Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cell Calcium ; 111: 102715, 2023 05.
Article in English | MEDLINE | ID: mdl-36933289

ABSTRACT

Cell swelling as a result of hypotonic stress is counteracted in mammalian cells by a process called regulatory volume decrease (RVD). We have recently discovered that RVD of human keratinocytes requires the LRRC8 volume-regulated anion channel (VRAC) and that Ca2+ exerts a modulatory function on RVD. However, the ion channel that is responsible for Ca2+ influx remains unknown. We investigated in this study whether the Ca2+-permeable TRPV4 ion channel, which functions as cell volume sensor in many cell types, may be involved in cell volume regulation during hypotonic stress response of human keratinocytes. We interfered with TRPV4 function in two human keratinocyte cell lines (HaCaT and NHEK-E6/E7) by using two TRPV4-specific inhibitors (RN1734 and GSK2193874), and by creating a CRISPR/Cas9-mediated genetic TRPV4-/- knockout in HaCaT cells. We employed electrophysiological patch clamp analysis, fluorescence-based Ca2+ imaging and cell volume measurements to determine the functional importance of TRPV4. We could show that both hypotonic stress and direct activation of TRPV4 by the specific agonist GSK1016790A triggered intracellular Ca2+ response. Strikingly, the Ca2+ increase upon hypotonic stress was neither affected by genetic knockout of TRPV4 in HaCaT cells nor by pharmacological inhibition of TRPV4 in both keratinocyte cell lines. Accordingly, hypotonicity-induced cell swelling, downstream activation of VRAC currents as well as subsequent RVD were unaffected both in TRPV4 inhibitor-treated keratinocytes and in HaCaT-TRPV4-/- cells. In summary, our study shows that keratinocytes do not require TRPV4 for coping with hypotonic stress, which implies the involvement of other, yet unidentified Ca2+ channels.


Subject(s)
Keratinocytes , TRPV Cation Channels , Animals , Humans , Osmotic Pressure , TRPV Cation Channels/metabolism , Cell Line , Keratinocytes/metabolism , Cell Size , Calcium/metabolism , Hypotonic Solutions/pharmacology , Hypotonic Solutions/metabolism , Mammals/metabolism
2.
BMC Cancer ; 22(1): 386, 2022 Apr 09.
Article in English | MEDLINE | ID: mdl-35397545

ABSTRACT

BACKGROUND: Invadopodia, actin-rich structures that release metallo-proteases at the interface with extra-cellular matrix, in a punctate manner are thought to be important drivers of tumour invasion. Invadopodia formation has been observed in-vitro and in-vivo in numerous metastatic cell lines derived from multiple tumour types. However, prostate cancer cell lines have not been routinely reported to generate invadopodia and the few instances have always required external stimulation. METHODS: In this study, the invasive potential of primary prostate adenocarcinoma cell lines, which have never been fully characterised before, was investigated both in-vitro invadopodia assays and in-vivo zebrafish dissemination assay. Subsequently, circulating tumour cells from prostate cancer patients were isolated and tested in the invadopodia assay. RESULTS: Retention of E-cadherin and N-cadherin expression indicated a transitional state of EMT progression, consistent with the idea of partial EMT that has been frequently observed in aggressive prostate cancer. All cell lines tested were capable of spontaneous invadopodia formation and possess a significant degradative ability in-vitro under basal conditions. These cell lines were invasive in-vivo and produced visible metastasis in the zebrafish dissemination assay. Importantly we have proceeded to demonstrate that circulating tumour cells isolated from prostate cancer patients exhibit invadopodia-like structures and degrade matrix with visible puncta. This work supports a role for invadopodia activity as one of the mechanisms of dissemination employed by prostate cancer cells. CONCLUSION: The combination of studies presented here provide clear evidence that invadopodia activity can play a role in prostate cancer progression.


Subject(s)
Neoplastic Cells, Circulating , Podosomes , Prostatic Neoplasms , Animals , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Male , Neoplasm Invasiveness/pathology , Neoplastic Cells, Circulating/metabolism , Podosomes/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Zebrafish
3.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33538776

ABSTRACT

B cells emerge from the bone marrow as transitional (TS) B cells that differentiate through T1, T2, and T3 stages to become naive B cells. We have identified a bifurcation of human B cell maturation from the T1 stage forming IgMhi and IgMlo developmental trajectories. IgMhi T2 cells have higher expression of α4ß7 integrin and lower expression of IL-4 receptor (IL4R) compared with the IgMlo branch and are selectively recruited into gut-associated lymphoid tissue. IgMhi T2 cells also share transcriptomic features with marginal zone B cells (MZBs). Lineage progression from T1 cells to MZBs via an IgMhi trajectory is identified by pseudotime analysis of scRNA-sequencing data. Reduced frequency of IgMhi gut-homing T2 cells is observed in severe SLE and is associated with reduction of MZBs and their putative IgMhi precursors. The collapse of the gut-associated MZB maturational axis in severe SLE affirms its existence in health.


Subject(s)
Cell Differentiation/immunology , Gastrointestinal Tract/immunology , Immunoglobulin M/metabolism , Lupus Nephritis/immunology , Lymphoid Tissue/immunology , Precursor Cells, B-Lymphoid/immunology , Adult , Aged , Blood Donors , Case-Control Studies , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Female , Humans , Integrin beta Chains/metabolism , Interleukin-4 Receptor alpha Subunit/metabolism , Lupus Nephritis/blood , Lupus Nephritis/pathology , Male , Middle Aged , Phenotype , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome , Young Adult
4.
Med ; 2(2): 149-163.e4, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33163984

ABSTRACT

BACKGROUND: Antibody responses to virus reflect exposure and potential protection. METHODS: We developed a highly specific and sensitive approach to measuring antibodies against SARS-CoV-2 for population-scale immune surveillance. Antibody positivity was defined as a dual-positive response against both the receptor-binding domain and nucleocapsid proteins of SARS-CoV-2. Antibodies were measured by immunoprecipitation assays in capillary blood from 15,771 children aged 1 to 18 years living in Bavaria, Germany, and participating in a public health type 1 diabetes screening program (ClinicalTrials.gov: NCT04039945), in 1,916 dried blood spots from neonates in a Bavarian screening study (ClinicalTrials.gov: NCT03316261), and in 75 SARS-CoV-2-positive individuals. Virus positive incidence was obtained from the Bavarian health authority data. FINDINGS: Dual-antibody positivity was detected in none of the 3,887 children in 2019 (100% specificity) and 73 of 75 SARS-CoV-2-positive individuals (97.3% sensitivity). Antibody surveillance in children during 2020 resulted in frequencies of 0.08% in January to March, 0.61% in April, 0.74% in May, 1.13% in June, and 0.91% in July. Antibody prevalence from April 2020 was 6-fold higher than the incidence of authority-reported cases (156 per 100,000 children), showed marked variation between the seven Bavarian regions (p < 0.0001), and was not associated with age or sex. Transmission in children with virus-positive family members was 35%. 47% of positive children were asymptomatic. No association with type 1 diabetes autoimmunity was observed. Antibody frequency in newborns was 0.47%. CONCLUSIONS: We demonstrate the value of population-based screening programs for pandemic monitoring. FUNDING: The work was supported by funding from the BMBF (FKZ01KX1818).


Subject(s)
COVID-19 , Diabetes Mellitus, Type 1 , Antibodies, Viral , COVID-19/diagnosis , Child , Diabetes Mellitus, Type 1/diagnosis , Humans , Infant, Newborn , Public Health , SARS-CoV-2
5.
J Immunol ; 204(12): 3129-3138, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32404353

ABSTRACT

We previously reported that costimulation blockade by abatacept limits the decline of ß-cell function and the frequency of circulating CD4+ central memory T cells (TCM) (CD45RO+CD62L+) in new-onset type 1 diabetes. In human subjects receiving placebo, we found a significant association between an increase in CD4+ TCM cells and the decline of ß-cell function. To extend and refine these findings, we examined changes in human CD4+ and CD8+ naive and memory T cell subsets at greater resolution using polychromatic flow and mass cytometry. In the placebo group, we successfully reproduced the original finding of a significant association between TCM and ß-cell function and extended this to other T cell subsets. Furthermore, we show that abatacept treatment significantly alters the frequencies of a majority of CD4+ conventional and regulatory T cell subsets; in general, Ag-naive subsets increase and Ag-experienced subsets decrease, whereas CD8+ T cell subsets are relatively resistant to drug effects, indicating a lesser reliance on CD28-mediated costimulation. Importantly, abatacept uncouples the relationship between changes in T cell subsets and ß-cell function that is a component of the natural history of the disease. Although these data suggest immunological markers for predicting change in ß-cell function in type 1 diabetes, the finding that abatacept blunts this relationship renders the biomarkers nonpredictive for this type of therapy. In sum, our findings point to a novel mechanism of action for this successful immunotherapy that may guide other disease-modifying approaches for type 1 diabetes.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Immunologic Memory/immunology , Abatacept/pharmacology , B-Lymphocytes/drug effects , Biomarkers/blood , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Diabetes Mellitus, Type 1/blood , Humans , Immunologic Memory/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
6.
Blood ; 136(7): 885-897, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32294156

ABSTRACT

Idiopathic aplastic anemia (AA) has 2 key characteristics: an autoimmune response against hematopoietic stem/progenitor cells and regulatory T-cells (Tregs) deficiency. We have previously demonstrated reduction in a specific subpopulation of Treg in AA, which predicts response to immunosuppression. The aims of the present study were to define mechanisms of Treg subpopulation imbalance and identify potential for therapeutic intervention. We have identified 2 mechanisms that lead to skewed Treg composition in AA: first, FasL-mediated apoptosis on ligand interaction; and, second, relative interleukin-2 (IL-2) deprivation. We have shown that IL-2 augmentation can overcome these mechanisms. Interestingly, when high concentrations of IL-2 were used for in vitro Treg expansion cultures, AA Tregs were able to expand. The expanded populations expressed a high level of p-BCL-2, which makes them resistant to apoptosis. Using a xenograft mouse model, the function and stability of expanded AA Tregs were tested. We have shown that these Tregs were able to suppress the macroscopic clinical features and tissue manifestations of T-cell-mediated graft-versus-host disease. These Tregs maintained their suppressive properties as well as their phenotype in a highly inflammatory environment. Our findings provide an insight into the mechanisms of Treg reduction in AA. We have identified novel targets with potential for therapeutic interventions. Supplementation of ex vivo expansion cultures of Tregs with high concentrations of IL-2 or delivery of IL-2 directly to patients could improve clinical outcomes in addition to standard immunosuppressive therapy.


Subject(s)
Anemia, Aplastic/immunology , Apoptosis/drug effects , Fas Ligand Protein/pharmacology , Interleukin-2/pharmacology , T-Lymphocytes, Regulatory/drug effects , Anemia, Aplastic/pathology , Animals , Apoptosis/immunology , Cells, Cultured , Female , Humans , Immune System Diseases/immunology , Immune System Diseases/pathology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Interleukin-2/deficiency , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , T-Lymphocytes, Regulatory/physiology
9.
Methods Mol Biol ; 1979: 285-303, 2019.
Article in English | MEDLINE | ID: mdl-31028645

ABSTRACT

Mass cytometry is a variation of conventional flow cytometry using metal tagged antibodies for cell staining instead of fluorochromes and detection in a mass cytometer, a modified mass spectrometer that allows for separation of discrete masses of these metal tags by time of flight (TOF). Currently, up to 50 different metal tags are available for cell analysis. The lack of any significant mass spectral overlap and autofluorescence background makes mass cytometry uniquely suited for complex high-dimensional phenotypic and functional analysis at the single cell level, thus accelerating biomarker discovery and drug screening. Here we describe a workflow for phenotyping of human peripheral blood mononuclear cells (PBMCs) covering cell staining, instrument setup of a Fluidigm Helios™ mass cytometer, and sample acquisition, and summarize a basic workflow of data analysis.


Subject(s)
Flow Cytometry/methods , Immunoconjugates/immunology , Immunophenotyping/methods , Leukocytes, Mononuclear/immunology , Cell Separation/methods , Cell Survival , Humans , Immunoconjugates/chemistry , Iridium/chemistry , Iridium/immunology , Isotopes/chemistry , Isotopes/immunology , Leukocytes, Mononuclear/cytology , Rhodium/chemistry , Rhodium/immunology , Single-Cell Analysis/methods , Staining and Labeling/methods
10.
Nat Commun ; 9(1): 3857, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242242

ABSTRACT

Human memory B cells and marginal zone (MZ) B cells share common features such as the expression of CD27 and somatic mutations in their IGHV and BCL6 genes, but the relationship between them is controversial. Here, we show phenotypic progression within lymphoid tissues as MZ B cells emerge from the mature naïve B cell pool via a precursor CD27-CD45RBMEM55+ population distant from memory cells. By imaging mass cytometry, we find that MZ B cells and memory B cells occupy different microanatomical niches in organised gut lymphoid tissues. Both populations disseminate widely between distant lymphoid tissues and blood, and both diversify their IGHV repertoire in gut germinal centres (GC), but nevertheless remain largely clonally separate. MZ B cells are therefore not developmentally contiguous with or analogous to classical memory B cells despite their shared ability to transit through GC, where somatic mutations are acquired.


Subject(s)
B-Lymphocytes , Lymphoid Tissue/cytology , Humans , Immunologic Memory , Lymphoid Tissue/immunology , Phenotype
11.
J Clin Invest ; 128(8): 3460-3474, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29851415

ABSTRACT

In type 1 diabetes, cytotoxic CD8+ T cells with specificity for ß cell autoantigens are found in the pancreatic islets, where they are implicated in the destruction of insulin-secreting ß cells. In contrast, the disease relevance of ß cell-reactive CD8+ T cells that are detectable in the circulation, and their relationship to ß cell function, are not known. Here, we tracked multiple, circulating ß cell-reactive CD8+ T cell subsets and measured ß cell function longitudinally for 2 years, starting immediately after diagnosis of type 1 diabetes. We found that change in ß cell-specific effector memory CD8+ T cells expressing CD57 was positively correlated with C-peptide change in subjects below 12 years of age. Autoreactive CD57+ effector memory CD8+ T cells bore the signature of enhanced effector function (higher expression of granzyme B, killer-specific protein of 37 kDa, and CD16, and reduced expression of CD28) compared with their CD57- counterparts, and network association modeling indicated that the dynamics of ß cell-reactive CD57+ effector memory CD8+ T cell subsets were strongly linked. Thus, coordinated changes in circulating ß cell-specific CD8+ T cells within the CD57+ effector memory subset calibrate to functional insulin reserve in type 1 diabetes, providing a tool for immune monitoring and a mechanism-based target for immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Immunologic Memory , Insulin-Secreting Cells/immunology , Adult , CD8-Positive T-Lymphocytes/pathology , Child , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Female , Humans , Insulin-Secreting Cells/pathology , Male
12.
Sci Transl Med ; 9(402)2017 08 09.
Article in English | MEDLINE | ID: mdl-28794283

ABSTRACT

Immunotherapy using short immunogenic peptides of disease-related autoantigens restores immune tolerance in preclinical disease models. We studied safety and mechanistic effects of injecting human leukocyte antigen-DR4(DRB1*0401)-restricted immunodominant proinsulin peptide intradermally every 2 or 4 weeks for 6 months in newly diagnosed type 1 diabetes patients. Treatment was well tolerated with no systemic or local hypersensitivity. Placebo subjects showed a significant decline in stimulated C-peptide (measuring insulin reserve) at 3, 6, 9, and 12 months versus baseline, whereas no significant change was seen in the 4-weekly peptide group at these time points or the 2-weekly group at 3, 6, and 9 months. The placebo group's daily insulin use increased by 50% over 12 months but remained unchanged in the intervention groups. C-peptide retention in treated subjects was associated with proinsulin-stimulated interleukin-10 production, increased FoxP3 expression by regulatory T cells, low baseline levels of activated ß cell-specific CD8 T cells, and favorable ß cell stress markers (proinsulin/C-peptide ratio). Thus, proinsulin peptide immunotherapy is safe, does not accelerate decline in ß cell function, and is associated with antigen-specific and nonspecific immune modulation.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Immunotherapy/methods , Peptides/therapeutic use , Proinsulin/therapeutic use , Adolescent , Adult , Autoantibodies/immunology , Autoantigens/immunology , C-Peptide/metabolism , Diabetes Mellitus, Type 1/metabolism , Double-Blind Method , Female , Humans , Immunophenotyping , Male , Middle Aged , T-Lymphocytes, Regulatory/metabolism , Young Adult
13.
Blood ; 128(9): 1193-205, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27281795

ABSTRACT

Idiopathic aplastic anemia (AA) is an immune-mediated and serious form of bone marrow failure. Akin to other autoimmune diseases, we have previously shown that in AA regulatory T cells (Tregs) are reduced in number and function. The aim of this study was to further characterize Treg subpopulations in AA and investigate the potential correlation between specific Treg subsets and response to immunosuppressive therapy (IST) as well as their in vitro expandability for potential clinical use. Using mass cytometry and an unbiased multidimensional analytical approach, we identified 2 specific human Treg subpopulations (Treg A and Treg B) with distinct phenotypes, gene expression, expandability, and function. Treg B predominates in IST responder patients, has a memory/activated phenotype (with higher expression of CD95, CCR4, and CD45RO within FOXP3(hi), CD127(lo) Tregs), expresses the interleukin-2 (IL-2)/STAT5 pathway and cell-cycle commitment genes. Furthermore, in vitro-expanded Tregs become functional and take on the characteristics of Treg B. Collectively, this study identifies human Treg subpopulations that can be used as predictive biomarkers for response to IST in AA and potentially other autoimmune diseases. We also show that Tregs from AA patients are IL-2-sensitive and expandable in vitro, suggesting novel therapeutic approaches such as low-dose IL-2 therapy and/or expanded autologous Tregs and meriting further exploration.


Subject(s)
Anemia, Aplastic/immunology , Anemia, Aplastic/therapy , Immunologic Memory , Immunosuppression Therapy/methods , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Female , Forkhead Transcription Factors/immunology , Humans , Interleukin-2/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Leukocyte Common Antigens/immunology , Male , Middle Aged , Receptors, CCR4/immunology , STAT5 Transcription Factor/immunology , fas Receptor/immunology
14.
Sci Rep ; 6: 20686, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26861911

ABSTRACT

Standardization of immunophenotyping requires careful attention to reagents, sample handling, instrument setup, and data analysis, and is essential for successful cross-study and cross-center comparison of data. Experts developed five standardized, eight-color panels for identification of major immune cell subsets in peripheral blood. These were produced as pre-configured, lyophilized, reagents in 96-well plates. We present the results of a coordinated analysis of samples across nine laboratories using these panels with standardized operating procedures (SOPs). Manual gating was performed by each site and by a central site. Automated gating algorithms were developed and tested by the FlowCAP consortium. Centralized manual gating can reduce cross-center variability, and we sought to determine whether automated methods could streamline and standardize the analysis. Within-site variability was low in all experiments, but cross-site variability was lower when central analysis was performed in comparison with site-specific analysis. It was also lower for clearly defined cell subsets than those based on dim markers and for rare populations. Automated gating was able to match the performance of central manual analysis for all tested panels, exhibiting little to no bias and comparable variability. Standardized staining, data collection, and automated gating can increase power, reduce variability, and streamline analysis for immunophenotyping.


Subject(s)
Flow Cytometry/standards , Immunophenotyping/standards , Laboratories/standards , Algorithms , Automation , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Flow Cytometry/methods , Humans , Immunophenotyping/methods , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
J Immunol ; 195(5): 2030-7, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26223658

ABSTRACT

Regulatory T cells (Tregs) are an essential component of the cellular immune response, occupying a key role in maintaining immunological tolerance and present an attractive therapeutic target in a range of immunopathologies. Comprehensive analysis of the human Treg compartment has been restricted due to technical limitations. The advent of mass cytometry enables simultaneous assessment of vastly increased phenotypic parameters at single-cell resolution. In this study, we used mass cytometry to examine the complexity of human Tregs using an extensive panel of surface markers associated with Treg function and phenotype. We applied unsupervised clustering analysis, revealing 22 distinct subpopulations of Tregs, representing previously identified and novel subpopulations. Our data represent the most in-depth phenotypic description of the human Treg compartment at single-cell resolution and show a hitherto unrecognized degree of phenotypic complexity among cells of the regulatory lineage.


Subject(s)
Biomarkers/metabolism , Flow Cytometry/methods , Single-Cell Analysis/methods , T-Lymphocytes, Regulatory/metabolism , Cell Lineage , Cells, Cultured , Cluster Analysis , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Immunophenotyping , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/immunology
16.
Diabetes ; 64(3): 916-925, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25249579

ABSTRACT

Autoreactive CD8 T cells play a central role in the destruction of pancreatic islet ß-cells that leads to type 1 diabetes, yet the key features of this immune-mediated process remain poorly defined. In this study, we combined high-definition polychromatic flow cytometry with ultrasensitive peptide-human leukocyte antigen class I tetramer staining to quantify and characterize ß-cell-specific CD8 T cell populations in patients with recent-onset type 1 diabetes and healthy control subjects. Remarkably, we found that ß-cell-specific CD8 T cell frequencies in peripheral blood were similar between subject groups. In contrast to healthy control subjects, however, patients with newly diagnosed type 1 diabetes displayed hallmarks of antigen-driven expansion uniquely within the ß-cell-specific CD8 T cell compartment. Molecular analysis of selected ß-cell-specific CD8 T cell populations further revealed highly skewed oligoclonal T cell receptor repertoires comprising exclusively private clonotypes. Collectively, these data identify novel and distinctive features of disease-relevant CD8 T cells that inform the immunopathogenesis of type 1 diabetes.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Adult , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/physiology , Female , Flow Cytometry , Glutamate Decarboxylase/immunology , Humans , Insulin-Secreting Cells/cytology , Male , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunology
17.
PLoS One ; 8(7): e65485, 2013.
Article in English | MEDLINE | ID: mdl-23843942

ABSTRACT

Discovery of novel immune biomarkers for monitoring of disease prognosis and response to therapy in immune-mediated inflammatory diseases is an important unmet clinical need. Here, we establish a novel framework for immunological biomarker discovery, comparing a conventional (liquid) flow cytometry platform (CFP) and a unique lyoplate-based flow cytometry platform (LFP) in combination with advanced computational data analysis. We demonstrate that LFP had higher sensitivity compared to CFP, with increased detection of cytokines (IFN-γ and IL-10) and activation markers (Foxp3 and CD25). Fluorescent intensity of cells stained with lyophilized antibodies was increased compared to cells stained with liquid antibodies. LFP, using a plate loader, allowed medium-throughput processing of samples with comparable intra- and inter-assay variability between platforms. Automated computational analysis identified novel immunophenotypes that were not detected with manual analysis. Our results establish a new flow cytometry platform for standardized and rapid immunological biomarker discovery with wide application to immune-mediated diseases.


Subject(s)
Biomarkers , Computational Biology/methods , Flow Cytometry/methods , Immunophenotyping/methods , Inflammation/immunology , Inflammation/metabolism , Adult , Antibodies , Female , Flow Cytometry/standards , Humans , Immunophenotyping/standards , Interferon-gamma/metabolism , Interleukin-10/metabolism , Male , Middle Aged , Reproducibility of Results , Sensitivity and Specificity , Young Adult
18.
Eur J Immunol ; 43(8): 2043-54, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23677517

ABSTRACT

Treg cells are critical for the prevention of autoimmune diseases and are thus prime candidates for cell-based clinical therapy. However, human Treg cells are "plastic", and are able to produce IL-17 under inflammatory conditions. Here, we identify and characterize the human Treg subpopulation that can be induced to produce IL-17 and identify its mechanisms. We confirm that a subpopulation of human Treg cells produces IL-17 in vitro when activated in the presence of IL-1ß, but not IL-6. "IL-17 potential" is restricted to population III (CD4(+) CD25(hi) CD127(lo) CD45RA(-) ) Treg cells expressing the natural killer cell marker CD161. We show that these cells are functionally as suppressive and have similar phenotypic/molecular characteristics to other subpopulations of Treg cells and retain their suppressive function following IL-17 induction. Importantly, we find that IL-17 production is STAT3 dependent, with Treg cells from patients with STAT3 mutations unable to make IL-17. Finally, we show that CD161(+) population III Treg cells accumulate in inflamed joints of patients with inflammatory arthritis and are the predominant IL-17-producing Treg-cell population at these sites. As IL-17 production from this Treg-cell subpopulation is not accompanied by a loss of regulatory function, in the context of cell therapy, exclusion of these cells from the cell product may not be necessary.


Subject(s)
Interleukin-17/biosynthesis , NK Cell Lectin-Like Receptor Subfamily B/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adult , Aged , CD4 Antigens/biosynthesis , Cells, Cultured , Female , Forkhead Transcription Factors/biosynthesis , Humans , Interleukin-1beta/metabolism , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-6/metabolism , Interleukin-7 Receptor alpha Subunit/biosynthesis , Leukocyte Common Antigens/biosynthesis , Lymphocyte Activation , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily B/immunology , STAT3 Transcription Factor/genetics
19.
Cytometry A ; 79(7): 496-506, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21638766

ABSTRACT

Asymmetric cell division is an important mechanism for generating cellular diversity, however, techniques for measuring the distribution of fate-regulating molecules during mitosis have been hampered by a lack of objectivity, quantitation, and statistical robustness. Here we describe a novel imaging flow cytometric approach that is able to report a cells proliferative history and cell cycle position using dye dilution, pH3, and PI staining to then measure the spatial distribution of fluorescent signals during mitosis using CCD-derived imagery. Using Jurkat cells, resolution of the fluorescently labeled populations was comparable to traditional PMT based cytometers thus eliminating the need to sort cells with specific division histories for microscopy. Subdividing mitotic stages by morphology allowed us to determine the time spent in each cell cycle phase using mathematical modeling approaches. Furthermore high sample throughput allowed us to collect statistically relevant numbers of cells without the need to use blocking agents that artificially enrich for mitotic events. The fluorescent imagery was used to measure PKCζ protein and EEA-1+ endosome distribution during different mitotic phases in Jurkat cells. While telophase cells represented the favorable population for measuring asymmetry, asynchronously dividing cells spent approximately 43 seconds in this stage, explaining why they were present at such low frequencies. This necessitated the acquisition of large cell numbers. Interestingly we found that PKCζ was inherited asymmetrically in 2.5% of all telophasic events whereas endosome inheritance was significantly more symmetrical. Furthermore, molecular polarity at early mitotic phases was a poor indicator of asymmetry during telophase highlighting that, though rare, telophasic events represented the best candidates for asymmetry studies. In summary, this technique combines the spatial information afforded by fluorescence microscopy with the statistical wealth and objectivity of traditional flow cytometry, overcoming the key limitations of existing approaches for studying asymmetry during mitosis.


Subject(s)
Cell Division/physiology , Flow Cytometry/methods , Image Cytometry/methods , Mitosis/physiology , Animals , Flow Cytometry/instrumentation , Humans , Image Cytometry/instrumentation , Jurkat Cells/cytology , Microscopy, Fluorescence/methods , Protein Kinase C/metabolism
20.
Blood ; 116(22): 4639-45, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20688957

ABSTRACT

Immune thrombocytopenia (ITP) is an autoantibody-mediated bleeding disorder with both accelerated platelet destruction and impaired platelet production. We and others have described impaired regulatory CD4(+)CD25(hi) T cells (Treg) numbers and/or suppressive function in ITP patients. Clinical trials using thrombopoietic agents to stimulate platelet production have shown favorable outcomes in ITP patients, but information on the immunologic responses of treated patients are lacking. We studied the immunologic profile of chronic ITP patients before (n = 10) and during treatment with thrombopoietin receptor (TPO-R) agonists (n = 9). Treg activity, as measured by suppression of proliferation of autologous CD4(+) CD25(-) cells, was improved in patients on treatment (P < .05), and the improvement correlated with reduction in interleukin-2-producing CD4(+) cells, consistent with dampening of immune responses. There was a concomitant increase in total circulating transforming growth factor-ß1 (TGF-ß1) levels (P = .002) in patients on treatment, and the levels of TGF-ß1 correlated with the degree of improvement in platelet counts (r = .8, P = .0002). This suggests that platelets in patients on TPO-R treatment may play a role in improving Treg function, either directly or indirectly by enhanced release of TGF-ß1 as a result of greater platelet turnover. In conclusion, our findings suggest that thrombopoietic agents in patients with ITP have profound effects to restore immune tolerance.


Subject(s)
Receptors, Thrombopoietin/agonists , T-Lymphocytes, Regulatory/immunology , Thrombocytopenia/drug therapy , Thrombocytopenia/immunology , Thrombopoiesis/drug effects , Adolescent , Adult , Aged , Blood Platelets/cytology , CD40 Ligand/blood , Cell Count , Cell Proliferation/drug effects , Chronic Disease , Cohort Studies , Female , Humans , Male , Middle Aged , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta1/blood , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL