Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Publication year range
1.
Cell ; 184(14): 3812-3828.e30, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34214472

ABSTRACT

We study a patient with the human papilloma virus (HPV)-2-driven "tree-man" phenotype and two relatives with unusually severe HPV4-driven warts. The giant horns form an HPV-2-driven multifocal benign epithelial tumor overexpressing viral oncogenes in the epidermis basal layer. The patients are unexpectedly homozygous for a private CD28 variant. They have no detectable CD28 on their T cells, with the exception of a small contingent of revertant memory CD4+ T cells. T cell development is barely affected, and T cells respond to CD3 and CD2, but not CD28, costimulation. Although the patients do not display HPV-2- and HPV-4-reactive CD4+ T cells in vitro, they make antibodies specific for both viruses in vivo. CD28-deficient mice are susceptible to cutaneous infections with the mouse papillomavirus MmuPV1. The control of HPV-2 and HPV-4 in keratinocytes is dependent on the T cell CD28 co-activation pathway. Surprisingly, human CD28-dependent T cell responses are largely redundant for protective immunity.


Subject(s)
CD28 Antigens/deficiency , Inheritance Patterns/genetics , Papillomaviridae/physiology , Skin/virology , T-Lymphocytes/immunology , Adult , Amino Acid Sequence , Animals , Base Sequence , CD28 Antigens/genetics , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Child , Endopeptidases/metabolism , Female , Genes, Recessive , HEK293 Cells , Homozygote , Humans , Immunity, Humoral , Immunologic Memory , Jurkat Cells , Keratinocytes/pathology , Male , Mice, Inbred C57BL , Oncogenes , Papilloma/pathology , Papilloma/virology , Pedigree , Protein Sorting Signals , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
J Immunol ; 199(12): 3972-3980, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29093061

ABSTRACT

Follicular regulatory T (TFR) cells are a newly defined regulatory T cell (Treg) subset that suppresses follicular helper T cell-mediated B cell responses in the germinal center reaction. The precise costimulatory signal requirements for proper TFR cell differentiation and function are still not known. Using conditional knockout strategies of CD28, we previously demonstrated that loss of CD28 signaling in Tregs caused autoimmunity in mice (termed CD28-ΔTreg mice), characterized by lymphadenopathy, accumulation of activated T cells, and cell-mediated inflammation of the skin and lung. In this study, we show that CD28 signaling is required for TFR cell differentiation. Treg-specific deletion of CD28 caused a reduction in TFR cell numbers and function, which resulted in increased germinal center B cells and Ab production. Moreover, residual CD28-deficient TFR cells showed a diminished suppressive capacity as assessed by their ability to inhibit Ab responses in vitro. Surprisingly, genetic deletion of B cells in CD28-ΔTreg mice prevented the development of lymphadenopathy and CD4+ T cell activation, and autoimmunity that mainly targeted skin and lung tissues. Thus, autoimmunity occurring in mice with CD28-deficient Tregs appears to be driven primarily by loss of TFR cell differentiation and function with resulting B cell-driven inflammation.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/immunology , B-Lymphocytes/immunology , CD28 Antigens/deficiency , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibody Formation , Autoimmune Diseases/etiology , Autoimmune Diseases/pathology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Female , Germinal Center/immunology , Immune Tolerance/immunology , Lung/immunology , Lung/pathology , Lymphadenopathy/etiology , Lymphadenopathy/immunology , Lymphadenopathy/prevention & control , Lymphocyte Depletion , Lymphopoiesis , Mice , Organ Specificity , Skin/immunology , Skin/pathology , Specific Pathogen-Free Organisms , T-Lymphocytes, Regulatory/classification
4.
Clin Exp Rheumatol ; 36(6): 1008-1013, 2018.
Article in English | MEDLINE | ID: mdl-29745892

ABSTRACT

OBJECTIVES: To determine whether the CD4+CD28null T-cells subpopulation predicts the occurrence of damage in SLE. METHODS: This longitudinal study was conducted in consecutive SLE patients seen every six months in our Rheumatology Department since 2012. Patients in whom CD4+CD28null T-cells had been measured and who had at least one subsequent visit were included in the study. Survival analyses (univariable and multivariable Cox-regression models) were performed to determine the risk of overall and domain damage (as per the SLICC Damage Index - SDI) as a function of the frequency of this T-cell subpopulation. The multivariable model was adjusted for pertinent confounders. All analyses were performed using SPSS 21.0. RESULTS: One hundred and nineteen patients were evaluated; their mean (SD) age was 43.5 (11.9) years, 113 (95.0%) were female. Disease duration was 7.8 (7.0) years, the SLEDAI 5.3 (4.1) and the SDI 1.0 (1.4). The percentage of CD4+CD28null T-cells was 17.4 (14.0). The mean follow-up was 2.1 (0.8) years, and the mean number of visits per patient 3.5 (1.1). Forty-six (38.7%) patients increase at least one SDI point. In the univariable and multivariable analyses, the percentage of CD4+CD28null predicted the occurrence of lung damage [HR: 1.042 (CI95%: 1.001-1.085); p=0.047 and HR: 1.099 (CI95%1.020-1.184); p=0.013, respectively] but neither the total SDI score nor all other SDI domain scores were predicted by the percentage of CD4+CD28null cells. CONCLUSIONS: In SLE patients, CD4+CD28null T-cells predict the occurrence of new lung damage, independently of other risk factors but not of overall damage or damage on other domains.


Subject(s)
CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Lung/immunology , Lupus Erythematosus, Systemic/immunology , Adult , Antimalarials/therapeutic use , CD28 Antigens/blood , CD28 Antigens/deficiency , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Disease Progression , Female , Humans , Immunosuppressive Agents/therapeutic use , Longitudinal Studies , Lung/drug effects , Lung/metabolism , Lung/pathology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/drug therapy , Male , Middle Aged , Peru , Prognosis , Risk Factors , Time Factors
5.
Proc Natl Acad Sci U S A ; 112(2): 524-9, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25548162

ABSTRACT

Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) is an essential regulator of T-cell responses, and its absence precipitates lethal T-cell hyperactivity. However, whether CTLA-4 acts simply to veto the activation of certain clones or plays a more nuanced role in shaping the quality of T-cell responses is not clear. Here we report that T cells in CTLA-4-deficient mice show spontaneous T-follicular helper (T(FH)) differentiation in vivo, and this is accompanied by the appearance of large germinal centers (GCs). Remarkably, short-term blockade with anti-CTLA-4 antibody in wild-type mice is sufficient to elicit T(FH) generation and GC development. The latter occurs in a CD28-dependent manner, consistent with the known role of CTLA-4 in regulating the CD28 pathway. CTLA-4 can act by down-regulating CD80 and CD86 on antigen presenting cells (APCs), thereby altering the level of CD28 engagement. To mimic reduced CD28 ligation, we used mice heterozygous for CD28, revealing that the magnitude of CD28 engagement is tightly linked to the propensity for T(FH) differentiation. In contrast, other parameters of T-cell activation, including CD62L down-regulation and Ki67 expression, were relatively insensitive to altered CD28 level. Altered T(FH) generation as a result of graded reduction in CD28 was associated with decreased numbers of GC B cells and a reduction in overall GC size. These data support a model in which CTLA-4 control of immunity goes beyond vetoing T-cell priming and encompasses the regulation of T(FH) differentiation by graded control of CD28 engagement.


Subject(s)
CD28 Antigens/metabolism , CTLA-4 Antigen/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity , Animals , Autoantibodies/biosynthesis , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD28 Antigens/deficiency , CD28 Antigens/genetics , CTLA-4 Antigen/deficiency , CTLA-4 Antigen/genetics , Cell Differentiation/immunology , Germinal Center/cytology , Germinal Center/immunology , Heterozygote , Ligands , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Immunological
6.
J Immunol ; 194(9): 4162-74, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25825447

ABSTRACT

In patients receiving allogeneic hematopoietic cell transplantation, chronic graft-versus-host disease (cGVHD) remains a frequent complication and resembles autoimmune diseases such as systemic lupus erythematosus and systemic sclerosis. Our previous work demonstrated the critical role of CD28 costimulation of donor T cells for GVHD induction. In this study, we investigate the role of CD28 costimulation of host T cells in cGVHD. CD28-intact mice as hosts showed systemic lupus erythematosus-type cGVHD, whereas CD28-deficient mice developed a distinct phenotype of cGVHD, with fibrotic damage in skin and internal organs, resembling systemic sclerosis. This phenotype was due to a lack of signaling through the C-terminal proline-rich motif within host CD28's cytoplasmic tail, a motif previously shown to be required for development of regulatory T cells (Tregs) and function of conventional T cells. Adoptive transfer experiments demonstrated that a defect in host CD4(+)CD25(+) Tregs, but not in conventional T cells, was responsible for disease phenotype. Host Treg deficiency altered the cytokine pattern of donor CD4(+) T cells and the Ag specificity of autoantibodies, and these might lead to phenotypic change. Thus, host CD28 signaling controlled the pathogenesis of cGVHD through effects on host Tregs, whose status impacts qualitatively on the allogeneic immune responses.


Subject(s)
CD28 Antigens/deficiency , Graft vs Host Disease/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD28 Antigens/genetics , CD28 Antigens/immunology , Chronic Disease , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous
7.
J Immunol ; 195(2): 477-87, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26041540

ABSTRACT

A substantial proportion of CD8(+) T cells in adults lack the expression of the CD28 molecule, and the aging of the immune system is associated with a steady expansion of this T cell subset. CD28(-)CD8(+) T cells are characterized by potent effector functions but impaired responses to antigenic challenge. CD28 acts as the primary T cell costimulatory receptor, but there are numerous additional receptors that can costimulate the activation of T cells. In this study, we have examined such alternative costimulatory pathways regarding their functional role in CD28(-)CD8(+) T cells. Our study showed that most costimulatory molecules have a low capacity to activate CD28-deficient T cells, whereas the engagement of the CD2 molecule by its ligand CD58 clearly costimulated proliferation, cytokine production, and effector function in this T cell subset. CD58 is broadly expressed on APCs including dendritic cells. Blocking CD58 mAb greatly reduced the response of human CD28(-)CD8(+) T cells to allogeneic dendritic cells, as well as to viral Ags. Our results clearly identify the CD58/CD2 axis as the primary costimulatory pathway for CD8 T cells that lack CD28. Moreover, we show that engagement of CD2 amplifies TCR signals in CD28(-)CD8(+) T cells, demonstrating that the CD2-CD58 interaction has a genuine costimulatory effect on this T cell subset. CD2 signals might promote the control of viral infection by CD28(-)CD8(+) T cells, but they might also contribute to the continuous expansion of CD28(-)CD8(+) T cells during chronic stimulation by persistent Ag.


Subject(s)
CD2 Antigens/immunology , CD28 Antigens/immunology , CD58 Antigens/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Signal Transduction/immunology , Aged , Amino Acid Sequence , Antigens, Viral/chemistry , Antigens, Viral/immunology , Antigens, Viral/pharmacology , CD2 Antigens/genetics , CD28 Antigens/deficiency , CD28 Antigens/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD58 Antigens/genetics , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gene Expression Regulation , Humans , Immunophenotyping , Lymphocyte Activation/drug effects , Molecular Sequence Data , Orthomyxoviridae/immunology , Peptides/chemistry , Peptides/immunology , Peptides/pharmacology , Primary Cell Culture , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
8.
Circulation ; 131(8): 709-20, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25527700

ABSTRACT

BACKGROUND: The number of CD4(+)CD28(null) (CD28(null)) T cells, a unique subset of T lymphocytes with proinflammatory and cell-lytic phenotype, increases markedly in patients with acute coronary syndrome (ACS). ACS patients harboring high numbers of CD28(null) T cells have increased risk of recurrent severe acute coronary events and unfavorable prognosis. The mechanisms that govern the increase in CD28(null) T cells in ACS remain elusive. We investigated whether apoptosis pathways regulating T-cell homeostasis are perturbed in CD28(null) T cells in ACS. METHODS AND RESULTS: We found that CD28(null) T cells in ACS were resistant to apoptosis induction via Fas-ligation or ceramide. This was attributable to a dramatic reduction in proapoptotic molecules Bim, Bax, and Fas in CD28(null) T cells, whereas antiapoptotic molecules Bcl-2 and Bcl-xL were similar in CD28(null) and CD28(+) T cells. We also show that Bim is phosphorylated in CD28(null) T cells and degraded by the proteasome. Moreover, we demonstrate for the first time that proteasomal inhibition restores the apoptosis sensitivity of CD28(null) T cells in ACS. CONCLUSIONS: We show that CD28(null) T cells in ACS harbor marked defects in molecules that regulate T-cell apoptosis, which tips the balance in favor of antiapoptotic signals and endows these cells with resistance to apoptosis. We demonstrate that the inhibition of proteasomal activity allows CD28(null) T cells to regain sensitivity to apoptosis. A better understanding of the molecular switches that control the apoptosis sensitivity of CD28(null) T cells may reveal novel strategies for targeted elimination of these T cells in ACS patients.


Subject(s)
Acute Coronary Syndrome/metabolism , Acute Coronary Syndrome/pathology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , CD28 Antigens/deficiency , CD4-Positive T-Lymphocytes/pathology , Membrane Proteins/metabolism , Proteasome Endopeptidase Complex/physiology , Proto-Oncogene Proteins/metabolism , Acute Coronary Syndrome/epidemiology , Antibodies, Anti-Idiotypic/pharmacology , Apoptosis/drug effects , Bcl-2-Like Protein 11 , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/physiology , Cells, Cultured , Fas-Associated Death Domain Protein/metabolism , Homeostasis/physiology , Humans , MAP Kinase Signaling System/physiology , Phenotype , Recurrence , Risk Factors , bcl-2-Associated X Protein/metabolism
9.
Circ J ; 80(9): 1971-9, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27396441

ABSTRACT

BACKGROUND: Inflammatory responses, especially by CD4(+)T cells activated by dendritic cells, are known to be important in the pathophysiology of cardiac repair after myocardial infarction (MI). Although co-stimulatory signals through B7 (CD80/86) and CD28 are necessary for CD4(+)T cell activation and survival, the roles of these signals in cardiac repair after MI are still unclear. METHODS AND RESULTS: C57BL/6 (Control) mice and CD28 knockout (CD28KO) mice were subjected to left coronary artery permanent ligation. The ratio of death by cardiac rupture within 5 days after MI was significantly higher in CD28KO mice compared with Control mice. Although there were no significant differences in the infarct size between the 2 groups, left ventricular end-diastolic and end-systolic diameters were significantly increased, and fractional shortening was significantly decreased in CD28KO mice compared with Control mice. Electron microscopic observation revealed that the extent of extracellular collagen fiber was significantly decreased in CD28KO mice compared with Control mice. The number of α-smooth muscle actin-positive myofibroblasts was significantly decreased, and matrix metalloproteinase-9 activity and the mRNA expression of interleukin-1ß were significantly increased in CD28KO mice compared with Control mice. CONCLUSIONS: Deletion of CD28 co-stimulatory signals exacerbates left ventricular remodeling and increases cardiac rupture after MI through prolongation of the inflammatory period and reduction of collagen fiber in the infarct scars. (Circ J 2016; 80: 1971-1979).


Subject(s)
CD28 Antigens/deficiency , Gene Deletion , Heart Rupture, Post-Infarction/metabolism , Myocardial Infarction/metabolism , Signal Transduction , Ventricular Remodeling , Animals , CD28 Antigens/metabolism , Gene Expression Regulation , Heart Rupture, Post-Infarction/genetics , Heart Rupture, Post-Infarction/pathology , Heart Rupture, Post-Infarction/physiopathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure
10.
J Immunol ; 193(3): 1223-32, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24951814

ABSTRACT

Adenoviral vectors have long been forerunners in the development of effective CD8 T cell-based vaccines; therefore, it is imperative that we understand the factors controlling the induction of robust and long-lasting transgene-specific immune responses by these vectors. In this study, we investigated the organ sites, molecules, and cell subsets that play a critical role in the priming of transgene-specific CD8 T cells after vaccination with a replication-deficient adenoviral vector. Using a human adenovirus serotype 5 (Ad5) vector and genetically engineered mice, we found that CD8(+) and/or CD103(+) dendritic cells in the draining lymph node played a critical role in the priming of Ad5-induced CD8 T cell responses. Moreover, we found that CD80/86, but not CD28, was essential for efficient generation of both primary effectors and memory CD8 T cells. Interestingly, the lack of CD28 expression resulted in a delayed primary response, whereas memory CD8 T cells generated in CD28-deficient mice appeared almost normal in terms of both phenotype and effector cytokine profile, but they exhibited a significantly reduced proliferative capacity upon secondary challenge while retaining immediate in vivo effector capabilities: in vivo cytotoxicity and short-term in vivo protective capacity. Overall, our data point to an absolute requirement for professional APCs and the expression of the costimulatory molecules CD80/86 for efficient CD8 T cell priming by adenoviral vectors. Additionally, our results suggest the existence of an alternative receptor for CD80/86, which may substitute, in part, for CD28.


Subject(s)
Adenovirus Infections, Human/prevention & control , Adenoviruses, Human/immunology , B7-1 Antigen/deficiency , B7-2 Antigen/deficiency , CD28 Antigens/deficiency , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Genetic Engineering , Adenovirus Infections, Human/genetics , Adenoviruses, Human/genetics , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , B7-1 Antigen/genetics , B7-2 Antigen/genetics , CD28 Antigens/genetics , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Dendritic Cells/metabolism , Dendritic Cells/virology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Ligands , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccination
11.
J Infect Dis ; 211(5): 835-45, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25240171

ABSTRACT

We postulated that the activation of proinflammatory signaling by methicillin-resistant Staphylococcus aureus (MRSA) strain USA300 is a major factor in the pathogenesis of severe pneumonia and a target for immunomodulation. Local activation of T cells in the lung was a conserved feature of multiple strains of S. aureus, in addition to USA300. The pattern of Vß chain activation was consistent with known superantigens, but deletion of SelX or SEK and SEQ was not sufficient to prevent T-cell activation, indicating the participation of multiple genes. Using Rag2(-/-), Cd4(-/-), and Cd28(-/-) mice, we observed significantly improved clearance of MRSA from the airways and decreased lung pathology, compared with findings for wild-type controls. The improved outcome correlated with decreased production of proinflammatory cytokines (tumor necrosis factor, KC, interleukin 6, and interleukin 1ß). Our data suggest that T-cell-mediated hypercytokinemia induced by infection with MRSA strain USA300 contributes to pathogenesis and may be a therapeutic target for improving outcomes of this common infection in a clinical setting.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Cytokines/metabolism , Methicillin-Resistant Staphylococcus aureus/immunology , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/pathology , Animals , CD28 Antigens/deficiency , CD4 Antigens/genetics , Cytokines/blood , DNA-Binding Proteins/deficiency , Disease Models, Animal , Gene Deletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Superantigens/genetics , Superantigens/immunology
12.
Immunology ; 146(2): 185-93, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26190355

ABSTRACT

Inflammation contributes to the development and perpetuation of several disorders and T lymphocytes orchestrate the inflammatory immune response. Although the role of T cells in inflammation is widely recognized, specific therapies that tackle inflammatory networks in disease are yet to be developed. CD4(+) CD28(null) T cells are a unique subset of helper T lymphocytes that recently shot back into the limelight as potential catalysts of inflammation in several inflammatory disorders such as autoimmunity, atherosclerosis and chronic viral infections. In contrast to conventional helper T cells, CD4(+) CD28(null) T cells have an inbuilt ability to release inflammatory cytokines and cytotoxic molecules that can damage tissues and amplify inflammatory pathways. It comes as no surprise that patients who have high numbers of these cells have more severe disease and poor prognosis. In this review, I provide an overview on the latest advances in the biology of CD4(+) CD28(null) T cells. Understanding the complex functions and dynamics of CD4(+) CD28(null) T cells may open new avenues for therapeutic intervention to prevent progression of inflammatory diseases.


Subject(s)
CD28 Antigens/immunology , Cytotoxicity, Immunologic , Inflammation/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , CD28 Antigens/deficiency , Cell Death , Cell Proliferation , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lymphocyte Activation , Phenotype , Prognosis , Severity of Illness Index , Signal Transduction , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology
13.
Immunology ; 144(4): 598-610, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25318356

ABSTRACT

NOD.H-2h4 mice develop spontaneous autoimmune thyroiditis (SAT) with chronic inflammation of thyroids by T and B cells. B-cell deficient (B(-/-) ) mice are resistant to SAT but develop SAT if regulatory T (Treg) cells are transiently depleted. We established a transfer model using splenocytes from CD28(-/-)  B(-/-) mice (effector cells and antigen-presenting cells) cultured with or without sorted Treg cells from Foxp3-GFP wild-type (WT) or B(-/-) mice. After transfer to mice lacking T cells, mice given Treg cells from B(-/-) mice had significantly lower SAT severity scores than mice given Treg cells from WT mice, indicating that Treg cells in B(-/-) mice are more effective suppressors of SAT than Treg cells in WT mice. Treg cells from B(-/-) mice differ from WT Treg cells in expression of CD27, tumour necrosis factor receptor (TNFR) II p75, and glucocorticoid-induced TNFR-related protein (GITR). After transient depletion using anti-CD25 or diphtheria toxin, the repopulating Treg cells in B(-/-) mice lack suppressor function, and expression of CD27, GITR and p75 is like that of WT Treg cells. If B(-/-) Treg cells develop with B cells in bone marrow chimeras, their phenotype is like that of WT Treg cells. Addition of B cells to cultures of B(-/-) Treg and T effector cells abrogates their suppressive function and their phenotype is like that of WT Treg cells. These results establish for the first time that Treg cells in WT and B(-/-) mice differ both functionally and in expression of particular cell surface markers. Both properties are altered after transient depletion and repopulation of B(-/-) Treg cells, and by the presence of B cells during Treg cell development or during interaction with effector T cells.


Subject(s)
B-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Thyroiditis, Autoimmune/immunology , Adoptive Transfer , Animals , B-Lymphocytes/metabolism , Biomarkers/metabolism , Bone Marrow Transplantation , CD28 Antigens/deficiency , CD28 Antigens/genetics , Cell Communication , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Genes, T-Cell Receptor alpha , Genetic Predisposition to Disease , Leukocyte Common Antigens/genetics , Mice, Inbred NOD , Mice, Knockout , Phenotype , Severity of Illness Index , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Thyroiditis, Autoimmune/genetics , Thyroiditis, Autoimmune/metabolism , Thyroiditis, Autoimmune/prevention & control , Transplantation Chimera
14.
Gastroenterology ; 147(1): 221-232.e7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24726754

ABSTRACT

BACKGROUND & AIMS: T-cell-mediated biliary injury is a feature of primary sclerosing cholangitis (PSC). We studied the roles of CD28(-) T cells in PSC and their regulation by vitamin D. METHODS: Peripheral and liver-infiltrating mononuclear cells were isolated from blood or fresh liver tissue. We analyzed numbers, phenotypes, functions, and localization patterns of CD28(-) T cells, along with their ability to activate biliary epithelial cells. We measured levels of tumor necrosis factor (TNF)α in liver tissues from patients with PSC and the effects of exposure to active vitamin D (1,25[OH]2D3) on expression of CD28. RESULTS: A significantly greater proportion of CD4(+) and CD8(+) T cells that infiltrated liver tissues of patients with PSC were CD28(-), compared with control liver tissue (CD4(+): 30.3% vs 2.5%, P < .0001; and CD8(+): 68.5% vs 31.9%, P < .05). The mean percentage of CD4(+)CD28(-) T cells in liver tissues from patients with PSC was significantly higher than from patients with primary biliary cirrhosis or nonalcoholic steatohepatitis (P < .05). CD28(-) T cells were activated CD69(+)CD45RA(-) C-C chemokine receptor (CCR)7(-) effector memory and perforin(+) granzyme B(+) cytotoxic cells, which express CD11a, CX3CR1, C-X3-C motif receptor 6 (CXCR6), and CCR10-consistent with their infiltration of liver and localization around bile ducts. Compared with CD28(+) T cells, activated CD28(-) T cells produced significantly higher levels of interferon γ and TNFα (P < .05), and induced up-regulation of intercellular cell adhesion molecule-1, HLA-DR, and CD40 by primary epithelial cells (3.6-fold, 1.5-fold, and 1.2-fold, respectively). Liver tissue from patients with PSC contained high levels of TNFα; TNFα down-regulated the expression of CD28 by T cells in vitro (P < .01); this effect was prevented by administration of 1,25(OH)2D3 (P < .05). CONCLUSIONS: Inflammatory CD28(-) T cells accumulate in livers of patients with PSC and localize around bile ducts. The TNFα-rich microenvironment of this tissue promotes inflammation; these effects are reversed by vitamin D in vitro.


Subject(s)
CD28 Antigens/deficiency , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cholangitis, Sclerosing/etiology , Liver/metabolism , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/drug effects , Cells, Cultured , Cholangitis, Sclerosing/metabolism , Cholangitis, Sclerosing/pathology , Cytokines/metabolism , Humans , In Vitro Techniques , Interferon-gamma/metabolism , Liver/pathology , Tumor Necrosis Factor-alpha/metabolism , Vitamin D/pharmacology
15.
Blood ; 121(1): 85-94, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23144170

ABSTRACT

Immunoglobulin class switching from IgM to IgG in response to peptides is generally T cell-dependent and vaccination in T cell-deficient individuals is inefficient. We show that a vaccine consisting of a dense array of peptides on liposomes induced peptide-specific IgG responses totally independent of T-cell help. Independency was confirmed in mice lacking T cells and in mice deficient for MHC class II, CD40L, and CD28. The IgG titers were high, long-lived, and comparable with titers obtained in wild-type animals, and the antibody response was associated with germinal center formation, expression of activation-induced cytidine deaminase, and affinity maturation. The T cell-independent (TI) IgG response was strictly dependent on ligation of TLR4 receptors on B cells, and concomitant TLR4 and cognate B-cell receptor stimulation was required on a single-cell level. Surprisingly, the IgG class switch was mediated by TIR-domain-containing adapter inducing interferon-ß (TRIF), but not by MyD88. This study demonstrates that peptides can induce TI isotype switching when antigen and TLR ligand are assembled and appropriately presented directly to B lymphocytes. A TI vaccine could enable efficient prophylactic and therapeutic vaccination of patients with T-cell deficiencies and find application in diseases where induction of T-cell responses contraindicates vaccination, for example, in Alzheimer disease.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Amyloid beta-Peptides/immunology , B-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , Peptide Fragments/immunology , Toll-Like Receptor 4/physiology , Vaccines, Subunit/immunology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Adoptive Transfer , Amino Acid Sequence , Amyloid beta-Peptides/administration & dosage , Animals , Antigen Presentation , B-Lymphocytes/metabolism , CD28 Antigens/deficiency , CD28 Antigens/immunology , CD40 Ligand/deficiency , CD40 Ligand/immunology , Germinal Center/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Lipopolysaccharide Receptors/immunology , Liposomes , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Molecular Sequence Data , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Vaccination , Vaccines, Subunit/administration & dosage
16.
J Immunol ; 191(10): 4940-9, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24098053

ABSTRACT

NOD.H-2h4 mice given NaI in their drinking water develop iodine-accelerated spontaneous autoimmune thyroiditis (ISAT) with chronic inflammation of the thyroid by T and B cells and production of anti-mouse thyroglobulin (MTg) autoantibody. CD28(-/-) NOD.H-2h4 mice, which have reduced numbers of CD4(+)Foxp3(+) regulatory T cells (Tregs), were developed to examine the role of Tregs in ISAT development. CD28(-/-) NOD.H2-h4 mice develop more severe ISAT than do wild-type (WT) mice, with collagen deposition (fibrosis) and low serum T4. CD28(-/-) mice have increased expression of proinflammatory cytokines IFN-γ and IL-6, consistent with increased mononuclear cell infiltration and tissue destruction in thyroids. Importantly, transferring purified CD4(+)Foxp3(+) Tregs from WT mice reduces ISAT severity in CD28(-/-) mice without increasing the total number of Tregs, suggesting that endogenous Tregs in CD28(-/-) mice are functionally ineffective. Endogenous CD28(-/-) Tregs have reduced surface expression of CD27, TNFR2 p75, and glucocorticoid-induced TNFR-related protein compared with transferred CD28(+/+) Tregs. Although anti-MTg autoantibody levels generally correlate with ISAT severity scores in WT mice, CD28(-/-) mice have lower anti-MTg autoantibody responses than do WT mice. The percentages of follicular B cells are decreased and those of marginal zone B cells are increased in spleens of CD28(-/-) mice, and they have fewer thyroid-infiltrating B cells than do WT mice. This suggests that CD28 deficiency has direct and indirect effects on the B cell compartment. B cell-deficient (B(-/-)) NOD.H-2h4 mice are resistant to ISAT, but CD28(-/-)B(-/-) mice develop ISAT comparable to WT mice and have reduced numbers of Tregs compared with WT B(-/-) mice.


Subject(s)
B-Lymphocytes/immunology , CD28 Antigens/deficiency , T-Lymphocytes, Regulatory/immunology , Thyroiditis, Autoimmune/immunology , Animals , Autoantibodies/immunology , CD4 Antigens/metabolism , Fibrosis/immunology , Forkhead Transcription Factors/metabolism , Inflammation , Interferon-gamma/biosynthesis , Interleukin-6/biosynthesis , Lymphocyte Count , Mice , Mice, Inbred NOD , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type II/metabolism , Sodium Iodide/administration & dosage , Thyroglobulin/immunology , Thyroid Gland/immunology , Thyroiditis, Autoimmune/chemically induced , Thyroxine/blood , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
17.
Arterioscler Thromb Vasc Biol ; 33(10): 2374-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23908246

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm is an inflammatory disease leading to destructive vascular remodeling and ultimately to lethal aortic rupture. Despite its frequent association with atherosclerosis, compelling studies have shown striking differences and potentially opposite roles of T-cell helper responses in aneurysm as compared with atherosclerosis, casting doubt on the relevance and suitability of T-cell-targeted therapies in this context. APPROACH AND RESULTS: Here, we show that selective depletion of T regulatory (Treg) cells using a CD25-specific monoclonal antibody significantly enhances the susceptibility of C57Bl/6 mice to angiotensin II-induced abdominal aortic aneurysm and promotes aortic rupture (n=25-44 mice/group). Similar results are observed in angiotensin II-treated Cd80(-/-)/Cd86(-/-) or Cd28(-/-) mice with impaired Treg cell homeostasis (n=18-23 mice/group). Treg cell depletion is associated with increased immune cell activation and a blunted interleukin (IL)-10 anti-inflammatory response, suggesting an immunoinflammatory imbalance. Interestingly, Il-10(-/-) mice (n=20 mice/group) show increased susceptibility to angiotensin II-induced abdominal aortic aneurysm and aortic rupture and are insensitive to Treg cell depletion. Finally, reconstitution of Cd28(-/-) Treg-deficient mice with Treg cells (n=22 mice/group) restores a balance in the immunoinflammatory response, rescues the animals from increased susceptibility to aneurysm, and prevents aortic dissection. CONCLUSIONS: These results identify a critical role for Treg cells and IL-10 in the control of aneurysm formation and its progression to rupture and suggest that therapies targeting Treg responses may be most suited to treat aneurysmal disease.


Subject(s)
Angiotensin II , Aorta, Abdominal/immunology , Aortic Aneurysm, Abdominal/prevention & control , Aortic Rupture/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Aortic Rupture/chemically induced , Aortic Rupture/immunology , Aortic Rupture/pathology , B7-1 Antigen/deficiency , B7-1 Antigen/genetics , B7-2 Antigen/deficiency , B7-2 Antigen/genetics , CD28 Antigens/deficiency , CD28 Antigens/genetics , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Reduction Procedures , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/transplantation , Time Factors
18.
J Immunol ; 189(6): 2758-67, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22908331

ABSTRACT

The interaction of CD28, which is constitutively expressed on T cells, with B7.1/B7.2 expressed on APCs is critical for T cell activation. CD28 is also expressed on murine and human plasma cells but its function on these cells remains unclear. There are two types of plasma cells: short-lived ones that appear in the secondary lymphoid tissue shortly after Ag exposure, and long-lived plasma cells that mainly reside in the bone marrow. We demonstrate that CD28-deficient murine short- and long-lived plasma cells produce significantly higher levels of Abs than do their wild-type counterparts. This was owing to both increased frequencies of plasma cells as well as increased Ab production per plasma cell. Plasma cells also express the ligand for CD28, B7.1, and B7.2. Surprisingly, deficiency of B7.1 and B7.2 in B cells also led to higher Ab levels, analogous to Cd28(-/-) plasma cells. Collectively, our results suggest that the CD28-B7 interaction operates as a key modulator of plasma cell function.


Subject(s)
B7 Antigens/physiology , CD28 Antigens/physiology , Cell Survival/immunology , Cellular Senescence/immunology , Plasma Cells/cytology , Plasma Cells/immunology , Animals , B7 Antigens/deficiency , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD28 Antigens/biosynthesis , CD28 Antigens/deficiency , Cell Line , Female , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plasma Cells/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Time Factors
19.
BMC Genomics ; 14: 744, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24176122

ABSTRACT

BACKGROUND: In the model eukaryote, Saccharomyces cerevisiae, previous experiments have identified those genes that exert the most significant control over cell growth rate. These genes are termed HFC for high flux control. Such genes are overrepresented within pathways controlling the mitotic cell cycle. RESULTS: We postulated that the increase/decrease in growth rate is due to a change in the rate of progression through specific cell cycle steps. We extended and further developed an existing logical model of the yeast cell cycle in order elucidate how the HFC genes modulated progress through the cycle. This model can simulate gene dosage-variation and calculate the cycle time, determine the order and relative speed at which events occur, and predict arrests and failures to correctly execute a step. To experimentally test our model's predictions, we constructed a tetraploid series of deletion mutants for a set of eight genes that control the G2/M transition. This system allowed us to vary gene copy number through more intermediate levels than previous studies and examine the impact of copy-number variation on growth, cell-cycle phenotype, and response to different cellular stresses. CONCLUSIONS: For the majority of strains, the predictions agreed with experimental observations, validating our model and its use for further predictions. Where simulation and experiment diverged, we uncovered both novel tetraploid-specific phenotypes and a switch in the determinative execution point of a key cell-cycle regulator, the Cdc28 kinase, from the G1/S to the S/G2 boundaries.


Subject(s)
DNA Copy Number Variations/genetics , Saccharomyces cerevisiae/genetics , CD28 Antigens/deficiency , CD28 Antigens/genetics , CD28 Antigens/metabolism , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division , Cyclin B/deficiency , Cyclin B/genetics , Cyclin B/metabolism , G2 Phase , Mitogen-Activated Protein Kinases/deficiency , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Models, Theoretical , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Tetraploidy
20.
J Mol Cell Cardiol ; 52(5): 1038-47, 2012 May.
Article in English | MEDLINE | ID: mdl-22285481

ABSTRACT

We and others have previously reported that granulocyte colony-stimulating factor (G-CSF) prevents left ventricular remodeling and dysfunction after myocardial infarction in animal models and human. We have also reported that G-CSF inhibits the progression of atherosclerosis in animal models, but its precise mechanism is still elusive. So, we examined the effects of G-CSF on atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice. Twelve-week-old male ApoE(-/-) mice were subcutaneously administrated with 200 µg/kg of G-CSF or saline once a day for 5 consecutive days per a week for 4 weeks. Atherosclerotic lesion of aortic sinus was significantly reduced in the G-CSF-treated mice compared with the saline-treated mice (35% reduction, P<0.05). G-CSF significantly reduced the expression level of interferon-γ by 31% and increased the expression level of interleukin-10 by 20% in atherosclerotic lesions of aortic sinus. G-CSF increased the number of CD4(+)CD25(+) regulatory T cells in lymph nodes and spleen, and enhanced the suppressive function of regulatory T cells in vitro. G-CSF markedly increased the number of Foxp3-positive regulatory T cells in atherosclerotic lesions of aortic sinus. Administration of anti-CD25 antibody (PC61) that depletes regulatory T cells abrogated these atheroprotective effects of G-CSF. Moreover, in ApoE(-/-)/CD28(-/-) mice, that lack regulatory T cells, the protective effects of G-CSF on atherosclerosis were not recognized. These findings suggest that regulatory T cells play an important role in the atheroprotective effects of G-CSF.


Subject(s)
Atherosclerosis/prevention & control , Granulocyte Colony-Stimulating Factor/pharmacology , T-Lymphocytes, Regulatory/physiology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/pathology , CD28 Antigens/deficiency , CD28 Antigens/genetics , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Granulocyte Colony-Stimulating Factor/therapeutic use , Lipids/blood , Lymph Nodes/pathology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sinus of Valsalva/metabolism , Sinus of Valsalva/pathology , Spleen/pathology , T-Lymphocytes, Regulatory/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL