Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 4(10): 101232, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37852182

ABSTRACT

In this issue of Cell Reports Medicine, Qin et al.1 present a comprehensive single-cell transcriptomics analysis of the tumor microenvironment of rectal cancer tumors before and after neoadjuvant chemotherapy.


Subject(s)
Neoadjuvant Therapy , Rectal Neoplasms , Humans , Neoplasm Staging , Rectal Neoplasms/drug therapy , Rectal Neoplasms/pathology , Tumor Microenvironment
2.
Nat Commun ; 14(1): 4703, 2023 08 05.
Article in English | MEDLINE | ID: mdl-37543621

ABSTRACT

TGFß signaling is associated with non-response to immune checkpoint blockade in patients with advanced cancers, particularly in the immune-excluded phenotype. While previous work demonstrates that converting tumors from excluded to inflamed phenotypes requires attenuation of PD-L1 and TGFß signaling, the underlying cellular mechanisms remain unclear. Here, we show that TGFß and PD-L1 restrain intratumoral stem cell-like CD8 T cell (TSCL) expansion and replacement of progenitor-exhausted and dysfunctional CD8 T cells with non-exhausted T effector cells in the EMT6 tumor model in female mice. Upon combined TGFß/PD-L1 blockade IFNγhi CD8 T effector cells show enhanced motility and accumulate in the tumor. Ensuing IFNγ signaling transforms myeloid, stromal, and tumor niches to yield an immune-supportive ecosystem. Blocking IFNγ abolishes the anti-PD-L1/anti-TGFß therapy efficacy. Our data suggest that TGFß works with PD-L1 to prevent TSCL expansion and replacement of exhausted CD8 T cells, thereby maintaining the T cell compartment in a dysfunctional state.


Subject(s)
B7-H1 Antigen , Breast Neoplasms , CD8-Positive T-Lymphocytes , Immune Checkpoint Inhibitors , Transforming Growth Factor beta , Female , Animals , Mice , Cell Differentiation , CD8-Positive T-Lymphocytes/immunology , Stem Cells , B7-H1 Antigen/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Interferon-gamma/immunology , T-Cell Exhaustion , Immune Checkpoint Inhibitors/pharmacology , Mice, Inbred BALB C , Cell Line, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , RNA-Seq
3.
J Exp Med ; 220(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-36820828

ABSTRACT

Coordinated gene expression programs enable development and function of T cell subsets. Follicular helper T (Tfh) cells coordinate humoral immune responses by providing selective and instructive cues to germinal center B cells. Here, we show that AP-1-independent NFAT gene expression, a program associated with hyporesponsive T cell states like anergy or exhaustion, is also a distinguishing feature of Tfh cells. NFAT signaling in Tfh cells, maintained by NFAT2 autoamplification, is required for their survival. ICOS signaling upregulates Bcl6 and induces an AP-1-independent NFAT program in primary T cells. Using lupus-prone mice, we demonstrate that genetic disruption or pharmacologic inhibition of NFAT signaling specifically impacts Tfh cell maintenance and leads to amelioration of autoantibody production and renal injury. Our data provide important conceptual and therapeutic insights into the signaling mechanisms that regulate Tfh cell development and function.


Subject(s)
Autoimmunity , T-Lymphocytes, Helper-Inducer , Mice , Animals , Transcription Factor AP-1/metabolism , B-Lymphocytes , Germinal Center , Cell Differentiation
4.
Nature ; 611(7934): 148-154, 2022 11.
Article in English | MEDLINE | ID: mdl-36171287

ABSTRACT

Recent single-cell studies of cancer in both mice and humans have identified the emergence of a myofibroblast population specifically marked by the highly restricted leucine-rich-repeat-containing protein 15 (LRRC15)1-3. However, the molecular signals that underlie the development of LRRC15+ cancer-associated fibroblasts (CAFs) and their direct impact on anti-tumour immunity are uncharacterized. Here in mouse models of pancreatic cancer, we provide in vivo genetic evidence that TGFß receptor type 2 signalling in healthy dermatopontin+ universal fibroblasts is essential for the development of cancer-associated LRRC15+ myofibroblasts. This axis also predominantly drives fibroblast lineage diversity in human cancers. Using newly developed Lrrc15-diphtheria toxin receptor knock-in mice to selectively deplete LRRC15+ CAFs, we show that depletion of this population markedly reduces the total tumour fibroblast content. Moreover, the CAF composition is recalibrated towards universal fibroblasts. This relieves direct suppression of tumour-infiltrating CD8+ T cells to enhance their effector function and augments tumour regression in response to anti-PDL1 immune checkpoint blockade. Collectively, these findings demonstrate that TGFß-dependent LRRC15+ CAFs dictate the tumour-fibroblast setpoint to promote tumour growth. These cells also directly suppress CD8+ T cell function and limit responsiveness to checkpoint blockade. Development of treatments that restore the homeostatic fibroblast setpoint by reducing the population of pro-disease LRRC15+ myofibroblasts may improve patient survival and response to immunotherapy.


Subject(s)
Cancer-Associated Fibroblasts , Membrane Proteins , Myofibroblasts , Pancreatic Neoplasms , Stromal Cells , Animals , Humans , Mice , Cancer-Associated Fibroblasts/metabolism , CD8-Positive T-Lymphocytes/immunology , Membrane Proteins/metabolism , Myofibroblasts/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Receptors, Transforming Growth Factor beta , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , B7-H1 Antigen
5.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33446502

ABSTRACT

Haematopoiesis relies on tightly controlled gene expression patterns as development proceeds through a series of progenitors. While the regulation of hematopoietic development has been well studied, the role of noncoding elements in this critical process is a developing field. In particular, the discovery of new regulators of lymphopoiesis could have important implications for our understanding of the adaptive immune system and disease. Here we elucidate how a noncoding element is capable of regulating a broadly expressed transcription factor, Ikaros, in a lymphoid lineage-specific manner, such that it imbues Ikaros with the ability to specify the lymphoid lineage over alternate fates. Deletion of the Daedalus locus, which is proximal to Ikaros, led to a severe reduction in early lymphoid progenitors, exerting control over the earliest fate decisions during lymphoid lineage commitment. Daedalus locus deletion led to alterations in Ikaros isoform expression and a significant reduction in Ikaros protein. The Daedalus locus may function through direct DNA interaction as Hi-C analysis demonstrated an interaction between the two loci. Finally, we identify an Ikaros-regulated erythroid-lymphoid checkpoint that is governed by Daedalus in a lymphoid-lineage-specific manner. Daedalus appears to act as a gatekeeper of Ikaros's broad lineage-specifying functions, selectively stabilizing Ikaros activity in the lymphoid lineage and permitting diversion to the erythroid fate in its absence. These findings represent a key illustration of how a transcription factor with broad lineage expression must work in concert with noncoding elements to orchestrate hematopoietic lineage commitment.


Subject(s)
Hematopoiesis/genetics , Ikaros Transcription Factor/genetics , Lymphopoiesis/genetics , RNA, Untranslated/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Gene Expression Regulation, Developmental/genetics , Mice
6.
Cell Res ; 30(11): 1053, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33028962

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Cell Res ; 30(8): 649-659, 2020 08.
Article in English | MEDLINE | ID: mdl-32709897

ABSTRACT

The maintenance of organismal homeostasis requires partitioning and transport of biochemical molecules between organ systems, their composite cells, and subcellular organelles. Although transcriptional programming undeniably defines the functional state of cells and tissues, underlying biochemical networks are intricately intertwined with transcriptional, translational, and post-translational regulation. Studies of the metabolic regulation of immunity have elegantly illustrated this phenomenon. The cells of the immune system interface with a diverse set of environmental conditions. Circulating immune cells perfuse peripheral organs in the blood and lymph, patrolling for pathogen invasion. Resident immune cells remain in tissues and play more newly appreciated roles in tissue homeostasis and immunity. Each of these cell populations interacts with unique and dynamic tissue environments, which vary greatly in biochemical composition. Furthermore, the effector response of immune cells to a diverse set of activating cues requires unique cellular adaptations to supply the requisite biochemical landscape. In this review, we examine the role of spatial partitioning of metabolic processes in immune function. We focus on studies of lymphocyte metabolism, with reference to the greater immunometabolism literature when appropriate to illustrate this concept.


Subject(s)
Signal Transduction , T-Lymphocytes/metabolism , Animals , Gene Expression Regulation , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mitochondria/metabolism , Models, Biological , T-Lymphocytes/immunology
8.
J Exp Med ; 217(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32525985

ABSTRACT

CD8+ tissue-resident memory T cells (TRM cells) are poised at the portals of infection and provide long-term protective immunity. Despite their critical roles, the precise mechanics governing TRM cell reactivation in situ are unknown. Using a TCR-transgenic Nur77-GFP reporter to distinguish "antigen-specific" from "bystander" reactivation, we demonstrate that lung CD8+ TRM cells are reactivated more quickly, yet less efficiently, than their counterparts in the draining LNs (TLN cells). Global profiling of reactivated memory T cells revealed tissue-defined and temporally regulated recall response programs. Unlike the reactivation of CD8+ TLN cells, which is strictly dependent on CD11c+XCR1+ APCs, numerous antigen-presenting partners, both hematopoietic and non-hematopoietic, were sufficient to reactivate lung CD8+ TRM cells, but the quality of TRM cell functional responses depended on the identity of the APCs. Together, this work uncovers fundamental differences in the activation kinetics, mechanics, and effector responses between CD8+ memory T cells in peripheral vs. lymphoid organs, revealing a novel tissue-specific paradigm for the reactivation of memory CD8+ T cells.


Subject(s)
Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lung/immunology , Lymph Nodes/immunology , Lymphocyte Activation , Animals , CD11 Antigens/genetics , CD11 Antigens/immunology , Mice , Mice, Knockout , Organ Specificity/genetics , Organ Specificity/immunology , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology
9.
Sci Transl Med ; 12(538)2020 04 08.
Article in English | MEDLINE | ID: mdl-32269165

ABSTRACT

The kidney is a frequent target of autoimmune injury, including in systemic lupus erythematosus; however, how immune cells adapt to kidney's unique environment and contribute to tissue damage is unknown. We found that renal tissue, which normally has low oxygen tension, becomes more hypoxic in lupus nephritis. In the injured mouse tissue, renal-infiltrating CD4+ and CD8+ T cells express hypoxia-inducible factor-1 (HIF-1), which alters their cellular metabolism and prevents their apoptosis in hypoxia. HIF-1-dependent gene-regulated pathways were also up-regulated in renal-infiltrating T cells in human lupus nephritis. Perturbation of these environmental adaptations by selective HIF-1 blockade inhibited infiltrating T cells and reversed tissue hypoxia and injury in murine models of lupus. The results suggest that targeting HIF-1 might be effective for treating renal injury in autoimmune diseases.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Animals , CD8-Positive T-Lymphocytes , Hypoxia , Kidney , Mice
11.
Nature ; 571(7765): 403-407, 2019 07.
Article in English | MEDLINE | ID: mdl-31217581

ABSTRACT

Activated CD4 T cells proliferate rapidly and remodel epigenetically before exiting the cell cycle and engaging acquired effector functions. Metabolic reprogramming from the naive state is required throughout these phases of activation1. In CD4 T cells, T-cell-receptor ligation-along with co-stimulatory and cytokine signals-induces a glycolytic anabolic program that is required for biomass generation, rapid proliferation and effector function2. CD4 T cell differentiation (proliferation and epigenetic remodelling) and function are orchestrated coordinately by signal transduction and transcriptional remodelling. However, it remains unclear whether these processes are regulated independently of one another by cellular biochemical composition. Here we demonstrate that distinct modes of mitochondrial metabolism support differentiation and effector functions of mouse T helper 1 (TH1) cells by biochemically uncoupling these two processes. We find that the tricarboxylic acid cycle is required for the terminal effector function of TH1 cells through succinate dehydrogenase (complex II), but that the activity of succinate dehydrogenase suppresses TH1 cell proliferation and histone acetylation. By contrast, we show that complex I of the electron transport chain, the malate-aspartate shuttle and mitochondrial citrate export are required to maintain synthesis of aspartate, which is necessary for the proliferation of T helper cells. Furthermore, we find that mitochondrial citrate export and the malate-aspartate shuttle promote histone acetylation, and specifically regulate the expression of genes involved in T cell activation. Combining genetic, pharmacological and metabolomics approaches, we demonstrate that the differentiation and terminal effector functions of T helper cells are biochemically uncoupled. These findings support a model in which the malate-aspartate shuttle, mitochondrial citrate export and complex I supply the substrates needed for proliferation and epigenetic remodelling early during T cell activation, whereas complex II consumes the substrates of these pathways, which antagonizes differentiation and enforces terminal effector function. Our data suggest that transcriptional programming acts together with a parallel biochemical network to enforce cell state.


Subject(s)
Cell Differentiation , Mitochondria/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Acetylation , Animals , Aspartic Acid/metabolism , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Citric Acid/metabolism , Citric Acid Cycle , Electron Transport , Female , Histones/metabolism , Humans , Lymphocyte Activation/genetics , Malates/metabolism , Male , Mice , Succinate Dehydrogenase/metabolism , Th1 Cells/metabolism , Transcription, Genetic
12.
Cell ; 172(4): 744-757.e17, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29398113

ABSTRACT

Cell communication within tissues is mediated by multiple paracrine signals including growth factors, which control cell survival and proliferation. Cells and the growth factors they produce and receive constitute a circuit with specific properties that ensure homeostasis. Here, we used computational and experimental approaches to characterize the features of cell circuits based on growth factor exchange between macrophages and fibroblasts, two cell types found in most mammalian tissues. We found that the macrophage-fibroblast cell circuit is stable and robust to perturbations. Analytical screening of all possible two-cell circuit topologies revealed the circuit features sufficient for stability, including environmental constraint and negative-feedback regulation. Moreover, we found that cell-cell contact is essential for the stability of the macrophage-fibroblast circuit. These findings illustrate principles of cell circuit design and provide a quantitative perspective on cell interactions.


Subject(s)
Cell Communication/physiology , Cell Proliferation/physiology , Fibroblasts/metabolism , Macrophages/metabolism , Animals , Cell Survival/physiology , Female , Fibroblasts/cytology , Macrophages/cytology , Male , Mice , Mice, Transgenic
13.
Cancer Cell ; 32(3): 280-281, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28898693

ABSTRACT

The tumor microenvironment presents metabolic constraints to immunosurveiling cells. In this issue of Cancer Cell, Zhang et al. demonstrate that CD8+ TILs reprogram under hypoxic and hypoglycemic conditions, regaining effector function by engaging fatty acid catabolism, which is promoted by fenofibrate and synergistic with immune checkpoint blockade therapy.


Subject(s)
Glucose , Lymphocytes, Tumor-Infiltrating/immunology , Fatty Acids , Humans , Oxygen , Tumor Microenvironment/immunology
14.
Cancer Immunol Res ; 5(1): 52-60, 2017 01.
Article in English | MEDLINE | ID: mdl-27923824

ABSTRACT

Janus kinase (JAK) inhibitors have achieved positive responses in myeloproliferative neoplasms, but at the expense of decreased natural killer (NK) cell numbers and compromised function. Selective JAK2 inhibition may also have a role in preventing and treating graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Although JAK inhibitors can impair monocyte-derived dendritic cell (moDC) activation and function and suppress effector T-cell responses, the effects on NK cells and the relevant mechanisms remain undefined. Using common γc cytokines and distinct human dendritic cell (DC) subtypes, we compared the effects of a JAK2-specific (TG101348) with a less selective JAK1/2 (ruxolitinib) inhibitor on NK-cell activation and function. Ruxolitinib treatment completely blocked IL2, IL15, and DC-mediated STAT5 phosphorylation, along with the capacity of NK cells to secrete IFNγ or lyse NK cell-sensitive targets. Only NK-cell proliferation stimulated by moDCs resisted ruxolitinib treatment. In contrast, TG101348 treatment of stimulated NK cells resulted in far less functional compromise. TG101348 completely inhibited only soluble IL15-mediated STAT5 phosphorylation, which Langerhans-type DCs (LCs), presenting membrane-bound IL15 in trans, could salvage. These results demonstrate that ruxolitinib's nonselective inhibition of JAK1/2 results in profound NK-cell dysfunction by blocking downstream pSTAT5, hence providing a persuasive rationale for the development of selective JAK2 inhibitors for immunotherapeutic applications. Cancer Immunol Res; 5(1); 52-60. ©2016 AACR.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Protein Kinase Inhibitors/pharmacology , Cell Line , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Humans , Immunophenotyping , Interferon-gamma/metabolism , Lymphocyte Count , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Phosphorylation , Receptors, Natural Killer Cell/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects
15.
Oncoimmunology ; 7(1): e1372081, 2017.
Article in English | MEDLINE | ID: mdl-29296525

ABSTRACT

Purpose: We conducted a phase I vaccine trial to determine safety, toxicity, and immunogenicity of autologous Langerhans-type dendritic cells (LCs), electroporated with murine tyrosinase-related peptide-2 (mTRP2) mRNA in patients with resected AJCC stage IIB, IIC, III, or IV (MIa) melanoma. Experimental Design: Nine patients received a priming immunization plus four boosters at three week intervals. Vaccines comprised 10 × 106 mRNA-electroporated LCs, based on absolute number of CD83+CD86brightHLA-DRbrightCD14neg LCs by flow cytometry. Initial vaccines used freshly generated LCs, whereas booster vaccines used viably thawed cells from the cryopreserved initial product. Post-vaccination assessments included evaluation of delayed-type hypersensitivity (DTH) reactions after booster vaccines and immune response assays at one and three months after the final vaccine. Results: All patients developed mild DTH reactions at injection sites after booster vaccines, but there were no toxicities exceeding grade 1 (CTCAE, v4.0). At one and three months post-vaccination, antigen-specific CD4 and CD8 T cells increased secretion of proinflammatory cytokines (IFN-γ, IL-2, and TNF-α), above pre-vaccine levels, and also upregulated the cytotoxicity marker CD107a. Next-generation deep sequencing of the TCR-V-ß CDR3 documented fold-increases in clonality of 2.11 (range 0.85-3.22) for CD4 and 2.94 (range 0.98-9.57) for CD8 T cells at one month post-vaccines. Subset analyses showed overall lower fold-increases in clonality in three patients who relapsed (CD4: 1.83, CD8: 1.54) versus non-relapsed patients (CD4: 2.31, CD8: 3.99). Conclusions: TRP2 mRNA-electroporated LC vaccines are safe and immunogenic. Responses are antigen-specific in terms of cytokine secretion, cytolytic degranulation, and increased TCR clonality, which correlates with clinical outcomes.

16.
Cancer Immunol Res ; 4(1): 61-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26464015

ABSTRACT

Multiple myeloma is the most common indication for high-dose chemotherapy and autologous stem cell transplantation (ASCT), and lenalidomide maintenance after transplant is now standard. Although lenalidomide doubles progression-free survival, almost all patients eventually relapse. Posttransplant immunotherapy to improve outcomes after ASCT therefore has great merit but first requires delineation of the dynamics of immune reconstitution. We evaluated lymphocyte composition and function after ASCT to guide optimal timing of immunotherapy and to identify potential markers of relapse. Regulatory T cells (Treg) decline as CD8(+) T cells expand during early lymphocyte recovery after ASCT, markedly reducing the Treg:CD8(+) effector T-cell ratio. These CD8(+) T cells can respond to autologous dendritic cells presenting tumor antigen in vitro as early as day +12 after transplant, becoming antigen-specific cytolytic T-lymphocyte effectors and thereby demonstrating preservation of cellular reactivity. CD4(+) and CD8(+) T cells express the negative regulatory molecules, CTLA-4, PD-1, LAG-3, and TIM-3, before and after ASCT. A subpopulation of exhausted/senescent CD8(+) T cells, however, downregulates CD28 and upregulates CD57 and PD-1, characterizing immune impairment and relapse after ASCT. Relapsing patients have higher numbers of these cells at +3 months after transplant, but before detection of clinical disease, indicating their applicability in identifying patients at higher risk of relapse. PD-1 blockade also revives the proliferation and cytokine secretion of the hyporesponsive, exhausted/senescent CD8(+) T cells in vitro. Collectively, these results identify T-cell exhaustion/senescence as a distinguishing feature of relapse and support early introduction of immunotherapy to stimulate antitumor immunity after ASCT.


Subject(s)
Multiple Myeloma/immunology , Neoplasm Recurrence, Local/immunology , Stem Cell Transplantation , Adult , Aged , Autografts , CD4-CD8 Ratio , Female , Humans , Immunotherapy , Male , Middle Aged , Multiple Myeloma/therapy , Neoplasm Recurrence, Local/prevention & control , T-Lymphocytes, Regulatory/immunology
17.
Immunity ; 43(4): 690-702, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26410627

ABSTRACT

The differentiation of CD4(+) helper T cell subsets with diverse effector functions is accompanied by changes in metabolism required to meet their bioenergetic demands. We find that follicular B helper T (Tfh) cells exhibited less proliferation, glycolysis, and mitochondrial respiration, accompanied by reduced mTOR kinase activity compared to T helper 1 (Th1) cells in response to acute viral infection. IL-2-mediated activation of the Akt kinase and mTORc1 signaling was both necessary and sufficient to shift differentiation away from Tfh cells, instead promoting that of Th1 cells. These findings were not the result of generalized signaling attenuation in Tfh cells, because they retained the ability to flux calcium and activate NFAT-transcription-factor-dependent cytokine production. These data identify the interleukin-2 (IL-2)-mTORc1 axis as a critical orchestrator of the reciprocal balance between Tfh and Th1 cell fates and their respective metabolic activities after acute viral infection.


Subject(s)
Interleukin-2/physiology , Multiprotein Complexes/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases/physiology , Animals , Apoptosis , Calcium Signaling , Cell Cycle , Cell Division , Enzyme Activation , Glucose/metabolism , Glycolysis , Interleukin-2 Receptor alpha Subunit/physiology , Lymphocytic choriomeningitis virus/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , NFATC Transcription Factors/physiology , Oxygen Consumption , Positive Regulatory Domain I-Binding Factor 1 , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
18.
J Transl Med ; 11: 166, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23837662

ABSTRACT

BACKGROUND: mRNA electroporation of dendritic cells (DCs) facilitates processing and presentation of multiple peptides derived from whole antigen, tailored to different HLA molecules. Clinical responses to electroporated moDC vaccines, however, have been suboptimal. Human Langerhans-type DCs (LCs) are the most potent conventional DC subtype for inducing CD8+ cytotoxic T lymphocytes (CTLs) in vitro. We recently demonstrated that Wilms' tumor 1 (WT1) mRNA-electroporated LCs are superior to moDCs as stimulators of tumor antigen-specific CD8+ CTLs, even though they are comparable stimulators of allogeneic T cell proliferative responses. A detailed comparative evaluation of the effects of mRNA electroporation on LCs versus moDCs, however, is needed. METHODS: Immature and partially-matured human moDCs and LCs electroporated with mRNA were compared for transfection efficiency, phenotypic changes, viability, retention of transgene expression after cryopreservation, and immunogenicity. Student t test was used for each pairwise comparison. One-way analysis of variance was used for multiple group comparisons. RESULTS: Transfection efficiency after electroporation with enhanced green fluorescent protein (eGFP) mRNA was higher for immature than for partially-matured moDCs. In contrast, transfection efficiency was higher for partially-matured than for immature LCs, with the additional benefit that electroporation itself increased maturation and activation of CD83+HLA-DRbright LCs but not moDCs. Electroporation did not impair final maturation and activation of either DC subtype, after which both mRNA-electroporated LCs and moDCs were functionally similar in stimulating allogeneic T cell proliferation, a standard assay of DC immunogenicity. CONCLUSIONS: These findings support mRNA electroporation of DCs, and in particular LCs, as an effective non-viral method to stimulate specific, potent CD8+ CTL responses. The differences between LCs and moDCs regarding this form of antigen-loading have important implications for DC-based immunotherapies.


Subject(s)
Dendritic Cells/cytology , Electroporation , Islets of Langerhans/cytology , Monocytes/cytology , Transfection , Antigen Presentation , Cell Survival , Cytokines/metabolism , Genetic Predisposition to Disease , Granulocyte Colony-Stimulating Factor/metabolism , HLA Antigens/metabolism , Humans , Immunotherapy , Phenotype , RNA, Messenger/metabolism , T-Lymphocytes, Cytotoxic/cytology , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...