Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
EMBO Rep ; 24(1): e54944, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36341538

ABSTRACT

Melanoma tumors are highly metastatic partly due to the ability of melanoma cells to transition between invasive and proliferative states. However, the mechanisms underlying this plasticity are still not fully understood. To identify new epigenetic regulators of melanoma plasticity, we combined data mining, tumor models, proximity proteomics, and CUT&RUN sequencing. We focus on the druggable family of bromodomain epigenetic readers and identify TRIM28 as a new regulator of melanoma plasticity. We find that TRIM28 promotes the expression of pro-invasive genes and that TRIM28 controls the balance between invasiveness and growth of melanoma cells. We demonstrate that TRIM28 acts via the transcription factor JUNB that directly regulates the expression of pro-invasive and pro-growth genes. Mechanistically, TRIM28 controls the expression of JUNB by negatively regulating its transcriptional elongation by RNA polymerase II. In conclusion, our results demonstrate that a TRIM28-JUNB axis controls the balance between invasiveness and growth in melanoma tumors and suggest that the bromodomain protein TRIM28 could be targeted to reduce tumor spread.


Subject(s)
Gene Expression Regulation , Melanoma , Humans , Cell Line, Tumor , Tripartite Motif-Containing Protein 28/genetics , Melanoma/genetics
3.
Nat Med ; 28(2): 345-352, 2022 02.
Article in English | MEDLINE | ID: mdl-35027758

ABSTRACT

Chimeric antigen receptors (CARs) are receptors for antigen that direct potent immune responses. Tumor escape associated with low target antigen expression is emerging as one potential limitation of their efficacy. Here we edit the TRAC locus in human peripheral blood T cells to engage cell-surface targets through their T cell receptor-CD3 complex reconfigured to utilize the same immunoglobulin heavy and light chains as a matched CAR. We demonstrate that these HLA-independent T cell receptors (HIT receptors) consistently afford high antigen sensitivity and mediate tumor recognition beyond what CD28-based CARs, the most sensitive design to date, can provide. We demonstrate that the functional persistence of HIT T cells can be augmented by constitutive coexpression of CD80 and 4-1BBL. Finally, we validate the increased antigen sensitivity afforded by HIT receptors in xenograft mouse models of B cell leukemia and acute myeloid leukemia, targeting CD19 and CD70, respectively. Overall, HIT receptors are well suited for targeting cell surface antigens of low abundance.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Animals , Antigens, CD19 , Histocompatibility Antigens , Humans , Immunotherapy, Adoptive , Mice , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen/metabolism , Xenograft Model Antitumor Assays
4.
Sci Transl Med ; 13(623): eabh1962, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34878825

ABSTRACT

Despite the high remission rates achieved using T cells bearing a chimeric antigen receptor (CAR) against hematogical malignancies, there is still a considerable proportion of patients who eventually experience tumor relapse. Clinical studies have established that mechanisms of treatment failure include the down-regulation of target antigen expression and the limited persistence of effective CAR T cells. We hypothesized that dual targeting mediated by a CAR and a chimeric costimulatory receptor (CCR) could simultaneously enhance T cell cytotoxicity and improve durability. Concomitant high-affinity engagement of a CD38-binding CCR enhanced the cytotoxicity of BCMA-CAR and CD19-CAR T cells by increasing their functional binding avidity. In comparison to second-generation BCMA-CAR or CD19-CAR T cells, double-targeted CAR + CD38-CCR T cells exhibited increased sensitivity to recognize and lyse tumor variants of multiple myeloma and acute lymphoblastic leukemia with low antigen density in vitro. In addition, complimentary costimulation by 4-1BB and CD28 endodomains provided by the CAR and CCR combination conferred increased cytokine secretion and expansion and improved persistence in vivo. The cumulatively improved properties of CAR + CCR T cells enabled the in vivo eradication of antigen-low tumor clones, which were otherwise resistant to treatment with conventional CAR T cells. Therefore, multiplexing targeting and costimulation through the combination of a CAR and a CCR is a powerful strategy to improve the clinical outcomes of CAR T cells by enhancing cytotoxic efficacy and persistence, thus preventing relapses of tumor clones with low target antigen density.


Subject(s)
Multiple Myeloma , Receptors, Chimeric Antigen , Antigens, CD19 , Humans , Immunotherapy, Adoptive , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes
5.
Immunology ; 159(3): 335-343, 2020 03.
Article in English | MEDLINE | ID: mdl-31755557

ABSTRACT

TRIM21 is an interferon-stimulated E3 ligase that controls the activity of pattern-recognition signaling via ubiquitination of interferon regulatory factors and DDX41. Previous studies on the role of TRIM21 in innate immune responses have yielded contradictory results, suggesting that the role of TRIM21 is cell specific. Here, we report that bone-marrow-derived macrophages (BMDMs) generated from Trim21-/- mice have reduced expression of mature macrophage markers. Reflecting their reduced differentiation in response to macrophage colony-stimulating factor (M-CSF), Trim21-/- BMDMs had decreased expression of M-CSF signature genes. Although Trim21-/- BMDMs responded normally to Toll-like receptor 9 (TLR9) activation, they produced lower levels of pro-inflammatory cytokines in response to the TLR2 agonist PAM3CSK4. In line with this, the response to infection with the Bacillus Calmette-Guérin strain of Mycobacterium bovis was also diminished in Trim21-/- BMDMs. Our results indicate that TRIM21 controls responses to TLR2 agonists.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Ribonucleoproteins/metabolism , Toll-Like Receptor 2/metabolism , Animals , Cell Differentiation , Cells, Cultured , Host-Pathogen Interactions , Lipopeptides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium bovis/immunology , Mycobacterium bovis/pathogenicity , Phenotype , Ribonucleoproteins/deficiency , Ribonucleoproteins/genetics , Signal Transduction , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics
6.
Eur J Immunol ; 49(2): 313-322, 2019 02.
Article in English | MEDLINE | ID: mdl-30307034

ABSTRACT

Systemic autoimmune diseases are characterized by the overexpression of type I IFN stimulated genes, and accumulating evidence indicate a role for type I IFNs in these diseases. However, the underlying mechanisms for this are still poorly understood. To explore the role of type I IFN regulated miRNAs in systemic autoimmune disease, we characterized cellular expression of miRNAs during both acute and chronic type I IFN responses. We identified a T cell-specific reduction of miR-31-5p levels, both after intramuscular injection of IFNß and in patients with Sjögren's syndrome (SjS). To interrogate the role of miR-31-51p in T cells we transfected human CD4+ T cells with a miR-31-5p inhibitor and performed metabolic measurements. This identified an increase in basal levels of glucose metabolism after inhibition of miR-31-5p. Furthermore, treatment with IFN-α also increased the basal levels of human CD4+ T-cell metabolism. In all, our results suggest that reduced levels of miR-31-5p in T cells of SjS patients support autoimmune T-cell responses during chronic type I IFN exposure.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Energy Metabolism/immunology , MicroRNAs/immunology , Sjogren's Syndrome/immunology , CD4-Positive T-Lymphocytes/pathology , Energy Metabolism/drug effects , Female , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Interferon-beta/immunology , Interferon-beta/pharmacology , Male , Sjogren's Syndrome/pathology
7.
J Immunol ; 196(1): 91-6, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26590315

ABSTRACT

Patients with systemic lupus erythematosus (SLE) and primary Sjögren's syndrome (pSS) are typically characterized by the presence of autoantibodies and an IFN-signature. The strength of the IFN-signature positively correlates with disease severity, suggesting that type I IFNs are active players in these diseases. BAFF is a cytokine critical for development and proper selection of B cells, and the targeting of BAFF has emerged as a successful treatment strategy of SLE. Previous reports have suggested that BAFF expression is directly induced by type I IFNs, but the precise mechanism for this remains unknown. In this article, we demonstrate that BAFF is a bona fide ISG and that IFN regulatory factors (IRFs) control the expression of BAFF. We identify IRF1 and IRF2 as positive regulators of BAFF transcription and IRF4 and IRF8 as potent repressors; in addition, we have mapped the precise binding site for these factors in the BAFF promoter. IFN-ß injections induced BAFF expression mainly in neutrophils and monocytes, and BAFF expression in neutrophils from pSS patients strongly correlated with the strength of the IFN-signature. In summary, we show that BAFF expression is directly induced by type I IFNs via IRF1 and IRF2, whereas IRF4 and IRF8 are negative regulators of BAFF expression. These data suggest that type I IFN blockade in SLE and pSS patients will lead to downregulation of BAFF and a consequential reduction of autoreactive B cell clones and autoantibodies.


Subject(s)
B-Cell Activating Factor/immunology , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factor-2/immunology , Interferon Regulatory Factors/immunology , Interferon-beta/immunology , Autoantibodies/immunology , B-Cell Activating Factor/blood , B-Cell Activating Factor/genetics , B-Lymphocytes/immunology , Base Sequence , Binding Sites/genetics , Cells, Cultured , Female , Humans , Lupus Erythematosus, Systemic/immunology , Male , Molecular Sequence Data , Monocytes/immunology , Multiple Sclerosis/immunology , Neutrophils/immunology , Sjogren's Syndrome/immunology , Transcription, Genetic/genetics
8.
J Matern Fetal Neonatal Med ; 29(20): 3286-91, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26699657

ABSTRACT

OBJECTIVE: To evaluate the frequency of uterine rupture following induction of labor in women with a previous cesarean section. Misoprostol was compared to other methods of induction. METHODS: A retrospective cohort study of 208 women attempting induction of labor after one previous cesarean section. Delivery data were collected retrospectively and compared. Group 1(2009-2010) was compared with Group 2 (2012-2013). In Group 1, the main method of induction was vaginal PGE2 (prostaglandin-E2), amniotomy, oxytocin or a balloon catheter. In Group 2, the dominant method of induction was an oral solution of misoprostol. MAIN OUTCOME MEASURES: frequency of uterine rupture in the two groups. RESULTS: Nine cases (4.3%) of uterine rupture occurred. There was no significant difference in the frequency of uterine rupture following the change of method of induction from PGE2, amniotomy, oxytocin or mechanical dilatation with a balloon catheter to orally administered misoprostol (4.1 versus 4.6%, p = 0.9). All ruptures occurred in women with no prior vaginal delivery. CONCLUSION: The shift to oral misoprostol as the primary method of induction in women with a previous cesarean section did not increase the frequency of uterine rupture in the cohort studied.


Subject(s)
Cesarean Section/adverse effects , Labor, Induced/adverse effects , Misoprostol/adverse effects , Oxytocics/adverse effects , Postoperative Complications/etiology , Uterine Rupture/etiology , Administration, Oral , Adult , Female , Humans , Misoprostol/administration & dosage , Oxytocics/administration & dosage , Pregnancy , Retrospective Studies
9.
Mult Scler ; 20(13): 1727-36, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24821684

ABSTRACT

BACKGROUND: Interleukin-7 (IL-7) is a non-redundant cytokine for T-cell development and survival. The IL-7 signaling pathway has been genetically and functionally associated with several autoimmune diseases including multiple sclerosis (MS). OBJECTIVE: The objective of this paper is to elucidate the effect of the widely used immunomodulatory MS therapy interferon beta (IFNß) on IL-7 homeostasis. METHODS: Swedish MS patients were screened for IL-7 concentration in serum and blood cell counts. IL-7 receptor alpha chain (IL-7Rα) expression was determined by semi-quantitative real-time polymerase chain reaction (PCR) and flow cytometry. RESULTS: IFNß treatment led to significantly increased serum IL-7 levels (mean: 17 pg/ml) compared with healthy controls (mean: 7.6 pg/ml) and natalizumab-treated patients (mean: 5.3 pg/ml). In vitro and in vivo, peripheral blood leukocytes showed decreased IL-7Rα expression and IL-7 consumption upon IFNß exposure, suggesting that their IL-7 responsiveness is impaired during treatment. CONCLUSIONS: MS patients undergoing IFNß treatment have increased serum IL-7 levels and decreased IL-7 consumption. Given IL-7's important role in T-cell immunity, this relationship may be highly relevant for IFNß's treatment efficacy.


Subject(s)
Immunosuppressive Agents/therapeutic use , Interferon-beta/therapeutic use , Interleukin-7/blood , Multiple Sclerosis/blood , Multiple Sclerosis/drug therapy , Antibodies, Neutralizing/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Multiple Sclerosis/immunology , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-7/blood , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
J Immunol ; 191(7): 3753-63, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23975864

ABSTRACT

Tripartite-motif 21 (TRIM21) is an E3 ubiquitin ligase that regulates innate immune responses by ubiquitinating IFN regulatory factors (IRFs). TRIM21 is mainly found in hematopoietic cells in which its expression is induced by IFNs during viral. infections and in systemic autoimmune diseases such as systemic lupus erythematosus and Sjögren's syndrome. However, the exact molecular mechanism by which the expression of the Trim21 gene is regulated is unknown. In this study, we demonstrate that IFNs induce Trim21 expression in immune cells via IRFs and that IFN-α and IFN-ß are the most potent inducers of Trim21. A functional IFN-stimulated response element but no conserved IFN-γ-activated site was detected in the promoter of Trim21. IRF1 and IRF2 strongly induced Trim21 expression in an IFN-stimulated response element-dependent manner, whereas IRF4 and IRF8 strongly repressed the IRF1-mediated induction of Trim21. Consistent with this observation, baseline expression of Trim21 was elevated in Irf4(-/-) cells. TRIM21, IRF1, and IRF2 expression was increased in PBMCs from patients with Sjögren's syndrome compared with healthy controls. In contrast, IRF4 and IRF8 expression was not increased in PBMCs from patients. The IFN-γ-mediated induction of Trim21 was completely abolished by inhibiting protein synthesis with cycloheximide, and Trim21 expression could not be induced by IFN-γ in Irf1(-/-) cells, demonstrating that IFN-γ induces Trim21 indirectly via IRF1 and not directly via STAT1 activation. Our data demonstrate that multiple IRFs tightly regulate expression of Trim21 in immune cells, suggesting that a well-controlled expression of the E3 ligase TRIM21 is important for regulation of immune responses.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Ribonucleoproteins/genetics , Animals , Base Sequence , Cell Line , Conserved Sequence , Gene Order , Humans , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-2/metabolism , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Mice , Mice, Transgenic , Promoter Regions, Genetic , Response Elements , STAT1 Transcription Factor/metabolism , Sjogren's Syndrome/genetics , Sjogren's Syndrome/metabolism
11.
J Exp Med ; 206(8): 1661-71, 2009 Aug 03.
Article in English | MEDLINE | ID: mdl-19635858

ABSTRACT

Ro52/Trim21 is targeted as an autoantigen in systemic lupus erythematosus and Sjögren's syndrome. Polymorphisms in the Ro52 gene have been linked to these autoimmune conditions, but the molecular mechanism by which Ro52 may promote development of systemic autoimmune diseases has not been explored. To address this issue, we generated Ro52-null mice (Ro52(-/-)), which appear phenotypically normal if left unmanipulated. However, Ro52(-/-) mice develop severe dermatitis extending from the site of tissue injury induced by ear tags. The affected mice further develop several signs of systemic lupus with hypergammaglobulinemia, autoantibodies to DNA, proteinuria, and kidney pathology. Ro52, which was recently identified as an E3 ligase, mediates ubiquitination of several members of the interferon regulatory factor (IRF) family, and the Ro52-deficient mice have an enhanced production of proinflammatory cytokines that are regulated by the IRF transcription factors, including cytokines involved in the Th17 pathway (interleukin [IL] 6, IL-12/IL-23p40, and IL-17). Loss of IL-23/IL-17 by genetic deletion of IL-23/p19 in the Ro52(-/-) mice conferred protection from skin disease and systemic autoimmunity. These data reveal that the lupus-associated Ro52 protein is an important negative regulator of proinflammatory cytokine production, and they provide a mechanism by which a defective Ro52 function can lead to tissue inflammation and systemic autoimmunity through the IL-23-Th17 pathway.


Subject(s)
Autoimmune Diseases/etiology , Inflammation/etiology , Interleukin-23/metabolism , Ribonucleoproteins/deficiency , Animals , Autoimmune Diseases/pathology , Base Sequence , Cytokines/biosynthesis , DNA Primers/genetics , Disease Models, Animal , Feedback, Physiological , Humans , Inflammation/pathology , Inflammation Mediators/metabolism , Interferons/pharmacology , Interleukin-23/deficiency , Interleukin-23/genetics , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ribonucleoproteins/genetics , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/classification , T-Lymphocytes, Helper-Inducer/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...