Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
2.
Nat Med ; 26(2): 270-280, 2020 02.
Article in English | MEDLINE | ID: mdl-31959992

ABSTRACT

Anti-CD19 chimeric antigen receptor (CAR)-expressing T cells are an effective treatment for B-cell lymphoma, but often cause neurologic toxicity. We treated 20 patients with B-cell lymphoma on a phase I, first-in-human clinical trial of T cells expressing the new anti-CD19 CAR Hu19-CD828Z (NCT02659943). The primary objective was to assess safety and feasibility of Hu19-CD828Z T-cell therapy. Secondary objectives included assessments of blood levels of CAR T cells, anti-lymphoma activity, second infusions and immunogenicity. All objectives were met. Fifty-five percent of patients who received Hu19-CD828Z T cells obtained complete remission. Hu19-CD828Z T cells had clinical anti-lymphoma activity similar to that of T cells expressing FMC63-28Z, an anti-CD19 CAR tested previously by our group, which contains murine binding domains and is used in axicabtagene ciloleucel. However, severe neurologic toxicity occurred in only 5% of patients who received Hu19-CD828Z T cells, whereas 50% of patients who received FMC63-28Z T cells experienced this degree of toxicity (P = 0.0017). T cells expressing Hu19-CD828Z released lower levels of cytokines than T cells expressing FMC63-28Z. Lower levels of cytokines were detected in blood from patients who received Hu19-CD828Z T cells than in blood from those who received FMC63-28Z T cells, which could explain the lower level of neurologic toxicity associated with Hu19-CD828Z. Levels of cytokines released by CAR-expressing T cells particularly depended on the hinge and transmembrane domains included in the CAR design.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/immunology , Adolescent , Adult , Aged , Cytokines/metabolism , Feasibility Studies , Female , Humans , K562 Cells , Male , Middle Aged , Phenotype , Protein Domains , Remission Induction , Young Adult
3.
Blood ; 132(8): 804-814, 2018 08 23.
Article in English | MEDLINE | ID: mdl-29895668

ABSTRACT

After treatment with chimeric antigen receptor (CAR) T cells, interleukin-15 (IL-15) elevation and CAR T-cell expansion are associated with non-Hodgkin lymphoma (NHL) outcomes. However, the association of preinfusion CAR product T-cell functionality with clinical outcomes has not been reported. A single-cell analysis of the preinfusion CD19 CAR product from patients with NHL demonstrated that CAR products contain polyfunctional T-cell subsets capable of deploying multiple immune programs represented by cytokines and chemokines, including interferon-γ, IL-17A, IL-8, and macrophage inflammatory protein 1α. A prespecified T-cell polyfunctionality strength index (PSI) applied to preinfusion CAR product was significantly associated with clinical response, and PSI combined with CAR T-cell expansion or pretreatment serum IL-15 levels conferred additional significance. Within the total product cell population, associations with clinical outcomes were greater with polyfunctional CD4+ T cells compared with CD8+ cells. Grade ≥3 cytokine release syndrome was associated with polyfunctional T cells, and both grade ≥3 neurologic toxicity and antitumor efficacy were associated with polyfunctional IL-17A-producing T cells. The findings in this exploratory study show that a preinfusion CAR product T-cell subset with a definable polyfunctional profile has a major association with clinical outcomes of CAR T-cell therapy. This trial was registered at www.clinicaltrials.gov as #NCT00924326.


Subject(s)
Adoptive Transfer , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Lymphoma, Non-Hodgkin , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen/therapeutic use , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cytokines/immunology , Female , Humans , K562 Cells , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/pathology , Lymphoma, Non-Hodgkin/therapy , Male , Middle Aged
4.
PLoS One ; 8(2): e56209, 2013.
Article in English | MEDLINE | ID: mdl-23409157

ABSTRACT

Foxp3(+) regulatory T cells (Treg) play a crucial role in regulating immune tolerance. The use of Treg to restore immune tolerance is considered an attractive novel approach to inhibit autoimmune disease, including type 1 diabetes (T1D), and to prevent rejection of organ transplants. In view of the goal of developing autologous Treg-based cell therapy for patients with long-term (>15 years) T1D, it will be necessary to expand a sufficient amount of functional Treg in vitro in order to study and compare Treg from T1D patients and healthy subjects. Our results have demonstrated that there is a comparable frequency of Treg in the peripheral blood lymphocytes (PBLs) of patients with long-term T1D relative to those in healthy subjects; however, Th1 cells, but not Th17 cells, were increased in the T1D patients. Further, more Treg in PBLs from T1D patients than from healthy subjects expressed the CD45RO(+) memory cell phenotype, suggesting they were antigen-experienced cells. After isolation, Treg from both T1D patients and healthy subjects were successfully expanded with high purity. Although there was no difference in Helios expression on Treg in PBLs, in vitro expansion led to fewer Helios-expressing Treg from T1D patients than healthy subjects. While more Th1-like Treg expressing IFN-γ or TNF-α were found in the PBLs of T1D patients than healthy controls, there was no such difference in the expanded Treg. Importantly, expanded Treg from both subject groups were able to suppress autologous or allogeneic CD8(+) effector T cells equally well. Our findings demonstrate that a large number of ex vivo expanded functional Treg can be obtained from long-term T1D patients, although fewer expanded Treg expressed a high level of Helios. Thus, based on the positive outcomes, these potent expanded Treg from diabetic human patients may be useful in treating T1D or preventing islet graft rejection.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Ikaros Transcription Factor/metabolism , Recovery of Function , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Adult , Case-Control Studies , Cell Count , Cell Proliferation , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Female , Humans , Immune Tolerance , Interferon-gamma/metabolism , Leukocyte Common Antigens/metabolism , Male , Middle Aged , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th17 Cells/cytology , Tumor Necrosis Factor-alpha/metabolism , Young Adult
5.
Proc Natl Acad Sci U S A ; 109(24): 9493-8, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22645357

ABSTRACT

Autoreactive pathogenic T cells (Tpaths) and regulatory T cells (Tregs) express a distinct gene profiles; however, the genes and associated genetic/signaling pathways responsible for the functional determination of Tpaths vs. Tregs remain unknown. Here we show that Skp2, an E3 ubiquitin ligase that affects cell cycle control and death, plays a critical role in the function of diabetogenic Tpaths and Tregs. Down-regulation of Skp2 in diabetogenic Tpaths converts them into Foxp3-expressing Tregs. The suppressive function of the Tpath-converted Tregs is dependent on increased production of TGF-ß/IL-10, and these Tregs are able to inhibit spontaneous diabetes in NOD mice. Like naturally arising Foxp3(+) nTregs, the converted Tregs are anergic cells with decreased proliferation and activation-induced cell death. Skp2 down-regulation leads to Tpath-Treg conversion due at least in part to up-regulation of several genes involved in cell cycle control and genes in the Foxo family. Down-regulation of the cyclin-dependent kinase inhibitor p27 alone significantly attenuates the effect of Skp2 on Tpaths and reduces the suppressive function of converted Tregs; its effect is further improved with concomitant down-regulation of p21, Foxo1, and Foxo3. In comparison, Skp2 overexpression does not change Tpath function, but significantly decreases Foxp3 expression and abrogates the suppressive function of nTregs. These findings support the critical role of Skp2 in functional specification of Tpaths and Tregs, and demonstrate an important molecular mechanism mediating Skp2 function in balancing immune tolerance during autoimmune disease development.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Forkhead Transcription Factors/metabolism , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Animals , Mice , Mice, Inbred NOD , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism
6.
Epigenetics ; 6(10): 1207-16, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21931278

ABSTRACT

The epigenetic mechanism of folic acid (FA) action on dorsal root ganglion (DRG) cell proliferation and sensory neuron differentiation is not well understood. In this study, the ND7 cell line, derived from DRG cells, was used to elucidate this mechanism. In ND7 cells differentiated with dbcAMP and NGF, Hes1 and Pax3 levels decreased, whereas Neurog2 levels showed a modest increase. Chromatin immunoprecipitation (ChIP) assays examining epigenetic marks at the Hes1 promoter showed that FA favored increased H3K9 and H3K19 acetylation and decreased H3K27 methylation. Hence, FA plays a positive role in cell proliferation. In differentiated ND7 cells, H3K27 methylation decreased, whereas H3K9 and H3K18 acetylation increased at the Neurog2 promoter. FA did not favor this phenotypic outcome. Additionally, in differentiated ND7 Neurog2 associated with the NeuroD1 promoter, FA decreased this association. The results suggest that the switch from proliferation to sensory neuron differentiation in DRG cells is regulated by alterations in epigenetic marks, H3K9/18 acetylation and H3K27 methylation, at Hes1 and Neurog2 promoters, as well as by Neurog2 association with NeuroD1 promoter. FA although positive for proliferation, does not appear to play a role in differentiation.


Subject(s)
Epigenesis, Genetic/drug effects , Folic Acid/pharmacology , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/drug effects , Neurogenesis/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Chromatin/chemistry , Chromatin/metabolism , Chromatin/physiology , Histones/chemistry , Histones/metabolism , Histones/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Rats
7.
PLoS Pathog ; 7(6): e1002071, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21655109

ABSTRACT

HSV-1 is the leading cause of sporadic encephalitis in humans. HSV infection of susceptible 129S6 mice results in fatal encephalitis (HSE) caused by massive inflammatory brainstem lesions comprising monocytes and neutrophils. During infection with pathogenic microorganisms or autoimmune disease, IgGs induce proinflammatory responses and recruit innate effector cells. In contrast, high dose intravenous immunoglobulins (IVIG) are an effective treatment for various autoimmune and inflammatory diseases because of potent anti-inflammatory effects stemming in part from sialylated IgGs (sIgG) present at 1-3% in IVIG. We investigated the ability of IVIG to prevent fatal HSE when given 24 h post infection. We discovered a novel anti-inflammatory pathway mediated by low-dose IVIG that protected 129S6 mice from fatal HSE by modulating CNS inflammation independently of HSV specific antibodies or sIgG. IVIG suppressed CNS infiltration by pathogenic CD11b(+) Ly6C(high) monocytes and inhibited their spontaneous degranulation in vitro. FcγRIIb expression was required for IVIG mediated suppression of CNS infiltration by CD45(+) Ly6C(low) monocytes but not for inhibiting development of Ly6C(high) monocytes. IVIG increased accumulation of T cells in the CNS, and the non-sIgG fraction induced a dramatic expansion of FoxP3(+) CD4(+) T regulatory cells (Tregs) and FoxP3(-) ICOS(+) CD4(+) T cells in peripheral lymphoid organs. Tregs purified from HSV infected IVIG treated, but not control, mice protected adoptively transferred mice from fatal HSE. IL-10, produced by the ICOS(+) CD4(+) T cells that accumulated in the CNS of IVIG treated, but not control mice, was essential for induction of protective anti-inflammatory responses. Our results significantly enhance understanding of IVIG's anti-inflammatory and immunomodulatory capabilities by revealing a novel sIgG independent anti-inflammatory pathway responsible for induction of regulatory T cells that secrete the immunosuppressive cytokine IL-10 and further reveal the therapeutic potential of IVIG for treating viral induced inflammatory diseases.


Subject(s)
Encephalitis, Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Interleukin-10/metabolism , Animals , Antigens, Ly/metabolism , Blood-Brain Barrier/immunology , Brain Stem/pathology , CD4-Positive T-Lymphocytes/immunology , Encephalitis, Herpes Simplex/mortality , Encephalitis, Herpes Simplex/prevention & control , Encephalitis, Herpes Simplex/virology , Flow Cytometry , Herpesvirus 1, Human/pathogenicity , Humans , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/immunology , Interleukin-10/administration & dosage , Interleukin-10/immunology , Leukocytes/immunology , Macrophages/immunology , Mice , Mice, Transgenic , Monocytes/immunology , Neutrophils/immunology , T-Lymphocytes, Regulatory/immunology , Time Factors
8.
Mol Biol Cell ; 22(4): 503-12, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21169561

ABSTRACT

Pax3 plays a role in regulating Hes1 and Neurog2 activity and thereby stem cell maintenance and neurogenesis. A mechanism for Pax3 regulation of these two opposing events, during caudal neural tube development, is examined in this study. Pax3 acetylation on C-terminal lysine residues K437 and K475 may be critical for proper regulation of Hes1 and Neurog2. Removal of these lysine residues increased Hes1 but decreased Neurog2 promoter activity. SIRT1 deacetylase may be a key component in regulating Pax3 acetylation. Chromatin immunoprecipitation assays showed that SIRT1 is associated with Hes1 and Neurog2 promoters during murine embryonic caudal neural tube development at E9.5, but not at E12.5. Overexpression of SIRT1 decreased Pax3 acetylation, Neurog2 and Brn3a positive staining. Conversely, siRNA-mediated silencing of SIRT1 increased these factors. These studies suggest that Pax3 acetylation results in decreased Hes1 and increased Neurog2 activity, thereby promoting sensory neuron differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Paired Box Transcription Factors/metabolism , Sensory Receptor Cells/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation/methods , Homeodomain Proteins/genetics , Humans , Mice , Nerve Tissue Proteins/genetics , Neural Tube/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Transcription Factor HES-1
9.
J Biol Chem ; 285(47): 36922-32, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20833714

ABSTRACT

The mechanism(s) behind folate rescue of neural tube closure are not well understood. In this study we show that maternal intake of folate prior to conception reverses the proliferation potential of neural crest stem cells in homozygous Splotch embryos (Sp(-/-)) via epigenetic mechanisms. It is also shown that the pattern of differentiation seen in these cells is similar to wild-type (WT). Cells from open caudal neural tubes of Sp(-/-) embryos exhibit increased H3K27 methylation and decreased expression of KDM6B possibly due to up-regulation of KDM6B targeting micro-RNAs such as miR-138, miR-148a, miR-185, and miR-339-5p. In our model, folate reversed these epigenetic marks in folate-rescued Sp(-/-) embryos. Using tissue from caudal neural tubes of murine embryos we also examined H3K27me2 and KDM6B association with Hes1 and Neurog2 promoters at embryonic day E10.5, the proliferative stage, and E12.5, when neural differentiation begins. In Sp(-/-) embryos compared with WT, levels of H3K27me2 associated with the Hes1 promoter were increased at E10.5, and levels associated with the Neurog2 promoter were increased at E12.5. KDM6B association with Hes1 and Neurog2 promoters was inversely related to H3K27me2 levels. These epigenetic changes were reversed in folate-rescued Sp(-/-) embryos. Thus, one of the mechanisms by which folate may rescue the Sp(-/-) phenotype is by increasing the expression of KDM6B, which in turn decreases H3K27 methylation marks on Hes1 and Neurog2 promoters thereby affecting gene transcription.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Chromatin Assembly and Disassembly/drug effects , Folic Acid/administration & dosage , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Neural Tube/drug effects , Neural Tube/embryology , Promoter Regions, Genetic/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Central Nervous System/embryology , Chromatin Assembly and Disassembly/genetics , Chromatin Immunoprecipitation , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Epigenomics , Female , Fluorescent Antibody Technique , Folic Acid/pharmacology , Histones/genetics , Histones/metabolism , Homeodomain Proteins/metabolism , Immunoenzyme Techniques , Immunoprecipitation , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Luciferases/metabolism , Male , Methylation , Mice , Mice, Inbred C57BL , MicroRNAs/physiology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Tube Defects/prevention & control , PAX3 Transcription Factor , Paired Box Transcription Factors/physiology , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor HES-1 , Vitamin B Complex/administration & dosage , Vitamin B Complex/pharmacology
10.
Int J Dev Biol ; 53(1): 69-79, 2009.
Article in English | MEDLINE | ID: mdl-19123128

ABSTRACT

Pax3 regulates neural crest cell migration and is critical during neural crest development. TGFbs modify neural crest cell migration and differentiation. TGFbeta2 nullizygous embryos (TGFbeta2(-/-)Pax3(+/+)) display open neural tube and bifid spine, whereas in wild type embryos, the neural tube is closed. In previous work, we have demonstrated that Pax3 regulates TGFbeta2 by directly binding to cis-regulatory elements on its promoter. In this study, we found that the TGFbeta2 nullizygous phenotype can be reversed to the wild type phenotype by down-regulating one allele of Pax3, as in TGFbeta2(-/-)Pax3(+/-) embryos obtained through breeding TGFb2(+/-)Pax3(+/-) mice. The data in this paper suggest that Pax3 and TGFbeta2 interact in a coordinated gene regulatory network, linked by common downstream effector genes, to bring about this phenotypic reversal. Downstream effectors may include Hes1, Ngn2 and Sox9, as well as other genes involved in neuronal differentiation.


Subject(s)
Gene Regulatory Networks/genetics , Neural Crest/embryology , Neural Crest/metabolism , Paired Box Transcription Factors/metabolism , Signal Transduction , Transcription, Genetic/genetics , Transforming Growth Factor beta2/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/genetics , Heterozygote , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/deficiency , Paired Box Transcription Factors/genetics , Phenotype , Promoter Regions, Genetic/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factor HES-1 , Transforming Growth Factor beta2/deficiency , Transforming Growth Factor beta2/genetics
11.
Dev Biol ; 316(2): 510-23, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18308300

ABSTRACT

Pax3 is expressed early during embryonic development in spatially restricted domains including limb muscle, neural crest, and neural tube. Pax3 functions at the nodal point in melanocyte stem cell differentiation, cardiogenesis and neurogenesis. Additionally Pax3 has been implicated in migration and differentiation of precursor cell populations. Currently there are questions about how Pax3 regulates these diverse functions. In this study we found that in the absence of functional Pax3, as in Splotch embryos, the neural crest cells undergo premature neurogenesis, as evidenced by increased Brn3a positive staining in neural tube explants, in comparison with wild-type. Premature neurogenesis in the absence of functional Pax3 may be due to a change in the regulation of basic helix-loop-helix transcription factors implicated in proliferation and differentiation. Using promoter-luciferase activity measurements in transient co-transfection experiments and electro-mobility shift assays, we show that Pax3 regulates Hairy and enhancer of split homolog-1 (Hes1) and Neurogenin2 (Ngn2) by directly binding to their promoters. Chromatin immunoprecipitation assays confirmed that Pax3 bound to cis-regulatory elements within Hes1 and Ngn2 promoters. These observations suggest that Pax3 regulates Hes1 and Ngn2 and imply that it may couple migration with neural stem cell maintenance and neurogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Animals , Cell Movement , Genotype , Helix-Loop-Helix Motifs/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Neural Crest/physiology , Neural Tube/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor HES-1
SELECTION OF CITATIONS
SEARCH DETAIL
...