28 results on '"Levings MK"'
Search Results
2. Alleviating graft-versus-host disease by directing regulatory T cells to the gut … It is all about location, location, location.
- Author
-
Haque M, Boardman DA, and Levings MK
- Published
- 2023
- Full Text
- View/download PDF
3. Manufacturing next-generation regulatory T-cell therapies.
- Author
-
MacDonald KN, Salim K, and Levings MK
- Subjects
- Humans, T-Lymphocytes, Regulatory metabolism, Cell- and Tissue-Based Therapy
- Abstract
Regulatory T-cell (Treg) therapy has shown promise in treating autoimmune diseases, transplant rejection, or graft-versus-host disease in early clinical trials. These trials have demonstrated that cell therapy using polyclonal Tregs is feasible and safe, however, the field has been limited by the lack of polyclonal cell specificity and consequent large cell numbers required, and the difficulty in generating autologous products for some patients. Thus, the field is moving toward 'next generation' Treg cell therapies that include genetic modification strategies to engineer specificity and/or modify function, as well as methods to generate Tregs in vitro. In this review, we describe how genetic modification of Tregs using viral transduction or gene editing may be incorporated into Treg manufacturing protocols. We also describe how Tregs may be generated via FOXP3 gene editing or overexpression, or by differentiation from pluripotent stem cells. The application of these various types of engineered Tregs is discussed., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
4. Consequences of adjusting cell density and feed frequency on serum-free expansion of thymic regulatory T cells.
- Author
-
MacDonald KN, Hall MG, Ivison S, Gandhi S, Klein Geltink RI, Piret JM, and Levings MK
- Subjects
- Cell Count, Cells, Cultured, Culture Media, Serum-Free pharmacology, Forkhead Transcription Factors metabolism, Humans, Lactates metabolism, Lactates pharmacology, Oxygen metabolism, CD28 Antigens metabolism, T-Lymphocytes, Regulatory
- Abstract
Background: Given the promising results from phase 1/2 clinical trials of therapy involving regulatory T cells (Tregs), it is critical to develop Treg manufacturing methods that use well-defined reagents., Methods: Seeking to maximize expansion of human thymic Tregs activated with anti-CD3/CD28 antibody-coated beads and cultured in serum-free medium, the authors investigated the effect of adjusting process parameters including cell density and cell concentration, and feeding strategy on Treg yield and quality., Results: The authors found that levels of expansion and viability varied with cell density on the day of restimulation. Tregs restimulated at low cell densities (1 × 10
5 cells/cm2 ) initially had high growth rates, viability and FOXP3 expression, but these parameters decreased with time and were less stable than those observed in cultures of Tregs restimulated at high cell densities (5 × 105 cells/cm2 ), which had slower growth rates. High-density expansion was associated with expression of inhibitory molecules and lower intracellular oxygen and extracellular nutrient concentrations as well as extracellular lactate accumulation. Experiments to test the effect of low oxygen revealed that transient exposure to low oxygen levels had little impact on expansion, viability or phenotype. Similarly, blockade of inhibitory molecules had little effect. By contrast, replenishing nutrients by increasing the feeding frequency between 2 days and 4 days after restimulation increased FOXP3, viability and expansion in high-density cultures., Conclusion: These data show the previously undescribed consequences of adjusting cell density on Treg expansion and establish a Good Manufacturing Practice-relevant protocol using non-cell-based activation reagents and serum-free media that supports sustained expansion without loss of viability or phenotype., (Copyright © 2022 International Society for Cell & Gene Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
5. Novel T regulatory cells come of age: The curious incident of a mouse in Tennessee, delayed thymectomy and chimeric receptors!
- Author
-
Scott DW and Levings MK
- Subjects
- Animals, Humans, Mice, T-Lymphocytes, Regulatory immunology, Immune Tolerance immunology, Immunotherapy trends, T-Lymphocytes, Regulatory metabolism
- Published
- 2021
- Full Text
- View/download PDF
6. A method for expansion and retroviral transduction of mouse regulatory T cells.
- Author
-
Wu D, Wong MQ, Vent-Schmidt J, Boardman DA, Steiner TS, and Levings MK
- Subjects
- Adoptive Transfer, Animals, Cells, Cultured, Colitis genetics, Colitis immunology, Colitis metabolism, Colitis prevention & control, Disease Models, Animal, Flow Cytometry, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Genes, T-Cell Receptor beta, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Immunomagnetic Separation, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen immunology, Retroviridae metabolism, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory transplantation, Mice, Cell Proliferation, Genetic Vectors, Receptors, Chimeric Antigen metabolism, Retroviridae genetics, T-Lymphocytes, Regulatory metabolism, Transduction, Genetic
- Abstract
Adoptive cell therapy with genetically modified regulatory T cells (Tregs) is under clinical investigation for the treatment of transplant rejection and various autoimmune conditions. A limitation of modelling this approach in mice is the lack of optimized protocols for expanding and transducing mouse Tregs. Here we describe a protocol for purifying, expanding and retrovirally transducing mouse Tregs with a vector encoding a chimeric antigen receptor as a model transgene. We found that isolation of Tregs from C57Bl/6J Foxp3
E GFP mice solely based on eGFP expression resulted in sufficiently pure cells; co-sorting of CD25hi cells was not essential. Although expansion with rapamycin reduced Treg expansion, it promoted maximal in vitro suppressive activity. Retroviral transduction of Tregs following 2 days of stimulation with anti-CD3/CD28 beads achieved a transduction efficiency of ~40% and did not impair their suppressive capacity. When injected into a conventional T cell (Tconv)-transfer-induced colitis model, transduced Tregs inhibited colitis progression at ratios as low as 1 Treg to 100 Tconvs, and maintained Foxp3 and transgene expression throughout an 8-week period. This method facilitates the study of transduced Tregs in animal models and will enable the study of genetically engineered Treg therapy for a variety of inflammatory diseases., (Copyright © 2020 Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
7. Building a CAR-Treg: Going from the basic to the luxury model.
- Author
-
Rosado-Sánchez I and Levings MK
- Subjects
- Animals, HLA-A2 Antigen immunology, Humans, Immune Tolerance immunology, Organ Transplantation, Receptors, Antigen, T-Cell immunology, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive trends, T-Lymphocytes, Regulatory immunology
- Abstract
Regulatory T cells (Tregs) control immune homeostasis and prevent exacerbated immune responses, and can be used as cell therapy to dampen a variety of autoimmune or autoinflammatory responses. Treg therapy is significantly more effective if the cells are antigen-specific. One way to re-direct the specificity of Tregs is to engineer them to express a Chimeric Antigen Receptor (CAR). Proof-of-concept studies have shown the potential for "basic" models of CAR-Tregs to be used as cellular therapy in autoimmunity, organ transplantation and hematopoietic stem cell transplantation. In parallel, work in the context of cancer has significantly advanced knowledge of how to optimise CAR-T cell structure and function for more precise and potent function. In this review, we summarize the current state of knowledge about important considerations when generating CAR-Tregs. We also extrapolate from emerging findings with CAR-T cells about strategies to further improve CAR-Treg function, creating "luxury" models with refined activity., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
8. Donor-specific chimeric antigen receptor Tregs limit rejection in naive but not sensitized allograft recipients.
- Author
-
Sicard A, Lamarche C, Speck M, Wong M, Rosado-Sánchez I, Blois M, Glaichenhaus N, Mojibian M, and Levings MK
- Subjects
- Allografts, Animals, Graft Rejection prevention & control, Humans, Isoantigens, Mice, T-Lymphocytes, Regulatory, Tissue Donors, Receptors, Chimeric Antigen
- Abstract
Cell therapy with autologous donor-specific regulatory T cells (Tregs) is a promising strategy to minimize immunosuppression in transplant recipients. Chimeric antigen receptor (CAR) technology has recently been used successfully to generate donor-specific Tregs and overcome the limitations of enrichment protocols based on repetitive stimulations with alloantigens. However, the ability of CAR-Treg therapy to control alloreactivity in immunocompetent recipients is unknown. We first analyzed the effect of donor-specific CAR Tregs on alloreactivity in naive, immunocompetent mice receiving skin allografts. Tregs expressing an irrelevant or anti-HLA-A2-specific CAR were administered to Bl/6 mice at the time of transplanting an HLA-A2
+ Bl/6 skin graft. Donor-specific CAR-Tregs, but not irrelevant-CAR Tregs, significantly delayed skin rejection and diminished donor-specific antibodies (DSAs) and frequencies of DSA-secreting B cells. Donor-specific CAR-Treg-treated mice also had a weaker recall DSA response, but normal responses to an irrelevant antigen, demonstrating antigen-specific suppression. When donor-specific CAR Tregs were tested in HLA-A2-sensitized mice, they were unable to delay allograft rejection or diminish DSAs. The finding that donor-specific CAR-Tregs restrain de novo but not memory alloreactivity has important implications for their use as an adoptive cell therapy in transplantation., (© 2020 The American Society of Transplantation and the American Society of Transplant Surgeons.)- Published
- 2020
- Full Text
- View/download PDF
9. Cryopreservation timing is a critical process parameter in a thymic regulatory T-cell therapy manufacturing protocol.
- Author
-
MacDonald KN, Ivison S, Hippen KL, Hoeppli RE, Hall M, Zheng G, Dijke IE, Aklabi MA, Freed DH, Rebeyka I, Gandhi S, West LJ, Piret JM, Blazar BR, and Levings MK
- Subjects
- Cell Culture Techniques methods, Cell Culture Techniques standards, Cell Proliferation, Cell- and Tissue-Based Therapy methods, Cell- and Tissue-Based Therapy standards, Cells, Cultured, Child, Preschool, Cryopreservation standards, Culture Media chemistry, Culture Media pharmacology, Humans, Infant, Lymphocyte Activation, Manufactured Materials standards, T-Lymphocytes, Regulatory immunology, Time Factors, Cryopreservation methods, T-Lymphocytes, Regulatory cytology, T-Lymphocytes, Regulatory transplantation, Thymus Gland cytology, Tissue Engineering methods
- Abstract
Regulatory T cells (Tregs) are a promising therapy for several immune-mediated conditions but manufacturing a homogeneous and consistent product, especially one that includes cryopreservation, has been challenging. Discarded pediatric thymuses are an excellent source of therapeutic Tregs with advantages including cell quantity, homogeneity and stability. Here we report systematic testing of activation reagents, cell culture media, restimulation timing and cryopreservation to develop a Good Manufacturing Practice (GMP)-compatible method to expand and cryopreserve Tregs. By comparing activation reagents, including soluble antibody tetramers, antibody-conjugated beads and artificial antigen-presenting cells (aAPCs) and different media, we found that the combination of Dynabeads Treg Xpander and ImmunoCult-XF medium preserved FOXP3 expression and suppressive function and resulted in expansion that was comparable with a single stimulation with aAPCs. Cryopreservation tests revealed a critical timing effect: only cells cryopreserved 1-3 days, but not >3 days, after restimulation maintained high viability and FOXP3 expression upon thawing. Restimulation timing was a less critical process parameter than the time between restimulation and cryopreservation. This systematic testing of key variables provides increased certainty regarding methods for in vitro expansion and cryopreservation of Tregs. The ability to cryopreserve expanded Tregs will have broad-ranging applications including enabling centralized manufacturing and long-term storage of cell products., (Copyright © 2019 International Society for Cell and Gene Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
10. Tailoring the homing capacity of human Tregs for directed migration to sites of Th1-inflammation or intestinal regions.
- Author
-
Hoeppli RE, MacDonald KN, Leclair P, Fung VCW, Mojibian M, Gillies J, Rahavi SMR, Campbell AIM, Gandhi SK, Pesenacker AM, Reid G, Lim CJ, and Levings MK
- Subjects
- Animals, Cell Adhesion, Cell Movement, Cell Proliferation, Chemokine CXCL10 metabolism, Epigenesis, Genetic, Female, Humans, Immune Tolerance, Immunosuppression Therapy, Integrins metabolism, Interleukin-12 immunology, Male, Mice, Phenotype, Receptors, CCR metabolism, Receptors, CXCR3 metabolism, Thymus Gland immunology, Inflammation immunology, Intestines immunology, T-Lymphocytes, Regulatory cytology, T-Lymphocytes, Regulatory immunology, Th1 Cells cytology
- Abstract
Cell-based therapy with CD4
+ FOXP3+ regulatory T cells (Tregs) is a promising strategy to limit organ rejection and graft-vs-host disease. Ongoing clinical applications have yet to consider how human Tregs could be modified to direct their migration to specific inflammation sites and/or tissues for more targeted immunosuppression. We show here that stable, homing-receptor-tailored human Tregs can be generated from thymic Tregs isolated from pediatric thymus or adult blood. To direct migration to Th1-inflammatory sites, addition of interferon-γ and IL-12 during Treg expansion produced suppressive, epigenetically stable CXCR3+ TBET+ FOXP3+ T helper (Th)1-Tregs. CXCR3 remained expressed after injection in vivo and Th1-Tregs migrated efficiently towards CXCL10 in vitro. To induce tissue-specific migration, addition of retinoic acid (RA) during Treg expansion induced expression of the gut-homing receptors α4β7-integrin and CCR9. FOXP3+ RA-Tregs had elevated expression of the functional markers latency-associated peptide and glycoprotein A repetitions predominant, increased suppressive capacity in vitro and migrated efficiently to healthy and inflamed intestine after injection into mice. Homing-receptor-tailored Tregs were epigenetically stable even after long-term exposure to inflammatory conditions, suppressive in vivo and characterized by Th1- or gut-homing-specific transcriptomes. Tailoring human thymic Treg homing during in vitro expansion offers a new and clinically applicable approach to improving the potency and specificity of Treg therapy., (© 2018 The American Society of Transplantation and the American Society of Transplant Surgeons.)- Published
- 2019
- Full Text
- View/download PDF
11. Engineering therapeutic T cells to suppress alloimmune responses using TCRs, CARs, or BARs.
- Author
-
Sicard A, Levings MK, and Scott DW
- Subjects
- Antigens immunology, Genetic Therapy, Humans, Protein Engineering, Transplantation, Homologous, Receptors, Antigen, T-Cell immunology
- Abstract
Adoptive cell therapy with therapeutic T cells has become one of the most promising strategies to stimulate or suppress immune responses. Using virus-mediated genetic manipulation, the antigen specificity of T cells can now be precisely redirected. Tailored specificity has not only overcome technical limitations and safety concerns but also considerably broadened the spectrum of therapeutic applications. Different T cell-engineering strategies have now become available to suppress alloimmune responses. We first provide an overview of the allorecognition pathways and effector mechanisms that are responsible for alloimmune injuries in the setting of vascularized organ transplantation. We then discuss the potential to use different T cell-engineering approaches to suppress alloimmune responses. Specifically, expression of allospecific T cell receptors, single-chain chimeric antigen receptors, or antigen domains recognized by B cell receptors (B cell antibody receptors) in regulatory or cytotoxic T cells are considered. The ability of these strategies to control the direct or indirect pathways of allorecognition and the cellular or humoral alloimmune responses is discussed. An intimate understanding of the complex interplay that occurs between the engineered T cells and the alloimmune players is a necessary prerequisite for the design of safe and successful strategies for precise immunomodulation in transplantation., (© 2018 The American Society of Transplantation and the American Society of Transplant Surgeons.)
- Published
- 2018
- Full Text
- View/download PDF
12. Biomarker-guided stratification of autoimmune patients for biologic therapy.
- Author
-
Ivison S, Des Rosiers C, Lesage S, Rioux JD, and Levings MK
- Subjects
- Autoimmune Diseases diagnosis, Gene-Environment Interaction, Humans, Patient Selection, Predictive Value of Tests, Prognosis, Reference Standards, Treatment Outcome, Autoimmune Diseases therapy, Biological Therapy, Biomarkers, Pharmacological metabolism
- Abstract
Autoimmunity results from an intersection of genetic and environmental factors that cause patient-specific perturbations in immune homeostasis. Defining autoimmunity-associated genetic factors has led to mechanistic insight into underlying etiologies, and the development of many biologic therapies that target the immune system. However, biomarker-informed pairing of patients with optimal biologic therapy is lacking. Here, we discuss platforms commonly used to find biomarkers that predict response to biologic therapy in autoimmunity and highlight recent biomarker discoveries. We also outline how the lack of assay standardization is a barrier to successful biomarker validation. Finally, we argue that the successful development of companion biomarkers for biologic therapy requires collaborative approaches that integrate multiple platforms and enable comprehensive measurement of multiple immune pathways., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
13. Antigen-specific regulatory T cells: are police CARs the answer?
- Author
-
Dawson NAJ and Levings MK
- Subjects
- Gene Expression Regulation, Neoplastic, Genetic Variation, Receptors, Antigen genetics, Autoimmune Diseases therapy, Graft Rejection therapy, Immunotherapy, Adoptive methods, Receptors, Antigen metabolism, T-Lymphocytes, Regulatory physiology
- Abstract
Cellular therapy with T-regulatory cells (Tregs) is a promising strategy to control immune responses and restore immune tolerance in a variety of immune-mediated diseases, such as transplant rejection and autoimmunity. Multiple clinical trials are currently testing this approach, typically by infusing a single dose of polyclonal Tregs that have been expanded in vitro. However, evidence from animal models of Treg therapy has clearly shown that antigen-specific Tregs are vastly superior to polyclonal cells, meaning that fewer cells are needed for the desired therapeutic effect. Traditional methods to obtain antigen-specific Tregs include antigen-stimulated expansion or T-cell receptor (TCR) overexpression. However, these methods are limited by low cell numbers, complex manufacturing procedures, and knowledge of patient-specific TCRs which recognize disease-relevant MHC-peptide complexes. Recently, several groups have explored the potential to use chimeric antigen receptors (CARs) to generate antigen-specific Tregs. Here, we discuss the progress in this field and highlight the major outstanding questions that remain to be addressed as this approach moves toward clinical applications., (Copyright © 2017. Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
14. The role of FOXP3 in autoimmunity.
- Author
-
Pesenacker AM, Cook L, and Levings MK
- Subjects
- Alternative Splicing, Animals, Autoimmunity, Diabetes Mellitus, Type 1 therapy, Forkhead Transcription Factors genetics, Graft Rejection immunology, Humans, Immunomodulation, Protein Processing, Post-Translational, Protein Transport, Self Tolerance, Signal Transduction, T-Lymphocytes, Regulatory transplantation, Transplantation, Diabetes Mellitus, Type 1 immunology, Forkhead Transcription Factors metabolism, Graft Rejection prevention & control, Immunotherapy methods, T-Lymphocytes, Regulatory immunology
- Abstract
FOXP3 controls the development and function of T regulatory cells (Tregs). Autoimmunity is linked to changes in FOXP3 activity that can occur at multiple levels and lead to Treg dysfunction. For example, changes in IL-2 signaling, FOXP3 transcription and/or post-translational modifications can all contribute to loss of self-tolerance. As additional pathways of FOXP3 regulation are elucidated, new therapeutic approaches to increase Treg activity either by cell therapy or pharmacological intervention are being tested. Early success from pioneering studies of Treg-based therapy in transplantation has promoted the undertaking of similar studies in autoimmunity, with emerging evidence for the effectiveness of these approaches, particularly in the context of type 1 diabetes., (Copyright © 2016. Published by Elsevier Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
15. Obesity-Associated Adipose Tissue Inflammation and Transplantation.
- Author
-
Wu D, Dawson NA, and Levings MK
- Subjects
- Animals, Graft Rejection, Humans, Obesity physiopathology, Panniculitis physiopathology, Adipose Tissue immunology, Obesity complications, Organ Transplantation, Panniculitis etiology
- Abstract
Obesity is often associated with the development of adipose tissue (AT) inflammation, resulting in metabolic dysfunction and an increased risk for developing type 2 diabetes. It is also associated with multiple chronic diseases, including cardiovascular, liver, and kidney disease, and thus can contribute to organ failure. Several studies have investigated whether there is a correlation between obesity and outcomes in transplantation, but there is currently very limited information on the specific role of AT inflammation in the rejection process or on the overall function of the transplanted organ. Here, we provide a brief review of the current understanding of the cellular mechanisms that control obesity-associated AT inflammation and summarize knowledge about how obesity affects clinical outcomes following solid organ or hematopoietic stem cell transplantation. We also highlight opportunities for more research to better understand how obesity affects outcomes of transplantation., (© Copyright 2015 The American Society of Transplantation and the American Society of Transplant Surgeons.)
- Published
- 2016
- Full Text
- View/download PDF
16. Discarded Human Thymus Is a Novel Source of Stable and Long-Lived Therapeutic Regulatory T Cells.
- Author
-
Dijke IE, Hoeppli RE, Ellis T, Pearcey J, Huang Q, McMurchy AN, Boer K, Peeters AM, Aubert G, Larsen I, Ross DB, Rebeyka I, Campbell A, Baan CC, Levings MK, and West LJ
- Subjects
- Adult, Animals, Cells, Cultured, Child, Female, Flow Cytometry, Graft vs Host Disease immunology, Humans, Lymphocyte Activation, Mice, Mice, Inbred NOD, Mice, SCID, Middle Aged, Telomere Homeostasis, Thymus Gland immunology, Thymus Gland metabolism, Forkhead Transcription Factors metabolism, Graft vs Host Disease therapy, Interleukin-2 Receptor alpha Subunit metabolism, T-Lymphocytes, Regulatory immunology, Thymus Gland cytology
- Abstract
Regulatory T cell (Treg)-based therapy is a promising approach to treat many immune-mediated disorders such as autoimmune diseases, organ transplant rejection, and graft-versus-host disease (GVHD). Challenges to successful clinical implementation of adoptive Treg therapy include difficulties isolating homogeneous cell populations and developing expansion protocols that result in adequate numbers of cells that remain stable, even under inflammatory conditions. We investigated the potential of discarded human thymuses, routinely removed during pediatric cardiac surgery, to be used as a novel source of therapeutic Tregs. Here, we show that large numbers of FOXP3(+) Tregs can be isolated and expanded from a single thymus. Expanded thymic Tregs had stable FOXP3 expression and long telomeres, and suppressed proliferation and cytokine production of activated allogeneic T cells in vitro. Moreover, expanded thymic Tregs delayed development of xenogeneic GVHD in vivo more effectively than expanded Tregs isolated based on CD25 expression from peripheral blood. Importantly, in contrast to expanded blood Tregs, expanded thymic Tregs remained stable under inflammatory conditions. Our results demonstrate that discarded pediatric thymuses are an excellent source of therapeutic Tregs, having the potential to overcome limitations currently hindering the use of Tregs derived from peripheral or cord blood., (© Copyright 2015 The American Society of Transplantation and the American Society of Transplant Surgeons.)
- Published
- 2016
- Full Text
- View/download PDF
17. A novel function for FOXP3 in humans: intrinsic regulation of conventional T cells.
- Author
-
McMurchy AN, Gillies J, Gizzi MC, Riba M, Garcia-Manteiga JM, Cittaro D, Lazarevic D, Di Nunzio S, Piras IS, Bulfone A, Roncarolo MG, Stupka E, Bacchetta R, and Levings MK
- Subjects
- CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes physiology, Cell Lineage immunology, Cell Proliferation, Clone Cells cytology, Clone Cells physiology, Female, Forkhead Transcription Factors metabolism, Gene Expression Regulation immunology, Humans, Interferon-gamma metabolism, Interleukin-2 metabolism, Male, RNA, Small Interfering, Receptors, CCR4 genetics, Receptors, CCR4 metabolism, Th1 Cells metabolism, Th17 Cells metabolism, Transcriptome, Forkhead Transcription Factors genetics, Forkhead Transcription Factors physiology, Th1 Cells physiology, Th17 Cells physiology
- Abstract
The role of forkhead box P3 (FOXP3) is well-established in T-regulatory cells, but the function of transient FOXP3 expression in activated human conventional T (Tconv) cells is unknown. In the present study, we used 2 approaches to determine the role of FOXP3 in human Tconv cells. First, we obtained Tconv clones from a female subject who is hemizygous for a null mutation in FOXP3, allowing the comparison of autologous T-cell clones that do or do not express FOXP3. Second, we knocked down activation-induced FOXP3 in Tconv cells from healthy donors with small interfering RNAagainst FOXP3. We found that FOXP3-deficient Tconv cells proliferate more and produce more cytokines than wild-type Tconv cells and have differential expression of 274 genes. We also investigated the role of FOXP3 in Th1 and Th17 cells and found that the expression of activation-induced FOXP3 was higher and more sustained in Th17 cells compared with Th1 cells. Knocking down FOXP3 expression in Th17 cells significantly increased the production of IFN-γ and decreased the expression of CCR4, but had no effect on IL-17 expression. These data reveal a novel function of FOXP3 in Tconv cells and suggest that expression of this protein is important in the function of multiple CD4(+) T-cell lineages.
- Published
- 2013
- Full Text
- View/download PDF
18. Cellular magnetic resonance imaging of monocyte-derived dendritic cell migration from healthy donors and cancer patients as assessed in a scid mouse model.
- Author
-
Zhang X, de Chickera SN, Willert C, Economopoulos V, Noad J, Rohani R, Wang AY, Levings MK, Scheid E, Foley R, Foster PJ, and Dekaban GA
- Subjects
- Animals, Antigens, Differentiation metabolism, Cells, Cultured, Dendritic Cells cytology, Dendritic Cells transplantation, Disease Models, Animal, Feasibility Studies, Humans, Magnetic Resonance Imaging methods, Mice, Mice, SCID, Monocytes cytology, Neoplasms immunology, Neoplasms pathology, Cancer Vaccines, Cell Movement, Dendritic Cells metabolism, Immunotherapy, Adoptive, Neoplasms therapy
- Abstract
BACKGROUND AIMS. The use of dendritic cells (DC) as an adjuvant in cell-based immunotherapeutic cancer vaccines is a growing field of interest. A reliable and non-invasive method to track the fate of autologous DC following their administration to patients is required in order to confirm that clinically sufficient numbers are reaching the lymph node (LN). We demonstrate that an immunocompromised mouse model can be used to conduct translational studies employing cellular magnetic resonance imaging (MRI). Such studies can provide clinically relevant information regarding the migration potential of clinical-grade DC used in cancer immunotherapies. METHODS. Human monocyte-derived dendritic cells (mo-DC) were generated from negatively selected monocytes obtained from either healthy donors or cancer patients. DC were labeled with superparamagnetic iron oxide (SPIO) nanoparticles in order to track them in vivo in a CB17scid mouse model using cellular MRI. SPIO did not have any adverse effects on DC phenotype or function, independent of donor type. Cellular MRI readily detected migration of SPIO-loaded DC in CB17scid mice. No differences in migration were observed between DC obtained from healthy donors and those obtained from donors undergoing autologous stem cell transplant for cancer therapy. CONCLUSIONS. Cellular MRI provided semi-quantitative image data that corresponded with data obtained by digital morphometry, validating cellular MRI's potential to assess DC migration in DC-based cancer immunotherapy clinical trials.
- Published
- 2011
- Full Text
- View/download PDF
19. Cutaneous GVHD is associated with the expansion of tissue-localized Th1 and not Th17 cells.
- Author
-
Broady R, Yu J, Chow V, Tantiworawit A, Kang C, Berg K, Martinka M, Ghoreishi M, Dutz J, and Levings MK
- Subjects
- Adult, Case-Control Studies, Female, Flow Cytometry, Humans, Immunoenzyme Techniques, Male, Middle Aged, Prospective Studies, T-Lymphocytes immunology, Young Adult, Graft vs Host Disease immunology, Skin Diseases immunology, Th1 Cells immunology, Th17 Cells immunology
- Abstract
Studies in mice have shown that proinflammatory Th17 cells can cause acute graft-versus-host disease (aGVHD) related tissue damage; however, whether they play a role in human aGVHD remains unclear. In a prospective study, we measured the proportion of Th17 cells in the blood and skin of patients at the onset of aGVHD. We found no difference in the proportion or amount of IL-17 produced by T cells in the blood of patients with aGVHD (n = 20) compared with time-matched patients without GVHD (n = 14). Moreover, Th17 cells were not increased in the skin of patients with cutaneous aGVHD (n = 7) compared with healthy controls (n = 10). In contrast, we found significantly more interferon-γ-producing T cells in the skin of patients with aGVHD compared with controls. These data support the long-standing paradigm that tissue localized interferon-γ-producing cells are the perpetrators of aGVHD.
- Published
- 2010
- Full Text
- View/download PDF
20. ATG-induced expression of FOXP3 in human CD4(+) T cells in vitro is associated with T-cell activation and not the induction of FOXP3(+) T regulatory cells.
- Author
-
Broady R, Yu J, and Levings MK
- Subjects
- Animals, Antilymphocyte Serum immunology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Cytokines biosynthesis, Cytokines immunology, Flow Cytometry, Humans, Immunosuppressive Agents immunology, Interleukin-2 Receptor alpha Subunit biosynthesis, Interleukin-2 Receptor alpha Subunit immunology, Lymphocyte Activation immunology, Rabbits, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism, Antilymphocyte Serum pharmacology, CD4-Positive T-Lymphocytes drug effects, Forkhead Transcription Factors biosynthesis, Immunosuppressive Agents pharmacology, Lymphocyte Activation drug effects, T-Lymphocyte Subsets drug effects
- Abstract
Several recent reports have suggested that in vitro exposure of CD4(+) T cells to rabbit antithymocyte globulin (rATG), which is commonly used to prevent and treat graft-versus-host disease and allograft rejection, is an effective method to induce CD4(+)CD25(+)FOXP3(+) T regulatory cells (Tregs). We and others, however, have shown that FOXP3 is also expressed in activated T cells. We therefore investigated whether the induction of FOXP3 expression by rATG resulted in a stable population of suppressive Tregs. We found that exposure of peripheral blood mononuclear cells (PBMCs) or conventional T cells to rATG resulted in induction of transient rather than stable expression of CD25 and FOXP3. Furthermore, rATG-treated T effector cells acquired neither an immunosuppressive profile of cytokine production nor suppressive capacity, even at the time of maximal FOXP3 expression. These findings indicate that the notion that rATG can be used to induce Tregs in vitro for cellular therapy in vivo should be re-evaluated.
- Published
- 2009
- Full Text
- View/download PDF
21. Wild-type FOXP3 is selectively active in CD4+CD25(hi) regulatory T cells of healthy female carriers of different FOXP3 mutations.
- Author
-
Di Nunzio S, Cecconi M, Passerini L, McMurchy AN, Baron U, Turbachova I, Vignola S, Valencic E, Tommasini A, Junker A, Cazzola G, Olek S, Levings MK, Perroni L, Roncarolo MG, and Bacchetta R
- Subjects
- Adult, Autoimmune Diseases genetics, Autoimmune Diseases immunology, Autoimmune Diseases metabolism, Autoimmune Diseases pathology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes pathology, Case-Control Studies, Cell Differentiation, Female, Forkhead Transcription Factors immunology, Genes, X-Linked, Genetic Diseases, X-Linked genetics, Genetic Diseases, X-Linked immunology, Genetic Diseases, X-Linked metabolism, Genetic Diseases, X-Linked pathology, Heterozygote, Humans, Male, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, T-Lymphocyte Subsets pathology, T-Lymphocytes, Regulatory pathology, X Chromosome Inactivation, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Mutation, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism
- Abstract
Forkhead box P3 (FOXP3) is constitutively expressed by CD4(+)CD25(hi) regulatory T cells (nTregs). Mutations of FOXP3 cause a severe autoimmune syndrome known as immune dysregulation polyendocrinopathy enteropathy X-linked, in which nTregs are absent or dysfunctional. Whether FOXP3 is essential for both differentiation and function of human nTreg cells remains to be demonstrated. Because FOXP3 is an X-linked gene subject to X-chromosome inactivation (XCI), we studied 9 healthy female carriers of FOXP3 mutations to investigate the role of wild-type (WT) versus mutated FOXP3 in different cell subsets. Analysis of active WT versus mutated (mut)-FOXP3 allele distribution revealed a random pattern of XCI in peripheral blood lymphocytes and in naive and memory CD4(+)T cells, whereas nTregs expressed only the active WT-FOXP3. These data demonstrate that expression of WT-FOXP3 is indispensable for the presence of a normal nTreg compartment and suggest that FOXP3 is not necessary for effector T-cell differentiation in humans.
- Published
- 2009
- Full Text
- View/download PDF
22. MASTering Treg function to promote tolerance.
- Author
-
Levings MK and Thomson AW
- Subjects
- CD4-Positive T-Lymphocytes immunology, Graft Rejection immunology, Graft Survival immunology, Humans, Immune Tolerance, T-Lymphocytes, Regulatory immunology
- Published
- 2009
- Full Text
- View/download PDF
23. Flow cytometry-based methods for studying signaling in human CD4+CD25+FOXP3+ T regulatory cells.
- Author
-
Crellin NK, Garcia RV, and Levings MK
- Subjects
- Cell Line, Cell Line, Tumor, Cell Proliferation, Coculture Techniques, Humans, Immunophenotyping, Intracellular Fluid immunology, Intracellular Fluid metabolism, Lymphocyte Activation immunology, Phosphoproteins analysis, Phosphoproteins biosynthesis, Phosphoproteins metabolism, Phosphorylation, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, T-Lymphocytes, Regulatory cytology, T-Lymphocytes, Regulatory metabolism, Flow Cytometry methods, Forkhead Transcription Factors biosynthesis, Signal Transduction immunology, T-Lymphocytes, Regulatory immunology
- Abstract
T regulatory (Treg) cells have a fundamental role in the establishment and maintenance of peripheral tolerance. It is well established that Treg cells have a phenotype and function that is distinct from conventional T effector cells, although how these two T cell subsets differ in terms of molecular signaling cascades remains largely unknown. Analysis of signaling events in Treg cells using classical biochemistry has been hampered due to difficulties in isolating homogeneous populations and limited cell numbers. In order to overcome these challenges, we defined the optimal conditions for culture, in vitro expansion, and stimulation of human CD4(+)CD25(+) Treg and T effector cells to study intracellular signaling events by flow cytometry. In order to avoid the pitfalls associated with cell isolation based on CD25 expression, we developed methodology to analyze subpopulations of FOXP3 positive and negative cells from ex vivo CD4(+) T cells. In addition to examination of ex vivo cells, we optimized expansion conditions for analysis of signaling in Treg and T effector cell lines. Using these methods, we found that human FOXP3(+) Treg cells displayed a greater capacity to phosphorylate the extracellular regulated kinase (ERK) compared to T effector cells, upon TCR-mediated activation. In contrast, FOXP3(+) Treg cells showed a significantly diminished capacity to phosphorylate AKT. This methodology provides a foundation for future investigation into the molecular events that regulate the phenotype and function of Treg cells, and may ultimately lead to the identification of Treg-cell specific therapeutic targets.
- Published
- 2007
- Full Text
- View/download PDF
24. Altered activation of AKT is required for the suppressive function of human CD4+CD25+ T regulatory cells.
- Author
-
Crellin NK, Garcia RV, and Levings MK
- Subjects
- Antigens, CD metabolism, Antigens, Differentiation metabolism, CD4-Positive T-Lymphocytes enzymology, CTLA-4 Antigen, Cytokines biosynthesis, Enzyme Activation, Forkhead Transcription Factors metabolism, Granzymes metabolism, Humans, Mitogen-Activated Protein Kinases metabolism, Phosphorylation, Receptors, Antigen, T-Cell immunology, Signal Transduction, T-Lymphocytes, Regulatory enzymology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Interleukin-2 Receptor alpha Subunit metabolism, Proto-Oncogene Proteins c-akt metabolism, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism
- Abstract
Suppression by T regulatory cells (Treg cells) is a major mechanism by which the immune system controls responses to self and nonharmful foreign proteins. Although there are many different types of Treg cells, the best characterized are those that constitutively express cell-surface IL-2Ralpha (CD25). We investigated whether altered T-cell-receptor (TCR)-mediated signaling in pure populations of ex vivo human CD4+CD25+ Treg cells might underlie their unique phenotype, including hyporesponsiveness to TCR-mediated activation and lack of cytokine production. CD4+CD25+ Treg cells displayed a consistent defect in phosphorylation of AKT at serine 473 and reduced phosphorylation of the AKT substrates FOXO and S6. Restoration of AKT activity via lentiviral-mediated expression of an inducibly active form of the kinase revealed that reduced activity of this pathway was necessary for the suppressive function of CD4+CD25+ Treg cells. These data represent the first demonstration of a causal association between altered signaling and the function of CD4+CD25+ Treg cells. Moreover, we have created the first system allowing inducible abrogation of suppression through manipulation of the suppressor cells. This system will be a powerful tool to further study the mechanism(s) of suppression by CD4+CD25+ Treg cells.
- Published
- 2007
- Full Text
- View/download PDF
25. Differentiation of Tr1 cells by immature dendritic cells requires IL-10 but not CD25+CD4+ Tr cells.
- Author
-
Levings MK, Gregori S, Tresoldi E, Cazzaniga S, Bonini C, and Roncarolo MG
- Subjects
- CD4 Antigens immunology, Cell Differentiation drug effects, Cell Differentiation immunology, Cell Division, Humans, Immunophenotyping, Receptors, Interleukin-2 immunology, Reference Values, T-Lymphocytes cytology, T-Lymphocytes drug effects, CD4-Positive T-Lymphocytes immunology, Dendritic Cells immunology, Interleukin-10 pharmacology, Lymphocyte Activation immunology, T-Lymphocytes immunology
- Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that monitor the antigenic environment and activate naive T cells. The role of DCs is not only to sense danger but also to tolerize the immune system to antigens encountered in the absence of maturation/inflammatory stimuli. Indeed, if a naive T cell encounters its antigen on immature DCs (iDCs), it may differentiate into a T-regulatory (Tr) rather than a T-effector cell. However, little is known about the mechanisms by which iDCs differentiate Tr cells. We developed a standardized and highly reproducible protocol to differentiate Tr cells by repetitive exposure of naive peripheral blood CD4(+) T cells to allogeneic iDCs. The resultant Tr cells are phenotypically and functionally identical to type 1 Tr (Tr1) cells because their generation requires production of IL-10 by iDCs, and they suppress T-cell responses through an interleukin-10 (IL-10)- and a transforming growth factor beta (TGF-beta)-dependent mechanism. In addition, Tr1 cells induced by iDCs do not require the presence of CD4(+)CD25(+) Tr cells for their generation, nor do they express high constitutive levels of CD25 or the transcription factor FoxP3. Thus, iDCs can drive the differentiation of Tr1 cells and can be used to generate large numbers of alloantigen-specific Tr1 cells for clinical use as a cellular therapy to restore peripheral tolerance.
- Published
- 2005
- Full Text
- View/download PDF
26. The role of different subsets of T regulatory cells in controlling autoimmunity.
- Author
-
Roncarolo MG and Levings MK
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, Cell Differentiation, Clonal Anergy immunology, Cytokines immunology, Humans, Leukocyte Common Antigens immunology, Receptors, Interleukin-2 immunology, T-Lymphocytes, Helper-Inducer immunology, Autoimmunity immunology, CD4-Positive T-Lymphocytes immunology, T-Lymphocyte Subsets immunology
- Abstract
T regulatory cells-in addition to clonal deletion and anergy-are essential for the downregulation of T cell responses to both foreign and self antigens, and for the prevention of autoimmunity. Recent progress has been made in characterising the different subsets of T regulatory cells, the factors that drive their differentiation, and their mode of action. The resolution of these mechanisms will make it possible to use T regulatory cells therapeutically in human autoimmune diseases.
- Published
- 2000
- Full Text
- View/download PDF
27. Heterodimerization of the alpha and beta chains of the interleukin-3 (IL-3) receptor is necessary and sufficient for IL-3-induced mitogenesis.
- Author
-
Orban PC, Levings MK, and Schrader JW
- Subjects
- Animals, Cell Line, Dimerization, Humans, Mice, Mitosis drug effects, Receptors, Interleukin-3 chemistry, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins physiology, Structure-Activity Relationship, Transfection, Interleukin-3 pharmacology, Mitosis physiology, Receptors, Interleukin-3 physiology, Signal Transduction drug effects
- Abstract
The high-affinity receptor for interleukin-3 (IL-3) is a complex of the IL-3-binding subunit (alpha(IL-3)) and a larger beta chain-beta(c), or, in the mouse, beta(c) or its close relative beta(IL-3). There is evidence that the critical event that initiates signaling is not the approximation of the cytoplasmic domains of alpha(IL-3) and beta(IL-3), but is, rather, the formation of a beta-beta homodimer. Many of these studies involved the analyses of receptor chimeras where the cytoplasmic domains were derived from alpha(IL-3), beta(c) or beta(IL-3), and the extracellular domains were derived from other cytokine receptors, such as the erythropoietin receptor (EpoR). However, evidence that the EpoR may also associate with other receptors clouds the interpretation of these experiments. Therefore, we reevaluated the structure of the functional IL-3R using chimeric receptors with extracellular domains derived not from members of the cytokine-receptor family, but from CD8 or CD16. We show, by expression of these chimeras in Ba/F3 or CTLL-2 cells, that mitogenic signals were only generated by heterodimerization of the cytoplasmic domains of alpha(IL-3) and beta(IL-3). Homodimers of either alpha(IL-3) or beta(IL-3), alone or in combination, were nonfunctional. Furthermore, the ability of heterodimers to stimulate mitogenesis correlated with their ability to induce tyrosine phosphorylation of JAK-2. These data suggest that the physiological activation of the IL-3R involves the generation of simple heterodimers of alpha(IL-3) and beta(IL-3).
- Published
- 1999
28. Interleukin-4 synergizes with Raf-1 to promote long-term proliferation and activation of c-jun N-terminal kinase.
- Author
-
Levings MK, Bessette DC, and Schrader JW
- Subjects
- Animals, Cell Division drug effects, Cell Division physiology, Cell Line, Hematopoiesis physiology, JNK Mitogen-Activated Protein Kinases, Calcium-Calmodulin-Dependent Protein Kinases physiology, Hematopoiesis drug effects, Interleukin-4 pharmacology, Mitogen-Activated Protein Kinases, Proto-Oncogene Proteins c-raf physiology, Signal Transduction drug effects, ras Proteins physiology
- Abstract
This report shows that interleukin-4 (IL-4), which plays a key role in regulating immune responses, fails to support cellular growth. We investigated whether this failure of IL-4 to promote growth was because of its unique inability to activate the Ras/Raf/Erk pathway. Consistent with other reports, expression in Ba/F3, a factor-dependent hematopoietic cell line, of either activated Q61KN-Ras or a hormone-inducible activated Raf-1, resulted in suppression of apoptosis but not in long-term growth. However, in the presence of IL-4, Ba/F3 cells that expressed either Q61KN-Ras or activated Raf-1 grew continuously at a rate comparable with that stimulated by IL-3. Investigation of the biochemical events associated with the stimulation of long-term growth showed that, as expected, the presence of activated Raf-1 resulted in an increased activity of extracellular signal regulated kinase (ERK) mitogen-activated protein kinase (MAPK) but not of c-jun N-terminal kinase/stress-activated protein kinase (JNK). However, surprisingly, if IL-4 was present, cells expressing active Raf-1 exhibited increases in JNK activity. These observations point to a novel mechanism for JNK activation involving synergy between Raf-1 and pathways activated by IL-4 and suggest that in hematopoietic cells proliferation is correlated not only with "mitogen activated" ERK activity, but also with JNK activity.
- Published
- 1999
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.