Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Bioanalysis ; : 1-13, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864397

ABSTRACT

The 16th GCC Closed Forum was held in Orlando, FL, USA, on 23 June 2023. Representatives from international bioanalytical Contract Research Organizations were in attendance in order to discuss scientific and regulatory issues specific to bioanalysis. The issues discussed at the meeting included: IS response, flow cytometry, changes to the bioanalytical industry, NGS assays, biomarker assay for tissues, dPCR validation, immunogenicity harmonization and ICH M10 implementation. Conclusions and consensus from discussions of these topics are included in this article.

2.
J Allergy Clin Immunol ; 143(6): 2215-2226.e7, 2019 06.
Article in English | MEDLINE | ID: mdl-30578871

ABSTRACT

BACKGROUND: Inflammatory activation of CD8+ T cells can, when left unchecked, drive severe immunopathology. Hyperstimulation of CD8+ T cells through a broad set of triggering signals can precipitate hemophagocytic lymphohistiocytosis (HLH), a life-threatening systemic inflammatory disorder. OBJECTIVE: The mechanism linking CD8+ T-cell hyperactivation to pathology is controversial, with excessive production of IFN-γ and, more recently, excessive consumption of IL-2, which are proposed as competing hypotheses. We formally tested the proximal mechanistic events of each pathway in a mouse model of HLH. METHODS: In addition to reporting a complete autosomal recessive IFN-γ receptor 1-deficient patient with multiple aspects of HLH pathology, we used the mouse model of perforin (Prf1)KO mice infected with lymphocytic choriomeningitis virus to genetically eliminate either IFN-γ production or CD25 expression and assess the immunologic, hematologic, and physiologic disease measurement. RESULTS: We found a striking dichotomy between the mechanistic basis of the hematologic and inflammatory components of CD8+ T cell-mediated pathology. The hematologic features of HLH were completely dependent on IFN-γ production, with complete correction after loss of IFN-γ production without any role for CD8+ T cell-mediated IL-2 consumption. By contrast, the mechanistic contribution of the immunologic features was reversed, with no role for IFN-γ production but substantial correction after reduction of IL-2 consumption by hyperactivated CD8+ T cells. These results were complemented by the characterization of an IFN-γ receptor 1-deficient patients with HLH-like disease, in whom multiple aspects of HLH pathology were observed in the absence of IFN-γ signaling. CONCLUSION: These results synthesize the competing mechanistic models of HLH pathology into a dichotomous pathogenesis driven through discrete pathways. A holistic model provides a new paradigm for understanding HLH and, more broadly, the consequences of CD8+ T-cell hyperactivation, thereby paving the way for clinical intervention based on the features of HLH in individual patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , Interferon-gamma/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Lymphohistiocytosis, Hemophagocytic/immunology , Receptors, Interferon/deficiency , Animals , Consanguinity , Disease Models, Animal , Female , Humans , Infant , Lymphocyte Activation , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/genetics , Mice , Mice, Knockout , Models, Immunological , Morocco , Perforin/genetics , Signal Transduction , Interferon gamma Receptor
3.
Front Immunol ; 9: 1989, 2018.
Article in English | MEDLINE | ID: mdl-30319599

ABSTRACT

Mammalian genomes encode a plethora of long non-coding RNA (lncRNA). These transcripts are thought to regulate gene expression, influencing biological processes from development to pathology. Results from the few lncRNA that have been studied in the context of the immune system have highlighted potentially critical functions as network regulators. Here we explored the nature of the lncRNA transcriptome in regulatory T cells (Tregs), a subset of CD4+ T cells required to establish and maintain immunological self-tolerance. The identified Treg lncRNA transcriptome showed distinct differences from that of non-regulatory CD4+ T cells, with evidence of direct shaping of the lncRNA transcriptome by Foxp3, the master transcription factor driving the distinct mRNA profile of Tregs. Treg lncRNA changes were disproportionally reversed in the absence of Foxp3, with an enrichment for colocalisation with Foxp3 DNA binding sites, indicating a direct coordination of transcription by Foxp3 independent of the mRNA coordination function. We further identified a novel lncRNA Flatr, as a member of the core Treg lncRNA transcriptome. Flatr expression anticipates Foxp3 expression during in vitro Treg conversion, and Flatr-deficient mice show a mild delay in in vitro and peripheral Treg induction. These results implicate Flatr as part of the upstream cascade leading to Treg conversion, and may provide clues as to the nature of this process.


Subject(s)
Forkhead Transcription Factors/metabolism , RNA, Long Noncoding/metabolism , Self Tolerance/genetics , T-Lymphocytes, Regulatory/immunology , Animals , CRISPR-Cas Systems/genetics , Gene Expression Profiling , Gene Expression Regulation , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Long Noncoding/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism
4.
Immunity ; 45(5): 960-962, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27851924

ABSTRACT

Variation in protein expression is a feature of all cell populations. Using T cell subsets as a proof-of-concept, Lu et al. (2016) develop a framework for dissecting out the contributors to this cell-to-cell expression variation from high-parameter flow cytometry studies.


Subject(s)
Flow Cytometry , T-Lymphocyte Subsets
5.
Neurol Neuroimmunol Neuroinflamm ; 3(4): e240, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27231713

ABSTRACT

OBJECTIVE: We undertook a systems immunology approach of the adaptive immune system in multiple sclerosis (MS), overcoming tradeoffs between scale and level of detail, in order to identify the immunologic signature of MS and the changes wrought by current immunomodulatory treatments. METHODS: We developed a comprehensive flow cytometry platform measuring 38 immunologic cell types in the peripheral blood of 245 individuals in a routine clinical setting. These include patients with MS, untreated or receiving any of 4 current immunomodulatory treatments (interferon-ß, glatiramer acetate, natalizumab, or fingolimod), patients with autoimmune thyroid disease, and healthy controls. RESULTS: An increase in memory CD8(+) T cells and B cells was observed in untreated patients with MS. Interferon-ß and fingolimod induce significant changes upon multiple aspects of the peripheral immune system, with an unexpectedly prominent alteration of B cells. Overall, both treatments push the immune system in different directions, with only 2 significant effects shared across these treatments-an increase in transitional B cells and a decrease in class-switched B cells. We further identified heightened B cell-activating factor (BAFF) levels as regulating this shared B cell pathway. CONCLUSIONS: A systems immunology approach established different immunologic profiles induced by current immunomodulatory MS treatments, offering perspectives for personalized medicine. Pathways shared between the immunologic architecture of existing efficacious treatments identify targets for future treatment design.

6.
Nat Genet ; 48(5): 519-27, 2016 05.
Article in English | MEDLINE | ID: mdl-26998692

ABSTRACT

Type 1 (T1D) and type 2 (T2D) diabetes share pathophysiological characteristics, yet mechanistic links have remained elusive. T1D results from autoimmune destruction of pancreatic beta cells, whereas beta cell failure in T2D is delayed and progressive. Here we find a new genetic component of diabetes susceptibility in T1D non-obese diabetic (NOD) mice, identifying immune-independent beta cell fragility. Genetic variation in Xrcc4 and Glis3 alters the response of NOD beta cells to unfolded protein stress, enhancing the apoptotic and senescent fates. The same transcriptional relationships were observed in human islets, demonstrating the role of beta cell fragility in genetic predisposition to diabetes.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Insulin-Secreting Cells/pathology , Animals , Apoptosis , Cellular Senescence , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Diet , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Protein Folding , Repressor Proteins/genetics , Sex Factors , Stress, Physiological , Trans-Activators/genetics
7.
J Allergy Clin Immunol ; 138(1): 200-209.e8, 2016 07.
Article in English | MEDLINE | ID: mdl-26947179

ABSTRACT

BACKGROUND: Hemophagocytic lymphohistiocytosis (HLH) is a severe inflammatory condition driven by excessive CD8(+) T-cell activation. HLH occurs as both acquired and familial hemophagocytic lymphohistiocytosis (FHL) forms. In both conditions, a sterile or infectious trigger is required for disease initiation, which then becomes self-sustaining and life-threatening. Recent studies have attributed the key distal event to excessive IFN-γ production; however, the proximal events driving immune dysregulation have remained undefined. OBJECTIVE: We sought to investigate the role of regulatory T (Treg) cells in the pathophysiology of experimental FHL. METHODS: Because mutation in perforin is a common cause of FHL, we used an experimental FHL mouse model in which disease in perforin-deficient mice is triggered by lymphocytic choriomeningitis virus (LCMV). We assessed Treg and CD8(+) T-cell homeostasis and activation during the changing systemic conditions in the mice. In addition, human blood samples were collected and analyzed during the HLH episode. RESULTS: We found no primary Treg cell defects in perforin-deficient mice. However, Treg cell numbers collapsed after LCMV inoculation. The collapse of Treg cell numbers in LCMV-triggered perforin-deficient, but not wild-type, mice was accompanied by the combination of lower IL-2 secretion by conventional CD4(+) T cells, increased IL-2 consumption by activated CD8(+) T cells, and secretion of competitive soluble CD25. Moreover low Treg cell numbers were observed in untreated patients experiencing HLH flares. CONCLUSION: These results demonstrate that excessive CD8(+) T-cell activation rewires the IL-2 homeostatic network away from Treg cell maintenance and toward feed-forward inflammation. These results also provide a potential mechanistic pathway for the progression of infectious inflammation to persistent inflammation in patients with HLH.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Interleukin-2/metabolism , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Disease Models, Animal , Homeostasis , Humans , Immunomodulation , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/physiology , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/genetics , Mice , Mice, Knockout , Perforin/genetics
8.
Immunology ; 146(1): 122-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26059465

ABSTRACT

Leptin is an adipokine that regulates metabolism and plays an important role as a neuroendocrine hormone. Leptin mediates these functions via the leptin receptor, and deficiency in either leptin or its receptor leads to obesity in humans and mice. Leptin has far reaching effects on the immune system, as observed in obese mice, which display decreased thymic function and increased inflammatory responses. With expression of the leptin receptor on T cells and supporting thymic epithelium, aberrant signalling through the leptin receptor has been thought to be the direct cause of thymic involution in obese mice. Here, we demonstrate that the absence of leptin receptor on either thymic epithelial cells or T cells does not lead to the loss of thymic function, demonstrating that the thymoprotective effect of leptin is mediated by obesity suppression rather than direct signalling to the cellular components of the thymus.


Subject(s)
Leptin/metabolism , Obesity/metabolism , Receptors, Leptin/genetics , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Adipogenesis/genetics , Animals , Cell Differentiation/genetics , Epithelial Cells/metabolism , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Receptors, Leptin/biosynthesis , Receptors, Leptin/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
9.
Eur J Immunol ; 45(5): 1535-47, 2015 May.
Article in English | MEDLINE | ID: mdl-25627671

ABSTRACT

The thymus is the organ devoted to T-cell production. The thymus undergoes multiple rounds of atrophy and redevelopment before degenerating with age in a process known as involution. This process is poorly understood, despite the influence the phenomenon has on peripheral T-cell numbers. Here we have investigated the FVB/N mouse strain, which displays premature thymic involution. We find multiple architectural and cellular features that precede thymic involution, including disruption of the epithelial-endothelial relationship and a progressive loss of pro-T cells. The architectural features, reminiscent of the human thymus, are intrinsic to the nonhematopoietic compartment and are neither necessary nor sufficient for thymic involution. By contrast, the loss of pro-T cells is intrinsic to the hematopoietic compartment, and is sufficient to drive premature involution. These results identify pro-T-cell loss as the main driver of premature thymic involution, and highlight the plasticity of the thymic stroma, capable of maintaining function across diverse interstrain architectures.


Subject(s)
Thymus Gland/immunology , Thymus Gland/pathology , Aging/immunology , Aging/pathology , Animals , Atrophy/immunology , Atrophy/pathology , Cell Differentiation/immunology , Endothelium, Vascular/pathology , Epithelial Cells/pathology , Female , Humans , Infant , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Species Specificity , Stromal Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thymus Gland/blood supply
10.
Immunol Cell Biol ; 93(4): 417-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25533288

ABSTRACT

Costimulatory signals by CD28 are critical for thymic regulatory T-cell (Treg) development. To determine the functional relevance of CD28 for peripheral Treg post thymic selection, we crossed the widely used Forkhead box protein 3 (Foxp3)-CreYFP mice to mice bearing a conditional Cd28 allele. Treg-specific CD28 deficiency provoked a severe autoimmune syndrome as a result of a strong disadvantage in competitive fitness and proliferation of CD28-deficient Tregs. By contrast, Treg survival and lineage integrity were not affected by the lack of CD28. This data demonstrate that, even after the initial induction requirement, Treg maintain a higher dependency on CD28 signalling than conventional T cells for homeostasis. In addition, we found the Foxp3-CreYFP allele to be a hypomorph, with reduced Foxp3 protein levels. Furthermore, we report here the stochastic activity of the Foxp3-CreYFP allele in non-Tregs, sufficient to recombine some conditional alleles (including Cd28) but not others (including R26-RFP). This hypomorphism and 'leaky' expression of the Foxp3-CreYFP allele should be considered when analysing the conditionally mutated Treg.


Subject(s)
CD28 Antigens/metabolism , Forkhead Transcription Factors/metabolism , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Autoimmunity/genetics , CD28 Antigens/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Survival/genetics , Clonal Selection, Antigen-Mediated/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Homeostasis , Mice , Mice, Transgenic , Signal Transduction/genetics
11.
Immunology ; 141(3): 377-87, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24164480

ABSTRACT

Loss of ζ-associated protein 70 (Zap70) results in severe immunodeficiency in humans and mice because of the critical role of Zap70 in T-cell receptor (TCR) signalling. Here we describe a novel mouse strain generated by N-ethyl-N-nitrosourea mutagenesis, with the reduced protein stability (rps) mutation in Zap70. The A243V rps mutation resulted in decreased Zap70 protein and a reduced duration of TCR-induced calcium responses, equivalent to that induced by a 50% decrease in catalytically active Zap70. The reduction of signalling through Zap70 was insufficient to substantially perturb thymic differentiation of conventional CD4 and CD8 T cells, although Foxp3(+) regulatory T cells demonstrated altered thymic production and peripheral homeostasis. Despite the mild phenotype, the Zap70(A243V) variant lies just above the functional threshold for TCR signalling competence, as T cells relying on only a single copy of the Zap70(rps) allele for TCR signalling demonstrated no intracellular calcium response to TCR stimulation. This addition to the Zap70 allelic series indicates that a rate-limiting threshold for Zap70 protein levels exists at which signalling capacity switches from nearly intact to effectively null.


Subject(s)
Calcium Signaling , Mutation , Receptors, Antigen, T-Cell/metabolism , Severe Combined Immunodeficiency/enzymology , T-Lymphocyte Subsets/enzymology , ZAP-70 Protein-Tyrosine Kinase/deficiency , Amino Acid Sequence , Animals , Cell Differentiation , Disease Models, Animal , Female , Forkhead Transcription Factors/metabolism , Genetic Predisposition to Disease , Heterozygote , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Phenotype , Protein Stability , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Spleen/enzymology , Spleen/immunology , T-Lymphocyte Subsets/immunology , Thymocytes/enzymology , Thymocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism
12.
Nat Immunol ; 14(9): 959-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23852275

ABSTRACT

Foxp3⁺ regulatory T (Treg) cells are a crucial immunosuppressive population of CD4⁺ T cells, yet the homeostatic processes and survival programs that maintain the Treg cell pool are poorly understood. Here we report that peripheral Treg cells markedly alter their proliferative and apoptotic rates to rapidly restore numerical deficit through an interleukin 2-dependent and costimulation-dependent process. By contrast, excess Treg cells are removed by attrition, dependent on the Bim-initiated Bak- and Bax-dependent intrinsic apoptotic pathway. The antiapoptotic proteins Bcl-xL and Bcl-2 were dispensable for survival of Treg cells, whereas Mcl-1 was critical for survival of Treg cells, and the loss of this antiapoptotic protein caused fatal autoimmunity. Together, these data define the active processes by which Treg cells maintain homeostasis via critical survival pathways.


Subject(s)
Apoptosis/immunology , Forkhead Transcription Factors/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Apoptosis/genetics , Cell Survival/genetics , Cell Survival/immunology , Female , Forkhead Transcription Factors/genetics , Gene Deletion , Homeostasis/immunology , Interleukin-2/metabolism , Lymphocyte Count , Male , Mice , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction
13.
Orphanet J Rare Dis ; 8: 79, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23692804

ABSTRACT

BACKGROUND: Olmsted syndrome is a rare congenital skin disorder presenting with periorifical hyperkeratotic lesions and mutilating palmoplantar keratoderma, which is often associated with infections of the keratotic area. A recent study identified de novo mutations causing constitutive activation of TRPV3 as a cause of the keratotic manifestations of Olmsted syndrome. METHODS: Genetic, clinical and immunological profiling was performed on a case study patient with the clinical diagnosis of Olmsted syndrome. RESULTS: The patient was found to harbour a previously undescribed 1718G-C transversion in TRPV3, causing a G573A point mutation. In depth clinical and immunological analysis found multiple indicators of immune dysregulation, including frequent dermal infections, inflammatory infiltrate in the affected skin, hyper IgE production and elevated follicular T cells and eosinophils in the peripheral blood. CONCLUSIONS: These results provide the first comprehensive assessment of the immunological features of Olmsted syndrome. The systemic phenotype of hyper IgE and persistent eosinophilia suggest a primary or secondary role of immunological processes in the pathogenesis of Olmsted syndrome, and have important clinical consequences with regard to the treatment of Olmsted syndrome patients.


Subject(s)
Keratoderma, Palmoplantar/immunology , Keratoderma, Palmoplantar/physiopathology , Keratosis/immunology , Keratosis/physiopathology , Adult , Eosinophilia/genetics , Eosinophilia/immunology , Eosinophilia/physiopathology , Facial Dermatoses/genetics , Facial Dermatoses/pathology , Female , Humans , Hyperplasia/genetics , Hyperplasia/immunology , Hyperplasia/pathology , Immunoglobulin E/blood , Immunoglobulin E/genetics , Keratoderma, Palmoplantar/genetics , Keratosis/genetics , Male , Mutation , Phenotype , Skin/pathology , Syndrome , TRPV Cation Channels/genetics , Young Adult
14.
Ann Neurol ; 73(3): 433-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23483640

ABSTRACT

Association studies have implicated common variants in the 12q14.1 region containing CYP27B1 in multiple sclerosis (MS). Rare CYP27B1 mutations cause autosomal recessive vitamin D-dependent rickets type 1, and it has recently been reported that heterozygous CYP27B1 mutations are associated with increased MS susceptibility and lower active vitamin D levels. By sequencing CYP27B1 in 134 multiplex families and genotyping the most common variant R389H in 2,608 MS patients and 1,987 controls from Italy and Belgium (a total of 4,729 individuals), we were unable to replicate these observations. These results provide evidence against a major role for CYP27B1 mutations in MS.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Multiple Sclerosis/genetics , Mutation/genetics , Adult , Belgium , Case-Control Studies , Computational Biology , Female , Genotype , Humans , Italy , Male , Middle Aged , Multiple Sclerosis/blood , Radioimmunoassay , Vitamin D/analogs & derivatives , Vitamin D/blood , Young Adult
15.
Eur J Immunol ; 43(1): 75-84, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23041971

ABSTRACT

The autoimmune regulator (Aire), mediates central tolerance of peripheral self. Its activity in thymic epithelial cells (TECs) directs the ectopic expression of thousands of tissue-restricted antigens (TRAs), causing the deletion of autoreactive thymocytes. The molecular mechanisms orchestrating the breadth of transcriptional regulation by Aire remain unknown. One prominent model capable of explaining both the uniquely high number of Aire-dependent targets and their specificity posits that tissue-specific transcription factors induced by Aire directly activate their canonical targets, exponentially adding to the total number of Aire-dependent TRAs. To test this "Hierarchical Transcription" model, we analysed mice deficient in the pancreatic master transcription factor pancreatic and duodenal homeobox 1 (Pdx1), specifically in TECs (Pdx1(ΔFoxn1) ), for the expression and tolerance of pancreatic TRAs. Surprisingly, we found that lack of Pdx1 in TECs did not reduce the transcription of insulin or somatostatin, or alter glucagon expression. Moreover, in a model of thymic deletion driven by a neo-TRA under the control of the insulin promoter, Pdx1 in TECs was not required to affect thymocyte deletion or the generation of regulatory T (Treg) cells. These findings suggest that the capacity of Aire to regulate expression of a huge array of TRAs relies solely on an unconventional transcriptional mechanism, without intermediary transcription factors.


Subject(s)
Central Tolerance , Homeodomain Proteins/metabolism , Pancreas/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Autoantigens/genetics , Autoantigens/metabolism , Autoimmunity , Cells, Cultured , Clonal Deletion/genetics , Epithelial Cells/immunology , Homeodomain Proteins/genetics , Insulin/genetics , Insulin/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Organ Specificity , Promoter Regions, Genetic/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic , AIRE Protein
16.
Blood ; 118(7): 1845-53, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21715314

ABSTRACT

Foxp3(+) regulatory T cells play a pivotal role in maintaining self-tolerance and immune homeostasis. In the absence of regulatory T cells, generalized immune activation and multiorgan T cell-driven pathology occurs. Although the phenomenon of immunologic control by Foxp3(+) regulatory T cells is well recognized, the comparative effect over different arms of the immune system has not been thoroughly investigated. Here, we generated a cohort of mice with a continuum of regulatory T-cell frequencies ranging from physiologic levels to complete deficiency. This titration of regulatory T-cell depletion was used to determine how different effector subsets are controlled. We found that in vivo Foxp3(+) regulatory T-cell frequency had a proportionate relationship with generalized T-cell activation and Th1 magnitude, but it had a surprising disproportionate relationship with Th2 magnitude. The asymmetric regulation was associated with efficient suppression of Th2 cells through additional regulations on the apoptosis rate in Th2 cells and not Th1 cells and could be replicated by CTLA4-Ig or anti-IL-2 Ab. These results indicate that the Th2 arm of the immune system is under tighter control by regulatory T cells than the Th1 arm, suggesting that Th2-driven diseases may be more responsive to regulatory T-cell manipulation.


Subject(s)
Apoptosis , Forkhead Transcription Factors/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/cytology , Abatacept , Animals , Immunoconjugates/immunology , Immunosuppressive Agents/immunology , Interleukin-2/immunology , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...