymocyte developmental stages results in the inhibition of the development of CD4(+)CD8(+) thymocytes with an impaired positive selection. Zfat deficiency in peripheral T-cells results in a reduction in the number of T-cells with decreased expression of the interleukin-7 receptor-α (IL-7Rα) that is critical for T-cell homeostasis. In addition, T-cell antigen receptor stimulation-induced responses of Zfat-deficient T-cells are also impaired, with reduced IL-2Rα expression. This review highlights and discusses the roles of Zfat in thymocyte differentiation of T-cells and in the homeostasis of naive T-cells with recent work.


Subject(s)
Homeostasis , T-Lymphocytes/cytology , Transcription Factors/physiology , Animals , Cell Differentiation , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/physiology , Receptors, Antigen, T-Cell/immunology
9.
Vet Dermatol ; 25(5): 456-e72, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24913127

ABSTRACT

BACKGROUND: T Cells play a major role in the immunopathogenesis of canine atopic dermatitis (cAD). However, the significance of cutaneous regulatory T cells (Tregs) and CD8(+) T cells is currently unclear. HYPOTHESIS/OBJECTIVES: The study aimed to evaluate the presence and distribution of Tregs in cAD and healthy skin and to determine the cytokine production of cutaneous CD4(+) and CD8(+) T cells. ANIMALS: Biopsies were taken from four dogs with cAD (lesional and nonlesional skin) and four healthy control dogs. METHODS: Distribution patterns of T-cell subtypes in cAD lesional, nonlesional and control skin were evaluated by immunohistochemistry. Phenotypic characterization of T cells from skin explant cultures and enzymatic digestions was performed using flow cytometry. Cytokine production of sorted CD4(+) and CD8(+) explant-derived T cells was measured by RT-qPCR. RESULTS: Regulatory T cells phenotypically characterized by CD25(+) FoxP3(+) were found in both CD4(+) and CD8(+) subsets of skin explant and digestion samples. The percentages of CD4(+) CD25(+) cells that were FoxP3(+) were similar in cAD and control skin. In atopic lesional and nonlesional explant samples, lower FoxP3(+) percentages of CD8(+) CD25(+) cells were seen compared with control skin. The presence of predominantly periadnexal CD25(+) FoxP3(+) cells was confirmed by immunohistochemistry in lesional, nonlesional and control skin. The CD4(+) /CD8(+) ratio was less than one in cAD skin with both skin explant and digestion methods. CD4(+) and CD8(+) T-cell subsets of lesional and nonlesional cAD skin were capable of producing interleukin-13, interleukin-22 and interferon-γ. CONCLUSIONS AND CLINICAL IMPORTANCE: Both CD4(+) and CD8(+) T cells are likely to contribute to the immunopathogenesis of cAD through the production of interleukin-13, interleukin-22 and interferon-γ. In both subsets, functional analysis of FoxP3(+) cells is essential to determine their role.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Dermatitis, Atopic/veterinary , Dog Diseases/immunology , Forkhead Transcription Factors/physiology , Interferon-gamma/physiology , Interleukin-13/physiology , Interleukin-2 Receptor alpha Subunit/physiology , Interleukin-2/physiology , Skin/cytology , T-Lymphocyte Subsets/physiology , Animals , Cells, Cultured , Dermatitis, Atopic/immunology , Dogs , Flow Cytometry/veterinary , Forkhead Transcription Factors/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Real-Time Polymerase Chain Reaction/veterinary , Skin/immunology , T-Lymphocyte Subsets/immunology
11.
Am J Transplant ; 14(5): 1032-45, 2014 May.
Article in English | MEDLINE | ID: mdl-24618297

ABSTRACT

CD4(+) Foxp3(+) T regulatory cells (Tregs ) are essential for maintaining immunological tolerance, which could be harnessed for novel cell-based therapies to prevent allograft rejection and control autoimmunity. However, the use of Tregs for therapy is hindered by the inability to generate sufficient cell numbers to inhibit desired immune response(s) and achieve stable engraftment of the donor-Treg cell inoculums. The present study was undertaken to investigate the in vivo requirements to promote engraftment of adoptively transferred Tregs and induce tolerance. We established that not only is peripheral space required, but competition from endogenous Tregs must be minimized for successful donor-Treg engraftment with IL-2 critical for driving their proliferation and survival. Moreover, these studies revealed a critical level of donor-Treg engraftment was required for tolerance induction to skin transplants. These mouse studies lay the foundation for development of novel Treg approaches for tolerance induction in the clinic involving not only organ or cellular transplantation, but also to re-establish self-tolerance in autoimmune settings.


Subject(s)
Autoimmunity , Graft Rejection/immunology , Interleukin-2 Receptor alpha Subunit/physiology , T-Lymphocytes, Regulatory/immunology , Tissue Donors , Transplantation Tolerance/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Female , Immunotherapy , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocyte Depletion , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Skin Diseases/immunology , Skin Diseases/metabolism , Skin Diseases/therapy , Skin Transplantation
12.
Blood ; 123(16): 2540-9, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24574458

ABSTRACT

Just as normal stem cells require niche cells for survival, leukemia-initiating cells (LICs) may also require niche cells for their maintenance. Chronic myeloid leukemia (CML) is caused by the activity of BCR-ABL, a constitutively active tyrosine kinase. CML therapy with tyrosine kinase inhibitors is highly effective; however, due to the persistence of residual LICs, it is not curative. Several factors are known to support CML LICs, but purification of LICs and a thorough understanding of their niche signals have not yet been achieved. Using a CML-like mouse model of myeloproliferative disease, we demonstrate that CML LICs can be divided into CD25(+)FcεRIα(-) Lineage marker (Lin)(-) Sca-1(+)c-Kit(+) (F(-)LSK) cells and CD25(-)F(-)LSK cells. The CD25(+)F(-)LSK cells had multilineage differentiation capacity, with a preference toward cytokine-producing mast cell commitment. Although cells interconverted between CD25(-)F(-)LSK and CD25(+)F(-)LSK status, the CD25(+)F(-)LSK cells exhibited higher LIC capacity. Our findings suggest that interleukin-2 derived from the microenvironment and CD25 expressed on CML LICs constitute a novel signaling axis. The high levels of CD25 expression in the CD34(+)CD38(-) fraction of human CML cells indicate that CD25(+) LICs constitute an "LIC-derived niche" that could be preferentially targeted in therapy for CML.


Subject(s)
Interleukin-2 Receptor alpha Subunit/physiology , Interleukin-2/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Animals , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/metabolism , Signal Transduction/physiology , Th2 Cells/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
14.
Nephrol Dial Transplant ; 29(5): 1005-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24322577

ABSTRACT

BACKGROUND: Recent studies have reported that podocytes are postnatally generated from progenitor cells localized in Bowman's capsule or in the bone marrow. In the present study, we investigated whether or not podocyte regeneration is important in the repair of injured glomeruli after mild podocyte injury in mice. METHODS: Mild podocyte injury was induced in NEP25 mice (n = 8) by injecting an immunotoxin, LMB2 (0.625 ng/g body weight). Control mice, not injured by LMB2 injection (n = 7) was used as a comparison. Proliferating cells were labeled by continuous infusion of bromodeoxyuridine (BrdU). Podocytes, identified by nephrin, WT1 or podocin staining, that had incorporated BrdU were enumerated 4 weeks later. RESULTS: A total of 742 corpuscles were inspected in serial sections stained for BrdU and nephrin; 19% showed sclerosis. BrdU(+) cells were observed in both the glomeruli and Bowman's capsules, averaging 2.5 ± 3.1 in non-sclerotic corpuscles and 7.0 ± 5.8 in sclerotic corpuscles. Only one BrdU(+) cell was also positive for nephrin. Another cell, localized at a position consistent with its potential identification as a podocyte, was nephrin negative but had incorporated BrdU. WT1 staining similarly revealed that only two nuclei were doubly positive for BrdU and WT1. Additional 1676 corpuscles were inspected by double staining for BrdU and podocin; none were doubly positive. CONCLUSIONS: Podocytes are not replenished by proliferation of endogenous progenitor cells in mice with glomerular injury.


Subject(s)
Immunotoxins/toxicity , Interleukin-2 Receptor alpha Subunit/physiology , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Podocytes/cytology , Regeneration/physiology , Animals , Cell Proliferation , Female , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Glomerulus/injuries , Mice , Mice, Inbred C57BL , Podocytes/metabolism , Stem Cells/cytology , Stem Cells/metabolism
15.
Cancer Res ; 73(21): 6494-503, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24030978

ABSTRACT

Carbonic anhydrase XII (CA XII) is a membrane-tethered cell surface enzyme that is highly expressed on many human tumor cells. Carbonic anhydrase members in this class of exofacial molecules facilitate tumor metabolism by facilitating CO2 venting and intracellular pH regulation. Accordingly, inhibition of exofacial CAs has been proposed as a general therapeutic strategy to target cancer. The recent characterization of 6A10, the first CA XII-specific inhibitory monoclonal antibody, offered an opportunity to evaluate this strategy with regard to CA XII-mediated catalysis. Using functional assays, we showed that 6A10 inhibited exofacial CA activity in CA XII-expressing cancer cells. 6A10 reduced spheroid growth in vitro under culture conditions where CA XII was active (i.e., alkaline pH) and where its catalytic activity was likely rate-limiting (i.e., restricted extracellular HCO3-supply). These in vitro results argued that the antibody exerted its growth-retarding effect by acting on the catalytic process, rather than on antigen binding per se. Notably, when administered in a mouse xenograft model of human cancer, 6A10 exerted a significant delay on tumor outgrowth. These results corroborate the notion that exofacial CA is critical for cancer cell physiology and they establish the immunotherapeutic efficacy of targeting CA XII using an inhibitory antibody.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/chemistry , Cell Proliferation/drug effects , Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Blotting, Western , Carbonic Anhydrases/metabolism , Catalysis , Cell Membrane/drug effects , Cell Membrane/metabolism , Fluorescent Antibody Technique , Humans , Hydrogen-Ion Concentration , Immunoprecipitation , Interleukin-2 Receptor alpha Subunit/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/enzymology , Neoplasms/immunology , Spheroids, Cellular , Tumor Cells, Cultured
16.
Cancer Res ; 73(21): 6484-93, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24072748

ABSTRACT

Ribonucleotide reductase (RNR) is an attractive target for anticancer agents given its central function in DNA synthesis, growth, metastasis, and drug resistance of cancer cells. The current clinically established RNR inhibitors have the shortcomings of short half-life, drug resistance, and iron chelation. Here, we report the development of a novel class of effective RNR inhibitors addressing these issues. A novel ligand-binding pocket on the RNR small subunit (RRM2) near the C-terminal tail was proposed by computer modeling and verified by site-directed mutagenesis and nuclear magnetic resonance (NMR) techniques. A compound targeting this pocket was identified by virtual screening of the National Cancer Institute (NCI) diverse small-molecule database. By lead optimization, we developed the novel RNR inhibitor COH29 that acted as a potent inhibitor of both recombinant and cellular human RNR enzymes. COH29 overcame hydroxyurea and gemcitabine resistance in cancer cells. It effectively inhibited proliferation of most cell lines in the NCI 60 human cancer panel, most notably ovarian cancer and leukemia, but exerted little effect on normal fibroblasts or endothelial cells. In mouse xenograft models of human cancer, COH29 treatment reduced tumor growth compared with vehicle. Site-directed mutagenesis, NMR, and surface plasmon resonance biosensor studies confirmed COH29 binding to the proposed ligand-binding pocket and offered evidence for assembly blockade of the RRM1-RRM2 quaternary structure. Our findings offer preclinical validation of COH29 as a promising new class of RNR inhibitors with a new mechanism of inhibition, with broad potential for improved treatment of human cancer.


Subject(s)
Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Protein Conformation/drug effects , Ribonucleotide Reductases/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides/pharmacology , Blotting, Western , Cell Cycle/drug effects , Chromatography, High Pressure Liquid , Computer Simulation , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Flow Cytometry , Half-Life , Humans , Hydroxyurea/pharmacology , Interleukin-2 Receptor alpha Subunit/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Mutagenesis, Site-Directed , Mutation/genetics , Neoplasms/metabolism , Neoplasms/pathology , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/genetics , Small Molecule Libraries , Structure-Activity Relationship , Surface Plasmon Resonance , Tandem Mass Spectrometry , Thiazoles/pharmacology , Gemcitabine
17.
J Allergy Clin Immunol ; 130(3): 781-797.e11, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22935591

ABSTRACT

BACKGROUND: Allergic contact dermatitis is one of the most common occupational diseases. A main protective mechanism in those who do not develop allergic contact dermatitis is tolerance induction by repeated exposure to low doses of contact allergen, which is termed low zone tolerance (LZT). The mechanisms that determine the tolerance induction in subjects with LZT are still elusive. OBJECTIVE: We performed analysis of the role of CD4(+)CD25(+) forkhead box protein 3 (FOXP3)-positive regulatory T (Treg) cells and dendritic cells (DCs) in mice with LZT. METHODS: Mechanisms of tolerance induction were analyzed in a murine model of LZT by using FOXP3 and IL-10 reporter mice, as well as mice that allow the selective depletion of Treg cells or DCs. RESULTS: Depletion of CD4(+)CD25(+)FOXP3(+) Treg cells during tolerance induction completely abolishes the development of LZT, resulting in a pronounced contact hypersensitivity response. Adoptive transfer experiments, depletion studies, and use of cell type-specific deficient mice revealed that IL-10 production is critical for the suppressor function of Treg cells in mice with LZT and that tolerogenic CD8(+)CD11c(+) DCs located in the skin-draining lymph nodes are essential for LZT. In the absence of Treg cells, DCs did not develop tolerogenic functions, indicating that activated IL-10(+) Treg cells might imprint the tolerogenic DC phenotype. Cell communication analysis revealed that the education of tolerogenic DCs might involve a direct interaction with Treg cells mediated by gap junctions. Subsequently, induction of tolerogenic CD11c(+) DCs leads to the generation of hapten-specific CD8(+) Treg cells, which protect against contact hypersensitivity. CONCLUSIONS: Our data demonstrate critical interactions between CD4(+)CD25(+)FOXP3(+) Treg cells and tolerogenic CD8(+)CD11c(+) DCs during the induction of LZT.


Subject(s)
Cell Communication , Dendritic Cells/physiology , Dermatitis, Allergic Contact/prevention & control , Immune Tolerance , T-Lymphocytes, Regulatory/physiology , Animals , CD11c Antigen/analysis , Dermatitis, Allergic Contact/immunology , Forkhead Transcription Factors/analysis , Interleukin-10/physiology , Interleukin-2 Receptor alpha Subunit/physiology , Lymphocyte Activation , Mice , Mice, Transgenic , Receptors, CCR7/analysis
18.
J Immunol ; 189(2): 755-66, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706088

ABSTRACT

Hepatitis C virus (HCV) is remarkable at disrupting human immunity to establish chronic infection. Upregulation of inhibitory signaling pathways (such as T cell Ig and mucin domain protein-3 [Tim-3]) and accumulation of regulatory T cells (Tregs) play pivotal roles in suppressing antiviral effector T cell (Teff) responses that are essential for viral clearance. Although the Tim-3 pathway has been shown to negatively regulate Teffs, its role in regulating Foxp3(+) Tregs is poorly explored. In this study, we investigated whether and how the Tim-3 pathway alters Foxp3(+) Treg development and function in patients with chronic HCV infection. We found that Tim-3 was upregulated, not only on IL-2-producing CD4(+)CD25(+)Foxp3(-) Teffs, but also on CD4(+)CD25(+)Foxp3(+) Tregs, which accumulate in the peripheral blood of chronically HCV-infected individuals when compared with healthy subjects. Tim-3 expression on Foxp3(+) Tregs positively correlated with expression of the proliferation marker Ki67 on Tregs, but it was inversely associated with proliferation of IL-2-producing Teffs. Moreover, Foxp3(+) Tregs were found to be more resistant to, and Foxp3(-) Teffs more sensitive to, TCR activation-induced cell apoptosis, which was reversible by blocking Tim-3 signaling. Consistent with its role in T cell proliferation and apoptosis, blockade of Tim-3 on CD4(+)CD25(+) T cells promoted expansion of Teffs more substantially than Tregs through improving STAT-5 signaling, thus correcting the imbalance of Foxp3(+) Tregs/Foxp3(-) Teffs that was induced by HCV infection. Taken together, the Tim-3 pathway appears to control Treg and Teff balance through altering cell proliferation and apoptosis during HCV infection.


Subject(s)
Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/pathology , Receptors, Immunologic/physiology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/virology , Apoptosis Regulatory Proteins/physiology , Cell Proliferation , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/physiology , Hepatitis C, Chronic/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/physiology , Pilot Projects , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/pathology , Viremia/immunology , Viremia/metabolism , Viremia/pathology
19.
J Neuroinflammation ; 9: 139, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22727044

ABSTRACT

BACKGROUND: DNA vaccines represent promising therapeutic strategies in autoimmune disorders such as multiple sclerosis (MS). However, the precise mechanisms by which DNA vaccines induce immune regulation remain largely unknown. Here, we aimed to expand previous knowledge existing on the mechanisms of action of DNA vaccines in the animal model of MS, experimental autoimmune encephalomyelitis (EAE), by treating EAE mice with a DNA vaccine encoding the myelin oligodendrocyte glycoprotein (MOG), and exploring the therapeutic effects on the disease-induced inflammatory and neurodegenerative changes. METHODS: EAE was induced in C57BL6/J mice by immunization with MOG35₋55 peptide. Mice were intramuscularly treated with a MOG-DNA vaccine or vehicle in prophylactic and therapeutic approaches. Histological studies were performed in central nervous system (CNS) tissue. Cytokine production and regulatory T cell (Treg) quantification were achieved by flow cytometry. Gene expression patterns were determined using microarrays, and the main findings were validated by real-time PCR. RESULTS: MOG-DNA treatment reduced the clinical and histopathological signs of EAE when administered in both prophylactic and therapeutic settings. Suppression of clinical EAE was associated with dampening of antigen (Ag)-specific proinflammatory Th1 and Th17 immune responses and, interestingly, expansion of Treg in the periphery and upregulation in the CNS of genes encoding neurotrophic factors and proteins involved in remyelination. CONCLUSIONS: These results suggest for the first time that the beneficial effects of DNA vaccines in EAE are not limited to anti-inflammatory mechanisms, and DNA vaccines may also exert positive effects through hitherto unknown neuroprotective mechanisms.


Subject(s)
CD4 Antigens/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Forkhead Transcription Factors/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Up-Regulation/immunology , Vaccines, DNA/administration & dosage , Animals , CD4 Antigens/genetics , CD4 Antigens/physiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/physiology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Treatment Outcome , Up-Regulation/genetics , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
20.
J Immunol ; 188(11): 5785-91, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22539790

ABSTRACT

In the current study, we showed that in vivo administration of an anti-CD25 Ab (PC61) decreased the Th17 response in C57BL/6 mice immunized with the uveitogenic peptide interphotoreceptor retinoid-binding protein, while enhancing the autoreactive Th1 response. The depressed Th17 response was closely associated with decreased numbers of a splenic dendritic cell (DC) subset expressing CD11c(+)CD3(-)CD25(+) and decreased expansion of γδ T cells. We demonstrated that ablation of the CD25(+) DC subset accounted for the decreased activation and the expansion of γδ T cells, leading to decreased activation of IL-17(+) interphotoreceptor retinoid-binding protein-specific T cells. Our results show that an enhanced Th17 response in an autoimmune disease is associated with the appearance of a DC subset expressing CD25 and that treatment of mice with anti-CD25 Ab causes functional alterations in a number of immune cell types, namely DCs and γδ T cells, in addition to CD25(+)αßTCR(+) regulatory T cells.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Differentiation/immunology , Dendritic Cells/immunology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Th17 Cells/immunology , Uveitis/immunology , Uveitis/pathology , Animals , Autoimmune Diseases/metabolism , Dendritic Cells/metabolism , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Interleukin-2 Receptor alpha Subunit/physiology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Th17 Cells/pathology , Uveitis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...