Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1264882, 2023.
Article in English | MEDLINE | ID: mdl-38090585

ABSTRACT

The preclinical characterization of gene modified adoptive cellular immunotherapy candidates for clinical development often requires the use of mouse models. Gene-modified lymphocytes (GML) incorporating chimeric antigen receptors (CAR) and T-cell receptors (TCR) into immune effector cells require in vivo characterization of biological activity, mechanism of action, and preclinical safety. Typically, this characterization involves the assessment of dose-dependent, on-target, on-tumor activity in severely immunocompromised mice. While suitable for the purpose of evaluating T cell-expressed transgene function in a living host, this approach falls short in translating cellular therapy efficacy, safety, and persistence from preclinical models to humans. To comprehensively characterize cell therapy products in mice, we have developed a framework called "DIAL". This framework aims to enable an end-to-end understanding of genetically engineered cellular immunotherapies in vivo, from infusion to tumor clearance and long-term immunosurveillance. The acronym DIAL stands for Distribution, Infiltration, Accumulation, and Longevity, compartmentalizing the systemic attributes of gene-modified cellular therapy and providing a platform for optimization with the ultimate goal of improving therapeutic efficacy. This review will discuss both existent and emerging examples of DIAL characterization in mouse models, as well as opportunities for future development and optimization.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Animals , Mice , Immunotherapy, Adoptive , T-Lymphocytes , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Disease Models, Animal
2.
Cytotherapy ; 24(7): 720-732, 2022 07.
Article in English | MEDLINE | ID: mdl-35570170

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR) T cell therapy has yielded impressive clinical results in hematological malignancies and is a promising approach for solid tumor treatment. However, toxicity, including cytokine-release syndrome (CRS) and neurotoxicity, is a concern hampering its broader use. METHODS: In selecting a lead CAR-T candidate against the oncofetal antigen glypican 3 (GPC3), we compared CARs bearing a low- and high-affinity single-chain variable fragment (scFv) binding to a similar epitope and cross-reactive with murine GPC3. RESULTS: Where the high-affinity CAR-T cells were toxic in vivo, the low-affinity CAR maintained cytotoxic function against antigen-positive tumor cells but did not show toxicity against normal tissues. High-affinity CAR-induced toxicity was caused by on-target, off-tumor binding, based on the observation that higher doses of the high-affinity CAR-T caused toxicity in non-tumor-bearing mice and accumulated in organs with low expression of GPC3. To explore another layer of controlling CAR-T toxicity, we developed a means to target and eliminate CAR-T cells using anti-TNF-α antibody therapy after CAR-T infusion. The antibody was shown to function by eliminating early antigen-activated, but not all, CAR-T cells, allowing a margin where the toxic response could be effectively decoupled from antitumor efficacy with only a minor loss in tumor control. By exploring additional traits of the CAR-T cells after activation, we identified a mechanism whereby we could use approved therapeutics and apply them as an exogenous kill switch that eliminated early activated CAR-T following antigen engagement in vivo. CONCLUSIONS: By combining the reduced-affinity CAR with this exogenous control mechanism, we provide evidence that we can modulate and control CAR-mediated toxicity.


Subject(s)
Glypicans , Receptors, Chimeric Antigen , Animals , Cell Line, Tumor , Glypicans/metabolism , Immunotherapy, Adoptive/methods , Mice , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes , Tumor Necrosis Factor Inhibitors , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 115(9): 2174-2179, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29440413

ABSTRACT

ZAP-70 is a tyrosine kinase that is essential for initiation of T cell antigen receptor (TCR) signaling. We have found that T cell p38 MAP kinase (MAPK), which is directly phosphorylated and activated by ZAP-70 downstream of the TCR, in turn phosphorylates Thr-293 in the interdomain B region of ZAP-70. Mutant T cells expressing ZAP-70 with an alanine substitution at this residue (ZAP-70T293A) had enhanced TCR proximal signaling and increased effector responses. Lack of ZAP-70T293 phosphorylation increased association of ZAP-70 with the TCR and prolonged the existence of TCR signaling microclusters. These results identify a tight negative feedback loop in which ZAP-70-activated p38 reciprocally phosphorylates ZAP-70 and destabilizes the signaling complex.


Subject(s)
Genes, T-Cell Receptor/physiology , ZAP-70 Protein-Tyrosine Kinase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Acid Sequence , Gene Expression Regulation , Humans , Jurkat Cells , Phosphorylation , Signal Transduction , ZAP-70 Protein-Tyrosine Kinase/genetics , p38 Mitogen-Activated Protein Kinases/genetics
5.
PLoS One ; 12(11): e0188916, 2017.
Article in English | MEDLINE | ID: mdl-29190813

ABSTRACT

Flow cytometry is the primary immunological technique used to analyze multiple parameters on complex cell populations. We present a staining method that identifies major human mononuclear lymphoid and myeloid populations (CD4+ and CD8+ T cells, γδ T cells, B cells, NK cells and monocytes), using only two fluorochromes and a minimal number of cells. Our approach increases the number of markers recordable on most flow cytometers allowing for a deeper and more comprehensive immunophenotyping.


Subject(s)
Biomarkers/metabolism , Flow Cytometry/methods , Fluorescent Dyes/chemistry , Lymphocytes/classification , Cell Lineage , Humans , Immunophenotyping , Lymphocytes/immunology , Multiple Myeloma/immunology
6.
FEBS Lett ; 590(10): 1498-508, 2016 05.
Article in English | MEDLINE | ID: mdl-27086836

ABSTRACT

Pathogen-associated molecular pattern (PAMP) recognition leads to TANK-binding kinase (TBK1) polyubiquitination and activation by transautophosphorylation, resulting in IFN-ß production. Here, we describe a mouse model of optineurin insufficiency (OptnΔ(157) ) in which the TBK1-interacting N-terminus of optineurin was deleted. PAMP-stimulated cells from OptnΔ(157) mice had reduced TBK1 activity, no phosphorylation of optineurin Ser(187) , and mounted low IFN-ß responses. In contrast to pull-down assays where the presence of N-terminus was sufficient for TBK1 binding, both the N-terminal and the ubiquitin-binding regions of optineurin were needed for PAMP-induced binding. This report establishes optineurin as a positive regulator TBK1 via a bipartite interaction between these molecules.


Subject(s)
Eye Proteins/chemistry , Eye Proteins/genetics , Interferon-beta/metabolism , Pathogen-Associated Molecular Pattern Molecules/pharmacology , Protein Serine-Threonine Kinases/metabolism , Animals , Binding Sites , Cell Cycle Proteins , Eye Proteins/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Membrane Transport Proteins , Mice , Phosphorylation/drug effects , Protein Binding , Sequence Deletion
7.
Eur J Immunol ; 45(9): 2672-82, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26096449

ABSTRACT

Cellular inhibitor of apoptosis proteins (c-IAP) 1 and 2 are widely expressed ubiquitin protein ligases that regulate a variety of cellular functions, including the sensitivity of T cells to costimulation. 4-1BB is a TNF receptor family member that signals via a complex that includes TRAF family members and the c-IAPs to upregulate NF-κB and ERK, and has been implicated in memory T-cell survival. Here, we show that effector and memory T cells from mice expressing a dominant negative E3-inactive c-IAP2 (c-IAP2(H570A)) have impaired signaling downstream of 4-1BB. When infected with lymphocytic choriomeningitis virus, unlike mice in which c-IAPs were acutely downregulated by c-IAP antagonists, the primary response of c-IAP2(H570A) mice was normal. However, the number of antigen-specific CD8(+) but not CD4(+) T cells declined more rapidly and to a greater extent in c-IAP2(H570A) mice than in WT controls. Studies with T-cell adoptive transfer demonstrated that the enhanced decay of memory cells was T-cell intrinsic. Thus, c-IAP E3 activity is required for 4-1BB coreceptor signaling and maintenance of CD8(+) T-cell memory.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/genetics , Inhibitor of Apoptosis Proteins/metabolism , Lymphocytic Choriomeningitis/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Ubiquitin-Protein Ligases/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/genetics , Apoptosis/immunology , Baculoviral IAP Repeat-Containing 3 Protein , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/virology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Female , Gene Expression Regulation , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/immunology , Lymphocyte Activation/drug effects , Lymphocyte Count , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Transgenic , Mutation , NF-kappa B/genetics , NF-kappa B/immunology , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology , Ubiquitination
8.
J Immunol ; 191(12): 6231-40, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24244017

ABSTRACT

Optineurin is a widely expressed polyubiquitin-binding protein that has been implicated in regulating cell signaling via its NF-κB essential modulator-homologous C-terminal ubiquitin (Ub)-binding region. Its functions are controversial, with in vitro studies finding that optineurin suppressed TNF-mediated NF-κB activation and virus-induced activation of IFN regulatory factor 3 (IRF3), whereas bone marrow-derived macrophages (BMDMs) from mice carrying an optineurin Ub-binding point mutation had normal TLR-mediated NF-κB activation and diminished IRF3 activation. We have generated a mouse model in which the entire Ub-binding C-terminal region is deleted (Optn(470T)). Akin to C-terminal optineurin mutations found in patients with certain neurodegenerative diseases, Optn(470T) was expressed at substantially lower levels than the native protein, allowing assessment not only of the lack of Ub binding, but also of protein insufficiency. Embryonic lethality with incomplete penetrance was observed for 129 × C57BL/6 Optn(470T/470T) mice, but after further backcrossing to C57BL/6, offspring viability was restored. Moreover, the mice that survived were indistinguishable from wild type littermates and had normal immune cell distributions. Activation of NF-κB in Optn(470T) BMDM and BM-derived dendritic cells with TNF or via TLR4, T cells via the TCR, and B cells with LPS or anti-CD40 was normal. In contrast, optineurin and/or its Ub-binding function was necessary for optimal TANK binding kinase 1 and IRF3 activation, and both Optn(470T) BMDMs and bone marrow-derived dendritic cells had diminished IFN-ß production upon LPS stimulation. Importantly, Optn(470T) mice produced less IFN-ß upon LPS challenge. Therefore, endogenous optineurin is dispensable for NF-κB activation but necessary for optimal IRF3 activation in immune cells.


Subject(s)
Eye Proteins/physiology , Interferon Regulatory Factor-3/metabolism , Lymphocyte Subsets/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Animals , Cell Cycle Proteins , Chimera , Crosses, Genetic , Dendritic Cells/immunology , Eye Proteins/genetics , Gene Expression Regulation/immunology , Genes, Lethal , HEK293 Cells , Humans , Immunity, Innate , Interferon-beta/biosynthesis , Interferon-beta/genetics , Lipopolysaccharides/pharmacology , Lymphocyte Subsets/immunology , Macrophages/immunology , Male , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Receptors, Antigen, B-Cell/immunology , Sequence Deletion , Signal Transduction , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination
9.
J Immunol ; 191(5): 2282-9, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23913967

ABSTRACT

Engagement of the receptor CD27 by CD70 affects the magnitude and quality of T cell responses in a variety of infection models, and exaggerated signaling via this pathway results in enhanced immune responses and autoimmunity. One means by which signaling is regulated is tight control of cell surface CD70, which is expressed on dendritic cells (DCs), T cells, and B cells only upon activation. In this article, we show that a second level of regulation also is present. First, although undetectable on the cell surface by flow cytometry, immature DCs have a small pool of CD70 that continuously recycles from the plasma membrane. In addition, surface levels of CD70 on DCs and T cells were higher in mice deficient in CD27, or on DCs for which the interaction between CD70 and CD27 was precluded by blocking Abs. Binding of CD70 by its receptor resulted in downregulation of CD70 transcription and protein levels, suggesting that CD70-mediated "reverse signals" regulate its own levels. Therefore, the ability of CD70 to trigger costimulation is self-regulated when it binds its complementary receptor.


Subject(s)
CD27 Ligand/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , CD27 Ligand/metabolism , Dendritic Cells/metabolism , Down-Regulation , Flow Cytometry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
10.
PLoS One ; 8(6): e66161, 2013.
Article in English | MEDLINE | ID: mdl-23799077

ABSTRACT

Cellular Inhibitors of Apoptosis 1 and 2 (c-IAP1 and c-IAP2) are ubiquitin protein ligases (E3s) that constitutively ubiquitinate and induce proteasomal-mediated degradation of NF-κB Inducing Kinase (NIK) and repress non-canonical NF-κB activation. Mice expressing an E3-inactive c-IAP2 mutant (c-IAP2(H570A)) have constitutive activation of non-canonical NF-κB, resulting in B cell hyperplasia and T cell costimulation-independence. If, and if so to what extent, c-IAP1 and c-IAP2 are redundant in NF-κB regulation in these mice is not known. Here we have generated mice expressing a mutant c-IAP1 that lacks E3 activity (c-IAP1(H582A)). These mice were phenotypically normal and did not have constitutive NF-κB activation in B cells or MEFs. siRNA-mediated knockdown of c-IAP2 showed that accumulated c-IAP2, resulting from lack of c-IAP1-dependent degradation, compensated for absent c-IAP1 E3 activity. Surprisingly, c-IAP1(H582A) T cells had a lower p100/p52 ratio than wild type T cells, and in the absence of costimulation proliferated to a degree intermediate between wild type and c-IAP2(H570A) T cells. Therefore, although c-IAP1 and c-IAP2 both can repress constitutive NF-κB activation, the relative importance of each varies according to cell type.


Subject(s)
B-Lymphocytes/immunology , Inhibitor of Apoptosis Proteins/physiology , T-Lymphocytes/immunology , Animals , Flow Cytometry , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction
11.
J Immunol ; 190(2): 549-55, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23248260

ABSTRACT

c-IAP1 and c-IAP2 are ubiquitin protein ligases (E3s) that repress noncanonical NF-κB activation. We have created mice that bear a mutation in c-IAP2 that inactivates its E3 activity and interferes, in a dominant-negative fashion, with c-IAP1 E3 activity (c-IAP2(H570A)). The immune response of these animals was explored by infecting them with the Th1-inducing parasite Toxoplasma gondii. Surprisingly, c-IAP2(H570A) mice succumbed because of T cell production of high levels of proinflammatory cytokines. Unlike naive wild-type (WT) cells, which require signals generated by the TCR and costimulatory receptors to become fully activated, naive c-IAP2(H570A) T cells proliferated and produced high levels of IL-2 and IFN-γ to stimulation via TCR alone. c-IAP2(H570A) T cells had constitutive noncanonical NF-κB activation, and IκB kinase inhibition reduced their proliferation to anti-TCR alone to WT levels but had no effect when costimulation via CD28 was provided. Notably, T cells from nfkb2(-/-) mice, which cannot generate the p52 component of noncanonical NF-κB, were also costimulation independent, consistent with the negative role of this unprocessed protein in canonical NF-κB activation. Whereas T cells from nfkb2(+/-) mice behaved like WT, coexpression of a single copy of c-IAP2(H570A) resulted in cleavage of p100, upregulation of p52, and T cell costimulation independence. Thus, p100 represses and p52 promotes costimulation, and the ratio regulates T cell dependence on costimulatory signals.


Subject(s)
NF-kappa B p52 Subunit/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Enzyme Activation , I-kappa B Kinase/antagonists & inhibitors , Immunologic Memory , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Mutation , NF-kappa B p52 Subunit/chemistry , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/immunology , Toxoplasmosis/metabolism
12.
Cell Host Microbe ; 12(4): 509-20, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-22999859

ABSTRACT

Commensal bacteria impact host health and immunity through various mechanisms, including the production of immunomodulatory molecules. Bacteroides fragilis produces a capsular polysaccharide (PSA), which induces regulatory T cells and mucosal tolerance. However, unlike pathogens, which employ secretion systems, the mechanisms by which commensal bacteria deliver molecules to the host remain unknown. We reveal that Bacteroides fragilis releases PSA in outer membrane vesicles (OMVs) that induce immunomodulatory effects and prevent experimental colitis. Dendritic cells (DCs) sense OMV-associated PSA through TLR2, resulting in enhanced regulatory T cells and anti-inflammatory cytokine production. OMV-induced signaling in DCs requires growth arrest and DNA-damage-inducible protein (Gadd45α). DCs treated with PSA-containing OMVs prevent experimental colitis, whereas Gadd45α(-/-) DCs are unable to promote regulatory T cell responses or suppress proinflammatory cytokine production and host pathology. These findings demonstrate that OMV-mediated delivery of a commensal molecule prevents disease, uncovering a mechanism of interkingdom communication between the microbiota and mammals.


Subject(s)
Bacteroides fragilis/immunology , Bacteroides fragilis/metabolism , Exosomes/immunology , Exosomes/metabolism , Immunologic Factors/metabolism , Polysaccharides, Bacterial/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gene Expression Profiling , Immunologic Factors/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Polysaccharides, Bacterial/immunology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 2/drug effects , Toll-Like Receptor 2/immunology
13.
Blood ; 118(12): 3280-9, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21715315

ABSTRACT

Stimulation via the T-cell receptor (TCR) activates p38α and p38ß by phosphorylation of p38 Tyr-323 (p38(Y323)). Here we characterize knockin mice in which p38α and/or ß Tyr-323 has been replaced with Phe. We find that p38α accounts for two-thirds and p38ß the remainder of TCR-induced p38 activation. T cells from double knockin mice (p38αß(Y323F)) had defects in TCR-mediated proliferation and Th1 and Th17 skewing, the former corresponding with an inability to sustain T-bet expression. Introduction of p38α(Y323F) into Gadd45α-deficient mice, in which the alternative p38 pathway is constitutively active, reversed T-cell hyperproliferation and autoimmunity. Furthermore, p38αß(Y323F) mice had delayed onset and reduced severity of the inflammatory autoimmune diseases collagen-induced arthritis and experimental autoimmune encephalomyelitis. Thus, T cell-specific alternative activation of p38 is an important pathway in T-cell proliferation, Th skewing, and inflammatory autoimmunity, and may be an attractive tissue-specific target for intervention in these processes.


Subject(s)
Arthritis, Experimental/metabolism , Autoimmunity , Cell Cycle Proteins/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Inflammation/metabolism , Nuclear Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Signal Transduction/immunology , T-Lymphocytes/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Blotting, Western , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/immunology , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Female , Gene Knock-In Techniques , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lymphocyte Activation , Mice , Mice, Transgenic , Nuclear Proteins/deficiency , Nuclear Proteins/immunology , Phenylalanine/immunology , Phenylalanine/metabolism , Phosphorylation , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Tyrosine/immunology , Tyrosine/metabolism , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
14.
PLoS One ; 5(6): e11052, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20548796

ABSTRACT

BACKGROUND: HIV-1 Protease Inhibitors, namely PIs, originally designed to inhibit HIV-1 aspartic protease, can modulate the immune response by mechanisms largely unknown, and independent from their activity on viral replication. Here, we analyzed the ability of PIs to interfere with differentiation program of monocytes toward dendritic cell (DCs) lineage, a key process in the inflammatory response. METHODOLOGY/PRINCIPAL FINDINGS: Monocytes from healthy donors were isolated and induced to differentiate in vitro in the presence or absence of saquinavir, ritonavir, nelfinavir, indinavir or amprenavir (sqv, rtv, nlfv, idv, apv, respectively). These drugs demonstrated a differential ability to sustain the generation of immature DCs (iDCs) with an altered phenotype, including low levels of CD1a, CD86, CD36 and CD209. DCs generated in the presence of rtv also failed to acquire the typical phenotype of mature DCs (mDCs), and secreted lower amounts of IL-12 and IL-15. Accordingly, these aberrant mDCs failed to support activation of autologous Natural Killer (NK) cells, and resulted highly susceptible to NK cell-mediated cytotoxicity. CONCLUSIONS/SIGNIFICANCE: Our findings uncover novel functional properties of PIs within the DC-NK cell cross-talk, unveiling the heterogeneous ability of members of this class drugs to drive the generation of atypical monocyte-derived DCs (MDDCs) showing an aberrant phenotype, a failure to respond appropriately to bacterial endotoxin, a weak ability to prime autologous NK cells, and a high susceptibility to NK cell killing. These unexpected properties might contribute to limit inflammation and viral spreading in HIV-1 infected patients under PIs treatment, and open novel therapeutical perspectives for this class drugs as immunomodulators in autoimmunity and cancer.


Subject(s)
Dendritic Cells/drug effects , HIV Protease Inhibitors/pharmacology , HIV Protease/drug effects , Killer Cells, Natural/drug effects , Cell Proliferation , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunophenotyping , Killer Cells, Natural/immunology
15.
Blood ; 114(10): 2121-30, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19617575

ABSTRACT

The ability of the adaptive immune system to respond rapidly and robustly upon repeated antigen exposure is known as immunologic memory, and it is thought that acquisition of memory T-cell function is an irreversible differentiation event. In this study, we report that many phenotypic and functional characteristics of antigen-specific CD8 memory T cells are lost when they are deprived of contact with dendritic cells. Under these circumstances, memory T cells reverted from G(1) to the G(0) cell-cycle state and responded to stimulation like naive T cells, as assessed by proliferation, dependence upon costimulation, and interferon-gamma production, without losing cell surface markers associated with memory. The memory state was maintained by signaling via members of the tumor necrosis factor receptor superfamily, CD27 and 4-1BB. Foxo1, a transcription factor involved in T-cell quiescence, was reduced in memory cells, and stimulation of naive CD8 cells via CD27 caused Foxo1 to be phosphorylated and emigrate from the nucleus in a phosphatidylinositol-3 kinase-dependent manner. Consistent with these results, maintenance of G(1) in vivo was compromised in antigen-specific memory T cells in vesicular stomatitis virus-infected CD27-deficient mice. Therefore, sustaining the functional phenotype of T memory cells requires active signaling and maintenance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Dendritic Cells/immunology , Immunologic Memory/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Animals , Antigens, Viral/immunology , Cell Communication/genetics , Cell Nucleus/immunology , Forkhead Box Protein O1 , Forkhead Transcription Factors/immunology , G1 Phase/immunology , Immunologic Memory/genetics , Interferon-gamma/immunology , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/immunology , Phosphorylation/genetics , Phosphorylation/immunology , Resting Phase, Cell Cycle/immunology , Signal Transduction/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Vesicular Stomatitis/immunology , Vesiculovirus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...