Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Proc Natl Acad Sci U S A ; 112(50): 15414-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26621740

ABSTRACT

Human interleukin 12 and interleukin 23 (IL12/23) influence susceptibility or resistance to multiple diseases. However, the reasons underlying individual differences in IL12/23 sensitivity remain poorly understood. Here we report that in human peripheral blood mononuclear cells (PBMCs) and inflamed lungs, the majority of interleukin-12 receptor ß1 (IL12RB1) mRNAs contain a number of RNA-DNA differences (RDDs) that concentrate in sequences essential to IL12Rß1's binding of IL12p40, the protein subunit common to both IL-12 and IL-23. IL12RB1 RDDs comprise multiple RDD types and are detectable by next-generation sequencing and classic Sanger sequencing. As a consequence of these RDDs, the resulting IL12Rß1 proteins have an altered amino acid sequence that could not be predicted on the basis of genomic DNA sequencing alone. Importantly, the introduction of RDDs into IL12RB1 mRNAs negatively regulates IL12Rß1's binding of IL12p40 and is sensitive to activation. Collectively, these results suggest that the introduction of RDDs into an individual's IL12RB1 mRNA repertoire is a novel determinant of IL12/23 sensitivity.


Subject(s)
DNA/metabolism , RNA/metabolism , Receptors, Interleukin-12/metabolism , Adult , Base Sequence , Gene Expression Regulation/drug effects , Humans , Interleukin-12/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Models, Biological , Molecular Sequence Data , Phytohemagglutinins/pharmacology , Pneumonia/genetics , Pneumonia/pathology , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-12/genetics , Recombinant Proteins/biosynthesis
2.
J Clin Immunol ; 33(1): 30-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22930256

ABSTRACT

PURPOSE: A subset of patients with common variable immunodeficiency (CVID) develops granulomatous and lymphocytic interstitial lung disease (GLILD), a restrictive lung disease associated with early mortality. The optimal therapy for GLILD is unknown. This study was undertaken to see if rituximab and azathioprine (combination chemotherapy) would improve pulmonary function and/or radiographic abnormalities in patients with CVID and GLILD. METHODS: A retrospective chart review of patients with CVID and GLILD who were treated with combination chemotherapy was performed. Complete pulmonary function tests (PFTs) and high-resolution computed tomography (HRCT) scans of the chest were done prior to therapy and >6 months later. HRCT scans of the chest were blinded, randomized, and scored independently (in pairs) by two radiologists. The differences between pre- and post-treatment HRCT scores and PFT parameters were analyzed. RESULTS: Seven patients with CVID and GLILD met inclusion criteria. Post-treatment increases were noted in both FEV1 (p=0.034) and FVC (p=0.043). HRCT scans of the chest demonstrated improvement in total score (p=0.018), pulmonary consolidations (p=0.041), ground-glass opacities (p=0.020) nodular opacities (p=0.024), and both the presence and extent of bronchial wall thickening (p=0.014, 0.026 respectively). No significant chemotherapy-related complications occurred. CONCLUSIONS: Combination chemotherapy improved pulmonary function and decreased radiographic abnormalities in patients with CVID and GLILD.


Subject(s)
Common Variable Immunodeficiency/drug therapy , Common Variable Immunodeficiency/immunology , Granuloma/drug therapy , Granuloma/immunology , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/immunology , Administration, Oral , Adolescent , Adult , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Azathioprine/administration & dosage , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Common Variable Immunodeficiency/pathology , Drug Therapy, Combination , Female , Granuloma/pathology , Humans , Infusions, Intravenous , Lung Diseases, Interstitial/pathology , Male , Retrospective Studies , Rituximab , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Young Adult
3.
J Immunol ; 189(9): 4684-94, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23024274

ABSTRACT

IL12RB1 is essential for human resistance to multiple intracellular pathogens, including Mycobacterium tuberculosis. In its absence, the proinflammatory effects of the extracellular cytokines IL-12 and IL-23 fail to occur, and intracellular bacterial growth goes unchecked. Given the recent observation that mouse leukocytes express more than one isoform from il12rb1, we examined whether primary human leukocytes similarly express more than one isoform from IL12RB1. We observed that human leukocytes express as many as 13 distinct isoforms, the relative levels of each being driven by inflammatory stimuli both in vitro and in vivo. Surprisingly, the most abundant isoform present before stimulation is a heretofore uncharacterized intracellular form of the IL-12R (termed "isoform 2") that presumably has limited contact with extracellular cytokine. After stimulation, primary PBMCs, including the CD4(+), CD8(+), and CD56(+) lineages contained therein, alter the splicing of IL12RB1 RNA to increase the relative abundance of isoform 1, which confers IL-12/IL-23 responsiveness. These data demonstrate both a posttranscriptional mechanism by which cells regulate their IL-12/IL-23 responsiveness, and that leukocytes primarily express IL12RB1 in an intracellular form located away from extracellular cytokine.


Subject(s)
Gene Expression Regulation/immunology , Inflammation Mediators/physiology , Receptors, Interleukin-12/biosynthesis , Receptors, Interleukin-12/genetics , Signal Transduction/immunology , Adult , Alternative Splicing/genetics , Alternative Splicing/immunology , Amino Acid Sequence , Base Sequence , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 19/immunology , Exons/genetics , Exons/immunology , Genome, Human/genetics , Genome, Human/immunology , HEK293 Cells , Humans , Inflammation Mediators/isolation & purification , Jurkat Cells , Molecular Sequence Data , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , RNA Processing, Post-Transcriptional/genetics , RNA Processing, Post-Transcriptional/immunology , Receptors, Interleukin-12/isolation & purification , Signal Transduction/genetics
4.
J Vis Exp ; (53)2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21808226

ABSTRACT

Regulatory T cells (Tregs) are critical mediators of immune tolerance to self-antigens. In addition, they are crucial regulators of the immune response following an infection. Despite efforts to identify unique surface marker on Tregs, the only unique feature is their ability to suppress the proliferation and function of effector T cells. While it is clear that only in vitro assays can be used in assessing human Treg function, this becomes problematic when assessing the results from cross-sectional studies where healthy cells and cells isolated from subjects with autoimmune diseases (like Type 1 Diabetes-T1D) need to be compared. There is a great variability among laboratories in the number and type of responder T cells, nature and strength of stimulation, Treg:responder ratios and the number and type of antigen-presenting cells (APC) used in human in vitro suppression assays. This variability makes comparison between studies measuring Treg function difficult. The Treg field needs a standardized suppression assay that will work well with both healthy subjects and those with autoimmune diseases. We have developed an in vitro suppression assay that shows very little intra-assay variability in the stimulation of T cells isolated from healthy volunteers compared to subjects with underlying autoimmune destruction of pancreatic ß-cells. The main goal of this piece is to describe an in vitro human suppression assay that allows comparison between different subject groups. Additionally, this assay has the potential to delineate a small loss in nTreg function and anticipate further loss in the future, thus identifying subjects who could benefit from preventive immunomodulatory therapy. Below, we provide thorough description of the steps involved in this procedure. We hope to contribute to the standardization of the in vitro suppression assay used to measure Treg function. In addition, we offer this assay as a tool to recognize an early state of immune imbalance and a potential functional biomarker for T1D.


Subject(s)
Immune System Diseases/diagnosis , Immune System Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Flow Cytometry/methods , Humans , T-Lymphocytes, Regulatory/cytology
5.
PLoS One ; 5(12): e15154, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21151941

ABSTRACT

BACKGROUND: In type 1 diabetes (T1D), a prototypic autoimmune disease, effector T cells destroy beta cells. Normally, CD4(+)CD25(+high), or natural regulatory T cells (Tregs), counter this assault. In autoimmunity, the failure to suppress CD4(+)CD25(low) T cells is important for disease development. However, both Treg dysfunction and hyperactive responder T-cell proliferation contribute to disease. METHODS/PRINCIPAL FINDINGS: We investigated human CD4(+)CD25(low) T cells and compared them to CD4(+)CD25(-) T cells in otherwise equivalent in vitro proliferative conditions. We then asked whether these differences in suppression are exacerbated in T1D. In both single and co-culture with Tregs, the CD4(+)CD25(low) T cells divided more rapidly than CD4(+)CD25(-) T cells, which manifests as increased proliferation/reduced suppression. Time-course experiments showed that this difference could be explained by higher IL-2 production from CD4+CD25(low) compared to CD4+CD25- T cells. There was also a significant increase in CD4+CD25(low) T-cell proliferation compared to CD4+CD25- T cells during suppression assays from RO T1D and at-risk subjects (n = 28, p = 0.015 and p = 0.024 respectively). CONCLUSIONS/SIGNIFICANCE: The in vitro dual suppression assays proposed here could highlight the impaired sensitivity of certain responder T cells to the suppressive effect of Tregs in human autoimmune diseases.


Subject(s)
Diabetes Mellitus, Type 1/immunology , T-Lymphocytes/cytology , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Separation , Coculture Techniques , Flow Cytometry , Humans , Insulin-Secreting Cells/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/biosynthesis , Kinetics , Leukocytes, Mononuclear/cytology , Risk , T-Lymphocytes, Regulatory/cytology
6.
Cell Tissue Res ; 339(3): 585-95, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20143240

ABSTRACT

CD4+CD25+(high) regulatory T cells (Tregs) play a pivotal role in the control of the immune response. A growing body of evidence suggests the reduced function of these cells in autoimmune diseases, including type 1 diabetes (T1D). Restoration of their function can potentially delay further disease development. In the present study, we have converted conventional effector T cells into induced Tregs (iTregs) in recent-onset (RO) T1D (n=9) and compared them with the same cells generated in controls (n=12) and in long-standing (LS) T1D subjects (n=9). The functional potential of in-vitro-generated Tregs was measured by using an in vitro proliferation assay. We noted that the suppressive potential of iTregs exceeded that of natural regulatory T cells (nTregs) only in the RO T1D subjects. We showed that iTregs from RO T1D subjects had increased expression of Foxp3, E3 ubiquitin ligase (ITCH) and TGF-beta-inducible early gene 1 (TIEG1) compared with control and LS T1D subjects. We also expanded natural, thymically derived Tregs (nTregs) and compared the functional ability of these cells between subject groups. Expanded cells from all three subject groups were suppressive. RO T1D subjects were the only group in which both iTregs and expanded Tregs were functional, suggesting that the inflammatory milieu impacts in vitro Treg generation. Future longitudinal studies should delineate the actual contribution of the stage of disease to the quality of in-vitro-generated Tregs.


Subject(s)
Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/immunology , Immune Tolerance/immunology , Repressor Proteins/metabolism , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Ubiquitin-Protein Ligases/metabolism , Adult , Age of Onset , Apoptosis , Case-Control Studies , Cell Count , Child , Diabetes Mellitus, Type 1/epidemiology , Early Growth Response Transcription Factors/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Male
7.
Eur J Immunol ; 39(9): 2571-83, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19701891

ABSTRACT

The transcription factor Foxp3 is essential for the development of functional, natural Treg (nTreg), which plays a prominent role in self-tolerance. Suppressive Foxp3(+) Treg cells can be generated from naïve T cells ex vivo, following TCR and TGF-beta1 stimulations. However, the molecular contributions from the different arms of these pathways leading to Foxp3 expression are not fully understood. TGF-beta1-activated Smad3 plays a major role in the expression of Foxp3, since TGF-beta1-induced-Treg generation from Smad3(-/-) mice is markedly reduced and abolished by inactivating Smad2. In the TCR pathway, deletion of Bcl10, which activates NF-kappaB, markedly reduces both IL-2 and Foxp3 production. However, partial rescue of Foxp3 expression occurs on addition of exogenous IL-2. TGF-beta1 significantly attenuates NF-kappaB binding to the Foxp3 promoter, while inducing Foxp3 expression. Furthermore, deletion of p50, a NF-kappaB subunit, results in increased Foxp3 expression despite a decline in the IL-2 production. We posit several TCR-NF-kappaB pathways, some increasing (Bcl10-IL-2-Foxp3) while others decreasing (p50-Foxp3) Foxp3 expression, with the former predominating. A better understanding of Foxp3 regulation could be useful in dissecting the cause of Treg dysfunction in several autoimmune diseases and for generating more potent TGF-beta1-induced-Treg cells for therapeutic purposes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/biosynthesis , NF-kappa B p50 Subunit/metabolism , Smad3 Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Cells, Cultured , Forkhead Transcription Factors/genetics , Interleukin-2/pharmacology , Mice , Mice, Inbred C57BL , NF-kappa B p50 Subunit/antagonists & inhibitors , Peptides/pharmacology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Smad3 Protein/genetics , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta1/pharmacology
8.
PLoS One ; 4(8): e6527, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19654878

ABSTRACT

BACKGROUND: Type 1 diabetes (T1D) is a T-cell mediated autoimmune disease targeting the insulin-producing pancreatic beta cells. Naturally occurring FOXP3(+)CD4(+)CD25(high) regulatory T cells (T(regs)) play an important role in dominant tolerance, suppressing autoreactive CD4(+) effector T cell activity. Previously, in both recent-onset T1D patients and beta cell antibody-positive at-risk individuals, we observed increased apoptosis and decreased function of polyclonal T(regs) in the periphery. Our objective here was to elucidate the genes and signaling pathways triggering apoptosis in T(regs) from T1D subjects. PRINCIPAL FINDINGS: Gene expression profiles of unstimulated T(regs) from recent-onset T1D (n = 12) and healthy control subjects (n = 15) were generated. Statistical analysis was performed using a Bayesian approach that is highly efficient in determining differentially expressed genes with low number of replicate samples in each of the two phenotypic groups. Microarray analysis showed that several cytokine/chemokine receptor genes, HLA genes, GIMAP family genes and cell adhesion genes were downregulated in T(regs) from T1D subjects, relative to control subjects. Several downstream target genes of the AKT and p53 pathways were also upregulated in T1D subjects, relative to controls. Further, expression signatures and increased apoptosis in T(regs) from T1D subjects partially mirrored the response of healthy T(regs) under conditions of IL-2 deprivation. CD4(+) effector T-cells from T1D subjects showed a marked reduction in IL-2 secretion. This could indicate that prior to and during the onset of disease, T(regs) in T1D may be caught up in a relatively deficient cytokine milieu. CONCLUSIONS: In summary, expression signatures in T(regs) from T1D subjects reflect a cellular response that leads to increased sensitivity to apoptosis, partially due to cytokine deprivation. Further characterization of these signaling cascades should enable the detection of genes that can be targeted for restoring T(reg) function in subjects predisposed to T1D.


Subject(s)
Apoptosis , CD4 Antigens/immunology , Diabetes Mellitus, Type 1/pathology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2/administration & dosage , T-Lymphocytes/cytology , Apoptosis/genetics , Bayes Theorem , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Gene Expression Profiling , Humans , T-Lymphocytes/immunology
9.
PLoS One ; 2(1): e146, 2007 Jan 03.
Article in English | MEDLINE | ID: mdl-17206281

ABSTRACT

BACKGROUND: In experimental models, Type 1 diabetes T1D can be prevented by adoptive transfer of CD4+CD25+ (FoxP3+) suppressor or regulatory T cells. Recent studies have found a suppression defect of CD4+CD25+(high) T cells in human disease. In this study we measure apoptosis of CD4+CD25+(high) T cells to see if it could contribute to reduced suppressive activity of these cells. METHODS AND FINDINGS: T-cell apoptosis was evaluated in children and adolescent 35 females/40 males subjects comprising recent-onset and long-standing T1D subjects and their first-degree relatives, who are at variable risk to develop T1D. YOPRO1/7AAD and intracellular staining of the active form of caspase 3 were used to evaluate apoptosis. Isolated CD4+CD25+(high) and CD4+CD25- T cells were co-cultured in a suppression assay to assess the function of the former cells. We found that recent-onset T1D subjects show increased apoptosis of CD4+CD25+(high) T cells when compared to both control and long-standing T1D subjects p<0.0001 for both groups. Subjects at high risk for developing T1D 2-3Ab+ve show a similar trend p<0.02 and p<0.01, respectively. On the contrary, in long-standing T1D and T2D subjects, CD4+CD25+(high) T cell apoptosis is at the same level as in control subjects p = NS. Simultaneous intracellular staining of the active form of caspase 3 and FoxP3 confirmed recent-onset FoxP3+ve CD4+CD25+(high) T cells committed to apoptosis at a higher percentage 15.3+/-2.2 compared to FoxP3+ve CD4+CD25+(high) T cells in control subjects 6.1+/-1.7 p<0.002. Compared to control subjects, both recent-onset T1D and high at-risk subjects had significantly decreased function of CD4+CD25+(high) T cells p = 0.0007 and p = 0.007, respectively. CONCLUSIONS: There is a higher level of ongoing apoptosis in CD4+CD25+(high) T cells in recent-onset T1D subjects and in subjects at high risk for the disease. This high level of CD4+CD25+(high) T-cell apoptosis could be a contributing factor to markedly decreased suppressive potential of these cells in recent-onset T1D subjects.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Interleukin-2 Receptor alpha Subunit/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Child , Diabetes Mellitus, Type 1/genetics , Female , Genetic Predisposition to Disease , Humans , Male , Risk Factors
10.
Ann N Y Acad Sci ; 1005: 284-7, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14679077

ABSTRACT

We have created an immunology-related microarray chip containing primarily known genes with well-studied functional properties. By looking at known genes rather than expressed sequence tags, we hope to gain a better understanding of immunological pathways and how they work. The immunology gene chip contains genes from the following functional categories: T cell genes; B cell genes; dendritic cell genes; chemokine and cytokine genes; apoptosis genes; cell cycle genes; cell interaction genes; general hematology and immunology genes; and adhesion genes. We have also developed a novel three-color cDNA array platform in which arrays are directly visualized before hybridization, which allows us to select only high-quality chips for our experiments. In an effort to provide quantitative quality control for each array element as well as the entire chip, we have developed Matarray, a software package for image processing and data acquisition. With Matarray, we have built a quantitative data filtering and normalization scheme that has proved to be more efficient than the existing methods. The list of immunology chip genes is available from the authors.


Subject(s)
Oligonucleotide Array Sequence Analysis , Quality Control , Chemokines/genetics , Cytokines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...