Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Res ; 12(2): 195-213, 2024 02 02.
Article in English | MEDLINE | ID: mdl-38091375

ABSTRACT

The costimulatory receptor CD137 (also known as TNFRSF9 or 4-1BB) sustains effective cytotoxic T-cell responses. Agonistic anti-CD137 cancer immunotherapies are being investigated in clinical trials. Development of the first-generation CD137-agonist monotherapies utomilumab and urelumab was unsuccessful due to low antitumor efficacy mediated by the epitope recognized on CD137 or hepatotoxicity mediated by Fcγ receptors (FcγR) ligand-dependent CD137 activation, respectively. M9657 was engineered as a tetravalent bispecific antibody (mAb2) in a human IgG1 backbone with LALA mutations to reduce binding to FCγRs. Here, we report that M9657 selectively binds to mesothelin (MSLN) and CD137 with similar affinity in humans and cynomolgus monkeys. In a cellular functional assay, M9657 enhanced CD8+ T cell-mediated cytotoxicity and cytokine release in the presence of tumor cells, which was dependent on both MSLN expression and T-cell receptor/CD3 activation. Both FS122m, a murine surrogate with the same protein structure as M9657, and chimeric M9657, a modified M9657 antibody with the Fab portion replaced with an anti-murine MSLN motif, demonstrated in vivo antitumor efficacy against various tumors in wild-type and human CD137 knock-in mice, and this was accompanied by activated CD8+ T-cell infiltration in the tumor microenvironment. The antitumor immunity of M9657 and FS122m depended on MSLN expression density and the mAb2 structure. Compared with 3H3, a murine surrogate of urelumab, FS122m and chimeric M9657 displayed significantly lower on-target/off-tumor toxicity. Taken together, M9657 exhibits a promising profile for development as a tumor-targeting immune agonist with potent anticancer activity without systemic immune activation and associated hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury , Neoplasms , Humans , Animals , Mice , Mesothelin , Inflammation , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Tumor Microenvironment
2.
J Clin Invest ; 130(4): 1683-1698, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32091410

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) catalyzes symmetric dimethylation (SDM) of arginine, a posttranslational modification involved in oncogenesis and embryonic development. However, the role and mechanisms by which PRMT5 modulates Th cell polarization and autoimmune disease have not yet been elucidated. Here, we found that PRMT5 promoted SREBP1 SDM and the induction of cholesterol biosynthetic pathway enzymes that produce retinoid-related orphan receptor (ROR) agonists that activate RORγt. Specific loss of PRMT5 in the CD4+ Th cell compartment suppressed Th17 differentiation and protected mice from developing experimental autoimmune encephalomyelitis (EAE). We also found that PRMT5 controlled thymic and peripheral homeostasis in the CD4+ Th cell life cycle and invariant NK (iNK) T cell development and CD8+ T cell maintenance. This work demonstrates that PRMT5 expression in recently activated T cells is necessary for the cholesterol biosynthesis metabolic gene expression program that generates RORγt agonistic activity and promotes Th17 differentiation and EAE. These results point to Th PRMT5 and its downstream cholesterol biosynthesis pathway as promising therapeutic targets in Th17-mediated diseases.


Subject(s)
Autoimmunity , Cell Differentiation/immunology , Cholesterol/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Protein-Arginine N-Methyltransferases/immunology , Th17 Cells/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cholesterol/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Transgenic , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Protein-Arginine N-Methyltransferases/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/immunology , Th17 Cells/pathology
3.
Front Immunol ; 10: 524, 2019.
Article in English | MEDLINE | ID: mdl-30941147

ABSTRACT

Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) mediated by CD4+ T cells and modeled via experimental autoimmune encephalomyelitis (EAE). Inhibition of PRMT5, the major Type II arginine methyltransferase, suppresses pathogenic T cell responses and EAE. PRMT5 is transiently induced in proliferating memory inflammatory Th1 cells and during EAE. However, the mechanisms driving PRMT5 protein induction and repression as T cells expand and return to resting is currently unknown. Here, we used naive mouse and memory mouse and human Th1/Th2 cells as models to identify mechanisms controlling PRMT5 protein expression in initial and recall T cell activation. Initial activation of naive mouse T cells resulted in NF-κB-dependent transient Prmt5 transcription and NF-κB, mTOR and MYC-dependent PRMT5 protein induction. In murine memory Th cells, transcription and miRNA loss supported PRMT5 induction to a lesser extent than in naive T cells. In contrast, NF-κB/MYC/mTOR-dependent non-transcriptional PRMT5 induction played a major role. These results highlight the importance of the NF-κB/mTOR/MYC axis in PRMT5-driven pathogenic T cell expansion and may guide targeted therapeutic strategies for MS.


Subject(s)
Lymphocyte Activation/genetics , NF-kappa B/genetics , Protein-Arginine N-Methyltransferases/genetics , Proto-Oncogene Proteins c-myc/genetics , TOR Serine-Threonine Kinases/genetics , Transcription, Genetic/genetics , Animals , Cell Line , Encephalomyelitis, Autoimmune, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/genetics , Th1 Cells/physiology , Th2 Cells/physiology
5.
Trends Mol Med ; 23(6): 546-562, 2017 06.
Article in English | MEDLINE | ID: mdl-28478950

ABSTRACT

Multiple Sclerosis (MS) is a chronic inflammatory disease of the central nervous system. The inflammatory and neurodegenerative pathways driving MS are modulated by DNA, lysine, and arginine methylation, as evidenced by studies made possible by novel tools for methylation detection or loss of function. We present evidence that MS is associated with genetic variants and metabolic changes that impact on methylation. Further, we comprehensively review current understanding of how methylation can impact on central nervous system (CNS) resilience and neuroregenerative potential, as well as inflammatory versus regulatory T helper (Th) cell balance. These findings are discussed in the context of therapeutic relevance for MS, with broad implications in other neurologic and immune-mediated diseases.


Subject(s)
Central Nervous System/immunology , DNA Methylation/immunology , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Central Nervous System/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Methylation , Multiple Sclerosis/pathology , T-Lymphocytes, Regulatory/pathology
6.
Sci Rep ; 7: 40814, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28094319

ABSTRACT

Helminths cause chronic infections and affect the immune response to unrelated inflammatory diseases. Although helminths have been used therapeutically to ameliorate inflammatory conditions, their anti-inflammatory properties are poorly understood. Alternatively activated macrophages (AAMϕs) have been suggested as the anti-inflammatory effector cells during helminth infections. Here, we define the origin of AAMϕs during infection with Taenia crassiceps, and their disease-modulating activity on the Experimental Autoimmune Encephalomyelitis (EAE). Our data show two distinct populations of AAMϕs, based on the expression of PD-L1 and PD-L2 molecules, resulting upon T. crassiceps infection. Adoptive transfer of Ly6C+ monocytes gave rise to PD-L1+/PD-L2+, but not PD-L1+/PD-L2- cells in T. crassiceps-infected mice, demonstrating that the PD-L1+/PD-L2+ subpopulation of AAMϕs originates from blood monocytes. Furthermore, adoptive transfer of PD-L1+/PD-L2+ AAMϕs into EAE induced mice reduced disease incidence, delayed disease onset, and diminished the clinical disability, indicating the critical role of these cells in the regulation of autoimmune disorders.


Subject(s)
Adoptive Transfer/methods , Antigens, Ly/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Macrophage Activation , Monocyte-Macrophage Precursor Cells/immunology , Taenia/immunology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism
7.
J Immunol ; 198(4): 1439-1451, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087667

ABSTRACT

In the autoimmune disease multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), expansion of pathogenic, myelin-specific Th1 cell populations drives active disease; selectively targeting this process may be the basis for a new therapeutic approach. Previous studies have hinted at a role for protein arginine methylation in immune responses, including T cell-mediated autoimmunity and EAE. However, a conclusive role for the protein arginine methyltransferase (PRMT) enzymes that catalyze these reactions has been lacking. PRMT5 is the main PRMT responsible for symmetric dimethylation of arginine residues of histones and other proteins. PRMT5 drives embryonic development and cancer, but its role in T cells, if any, has not been investigated. In this article, we show that PRMT5 is an important modulator of CD4+ T cell expansion. PRMT5 was transiently upregulated during maximal proliferation of mouse and human memory Th cells. PRMT5 expression was regulated upstream by the NF-κB pathway, and it promoted IL-2 production and proliferation. Blocking PRMT5 with novel, highly selective small molecule PRMT5 inhibitors severely blunted memory Th expansion, with preferential suppression of Th1 cells over Th2 cells. In vivo, PRMT5 blockade efficiently suppressed recall T cell responses and reduced inflammation in delayed-type hypersensitivity and clinical disease in EAE mouse models. These data implicate PRMT5 in the regulation of adaptive memory Th cell responses and suggest that PRMT5 inhibitors may be a novel therapeutic approach for T cell-mediated inflammatory disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Immunologic Memory , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/physiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Cytokines/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Regulation , Humans , Inflammation , Interleukin-2/biosynthesis , Interleukin-2/immunology , Lymphocyte Activation , Methylation , Mice , NF-kappa B/immunology , Protein-Arginine N-Methyltransferases/genetics , Th1 Cells/immunology , Th2 Cells/immunology , Up-Regulation
8.
PLoS One ; 10(12): e0145342, 2015.
Article in English | MEDLINE | ID: mdl-26699615

ABSTRACT

Classically (M1) and alternatively activated (M2) macrophages exhibit distinct phenotypes and functions. It has been difficult to dissect macrophage phenotypes in vivo, where a spectrum of macrophage phenotypes exists, and also in vitro, where low or non-selective M2 marker protein expression is observed. To provide a foundation for the complexity of in vivo macrophage phenotypes, we performed a comprehensive analysis of the transcriptional signature of murine M0, M1 and M2 macrophages and identified genes common or exclusive to either subset. We validated by real-time PCR an M1-exclusive pattern of expression for CD38, G-protein coupled receptor 18 (Gpr18) and Formyl peptide receptor 2 (Fpr2) whereas Early growth response protein 2 (Egr2) and c-Myc were M2-exclusive. We further confirmed these data by flow cytometry and show that M1 and M2 macrophages can be distinguished by their relative expression of CD38 and Egr2. Egr2 labeled more M2 macrophages (~70%) than the canonical M2 macrophage marker Arginase-1, which labels 24% of M2 macrophages. Conversely, CD38 labeled most (71%) in vitro M1 macrophages. In vivo, a similar CD38+ population greatly increased after LPS exposure. Overall, this work defines exclusive and common M1 and M2 signatures and provides novel and improved tools to distinguish M1 and M2 murine macrophages.


Subject(s)
Biomarkers/metabolism , High-Throughput Nucleotide Sequencing/methods , Inflammation/metabolism , Macrophage Activation/physiology , Macrophages/metabolism , Transcriptome , Animals , Flow Cytometry , Immunoenzyme Techniques , Inflammation/chemically induced , Inflammation/genetics , Lipopolysaccharides/toxicity , Macrophages/cytology , Mice , Mice, Inbred C57BL , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
Inorg Chem ; 53(11): 5781-7, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24881486

ABSTRACT

The trimeric clusters [Fe(III)3(X-Sal-AHA)3(µ3-OCH3)](-), where X-Sal-AHA is a tetradentate chelate incorporating an α-hydroxy acid moiety (AHA) and a salicylidene moiety (X-Sal with X being 5-NO2, 3,5-diCl, all-H, 3-OCH3, or 3,5-di-t-Bu substituents on the phenolate ring), undergo a photochemical reaction resulting in reduction of two Fe(III) to Fe(II) for each AHA group that is oxidatively cleaved. However, photolysis of structurally analogous mixed Fe/Ga clusters demonstrate that a similar photolysis reaction will occur with only a single Fe(III) in the cluster. Quantum yields of iron reduction for the series of [Fe(III)3(X-Sal-AHA)3(µ3-OCH3)](-) complexes measured by monitoring Fe(II) production are twice those for ligand oxidation, measured by loss of the CD signal for the complex due to cleavage of the chiral AHA group.The quantum yields, 2-13% in the UVA and UVB ranges, are higher for complexes with electron-withdrawing X groups than for those with electron-donating X groups [corrected]. The observed final photolysis product of the chelate is different if irradiation is done in the air than if it is done under Ar. The first observed photochemical product is the aldehyde resulting from decarboxylation of the AHA. This is the final product under anaerobic conditions. In air, this is followed by an Fe- and O2-dependent reaction oxidizing the aldehyde to the corresponding carboxylate, then a second Fe- and light-dependent decarboxylation reaction giving a product that is two carbons smaller than the initial ligand. These reactivity studies have important biological implications for the photoactive marine siderophores. They suggest that different types of photochemical products for different siderophore structure types do not result from different initial photochemical steps, but rather from different susceptibility of the initial photochemical product to air oxidation.


Subject(s)
Hydroxy Acids/chemistry , Iron Compounds/chemistry , Photolysis , Siderophores/chemistry , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...