Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Int J Mol Sci ; 25(3)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38338672

ABSTRACT

The search for target antigens for CAR-T cell therapy against multiple myeloma defined the B-cell maturation antigen (BCMA) as an interesting candidate. Several studies with BCMA-directed CAR-T cell therapy showed promising results. Second-generation point-of-care BCMA.CAR-T cells were manufactured to be of a GMP (good manufacturing practice) standard using the CliniMACS Prodigy® device. Cytokine release in BCMA.CAR-T cells after stimulation with BCMA positive versus negative myeloma cell lines, U266/HL60, was assessed via intracellular staining and flow cytometry. The short-term cytotoxic potency of CAR-T cells was evaluated by chromium-51 release, while the long-term potency used co-culture (3 days/round) at effector/target cell ratios of 1:1 and 1:4. To evaluate the activation and exhaustion of CAR-T cells, exhaustion markers were assessed via flow cytometry. Stability was tested through a comparison of these evaluations at different timepoints: d0 as well as d + 14, d + 90 and d + 365 of cryopreservation. As results, (1) Killing efficiency of U266 cells correlated with the dose of CAR-T cells in a classical 4 h chromium-release assay. There was no significant difference after cryopreservation on different timepoints. (2) In terms of endurance of BCMA.CAR-T cell function, BCMA.CAR-T cells kept their ability to kill all tumor cells over six rounds of co-culture. (3) BCMA.CAR-T cells released high amounts of cytokines upon stimulation with tumor cells. There was no significant difference in cytokine release after cryopreservation. According to the results, BCMA.CAR-T cells manufactured under GMP conditions exerted robust and specific killing of target tumor cells with a high release of cytokines. Even after 1 year of cryopreservation, cytotoxic functions were maintained at the same level. This gives clinicians sufficient time to adjust the timepoint of BCMA.CAR-T cell application to the patient's course of the underlying disease.


Subject(s)
Multiple Myeloma , Receptors, Chimeric Antigen , Humans , B-Cell Maturation Antigen/metabolism , Point-of-Care Systems , Immunotherapy, Adoptive/methods , Multiple Myeloma/pathology , Cytokines/metabolism , T-Lymphocytes , Cryopreservation
2.
Mol Ther ; 30(11): 3358-3378, 2022 11 02.
Article in English | MEDLINE | ID: mdl-35821635

ABSTRACT

Chimeric antigen receptor (CAR) T cells have revolutionized treatment of B cell malignancies. However, enhancing the efficacy of engineered T cells without compromising their safety is warranted. The estrogen receptor-binding fragment-associated antigen 9 (EBAG9) inhibits release of cytolytic enzymes from cytotoxic T lymphocytes. Here, we examined the potency of EBAG9 silencing for the improvement of adoptive T cell therapy. MicroRNA (miRNA)-mediated EBAG9 downregulation in transplanted cytolytic CD8+ T cells (CTLs) from immunized mice improved their cytolytic competence in a tumor model. In tolerant female recipient mice that received organ transplants, a minor histocompatibility antigen was turned into a rejection antigen by Ebag9 deletion, indicating an immune checkpoint function for EBAG9. Considerably fewer EBAG9-silenced human CAR T cells were needed for tumor growth control in a xenotransplantation model. Transcriptome profiling did not reveal additional risks regarding genotoxicity or aberrant differentiation. A single-step retrovirus transduction process links CAR or TCR expression with miRNA-mediated EBAG9 downregulation. Despite higher cytolytic efficacy, release of cytokines associated with cytokine release syndrome remains unaffected. Collectively, EBAG9 silencing enhances effector capacity of TCR- and CAR-engineered T cells, results in improved tumor eradication, facilitates efficient manufacturing, and decreases the therapeutic dose.


Subject(s)
Antigens, Neoplasm , Immunotherapy, Adoptive , Neoplasms , Animals , Female , Humans , Mice , MicroRNAs/genetics , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Cytotoxic , Gene Silencing , Immune Checkpoint Proteins , Antigens, Neoplasm/genetics
3.
Proc Natl Acad Sci U S A ; 117(42): 26328-26339, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33020261

ABSTRACT

Dendritic cell (DC) maturation is a prerequisite for the induction of adaptive immune responses against pathogens and cancer. Transcription factor (TF) networks control differential aspects of early DC progenitor versus late-stage DC cell fate decisions. Here, we identified the TF C/EBPß as a key regulator for DC maturation and immunogenic functionality under homeostatic and lymphoma-transformed conditions. Upon cell-specific deletion of C/EBPß in CD11c+MHCIIhi DCs, gene expression profiles of splenic C/EBPß-/- DCs showed a down-regulation of E2F cell cycle target genes and associated proliferation signaling pathways, whereas maturation signatures were enriched. Total splenic DC cell numbers were modestly increased but differentiation into cDC1 and cDC2 subsets were unaltered. The splenic CD11c+MHCIIhiCD64+ DC compartment was also increased, suggesting that C/EBPß deficiency favors the expansion of monocytic-derived DCs. Expression of C/EBPß could be mimicked in LAP/LAP* isoform knockin DCs, whereas the short isoform LIP supported a differentiation program similar to deletion of the full-length TF. In accordance with E2F1 being a negative regulator of DC maturation, C/EBPß-/- bone marrow-derived DCs matured much faster enabling them to activate and polarize T cells stronger. In contrast to a homeostatic condition, lymphoma-exposed DCs exhibited an up-regulation of the E2F transcriptional pathways and an impaired maturation. Pharmacological blockade of C/EBPß/mTOR signaling in human DCs abrogated their protumorigenic function in primary B cell lymphoma cocultures. Thus, C/EBPß plays a unique role in DC maturation and immunostimulatory functionality and emerges as a key factor of the tumor microenvironment that promotes lymphomagenesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Dendritic Cells/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Differentiation , Cell Line , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Protein Isoforms/genetics , Signal Transduction , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Microenvironment/physiology
4.
Cell Mol Life Sci ; 78(7): 3525-3542, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33469705

ABSTRACT

Metastasis Associated in Colon Cancer 1 (MACC1) is a novel prognostic, predictive and causal biomarker for tumor progression and metastasis in many cancer types, including colorectal cancer. Besides its clinical value, little is known about its molecular function. Its similarity to SH3BP4, involved in regulating uptake and recycling of transmembrane receptors, suggests a role of MACC1 in endocytosis. By exploring the MACC1 interactome, we identified the clathrin-mediated endocytosis (CME)-associated proteins CLTC, DNM2 and AP-2 as MACC1 binding partners. We unveiled a MACC1-dependent routing of internalized transferrin receptor towards recycling. Elevated MACC1 expression caused also the activation and internalization of EGFR, a higher rate of receptor recycling, as well as earlier and stronger receptor activation and downstream signaling. These effects are limited by deletion of CME-related protein interaction sites in MACC1. Thus, MACC1 regulates CME and receptor recycling, causing increased growth factor-mediated downstream signaling and cell proliferation. This novel mechanism unveils potential therapeutic intervention points restricting MACC1-driven metastasis.


Subject(s)
Clathrin/metabolism , Colorectal Neoplasms/pathology , Endocytosis , Gene Expression Regulation, Neoplastic , Receptors, Transferrin/metabolism , Trans-Activators/metabolism , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Mice , Proteome/analysis , Proteome/metabolism , Receptors, Transferrin/genetics , Trans-Activators/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055086

ABSTRACT

Chimeric-antigen-receptor (CAR)-T-cell therapy is already widely used to treat patients who are relapsed or refractory to chemotherapy, antibodies, or stem-cell transplantation. Multiple myeloma still constitutes an incurable disease. CAR-T-cell therapy that targets BCMA (B-cell maturation antigen) is currently revolutionizing the treatment of those patients. To monitor and improve treatment outcomes, methods to detect CAR-T cells in human peripheral blood are highly desirable. In this study, three different detection reagents for staining BCMA-CAR-T cells by flow cytometry were compared. Moreover, a quantitative polymerase chain reaction (qPCR) to detect BCMA-CAR-T cells was established. By applying a cell-titration experiment of BCMA-CAR-T cells, both methods were compared head-to-head. In flow-cytometric analysis, the detection reagents used in this study could all detect BCMA-CAR-T cells at a similar level. The results of false-positive background staining differed as follows (standard deviation): the BCMA-detection reagent used on the control revealed a background staining of 0.04% (±0.02%), for the PE-labeled human BCMA peptide it was 0.25% (±0.06%) and for the polyclonal anti-human IgG antibody it was 7.2% (±9.2%). The ability to detect BCMA-CAR-T cells down to a concentration of 0.4% was similar for qPCR and flow cytometry. The qPCR could detect even lower concentrations (0.02-0.01%). In summary, BCMA-CAR-T-cell monitoring can be reliably performed by both flow cytometry and qPCR. In flow cytometry, reagents with low background staining should be preferred.


Subject(s)
B-Cell Maturation Antigen/metabolism , Flow Cytometry , Polymerase Chain Reaction , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , B-Cell Maturation Antigen/genetics , Biomarkers , Flow Cytometry/methods , Flow Cytometry/standards , Humans , Immunophenotyping , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/standards , Real-Time Polymerase Chain Reaction , Receptors, Chimeric Antigen/genetics , Reproducibility of Results , Sensitivity and Specificity , T-Lymphocytes/immunology
6.
Immunity ; 37(5): 854-66, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23084031

ABSTRACT

Immunological control of infections or tumors depends on the release of effector cytokines and polarized secretion of cytotoxic granules from T cells and natural killer cells. Here we show that the sorting receptor Sortilin controlled both processes. In murine Sortilin-deficient cytotoxic T lymphocytes, regulated secretion of granzyme A and cytotoxic killing was enhanced and correlated with increased vesicle-associated membrane protein 7 availability. In contrast, loss of Sortilin reduced the release of interferon-γ upon infections and in autoimmune colitis. Exit of interferon-γ from the Golgi apparatus required the presence of Sortilin. Furthermore, we tracked the transport route of interferon-γ beyond this Sortilin-dependent Golgi to early endosome step. In wild-type T cells, trafficking of interferon-γ from the endosomal sorting platform to the plasma membrane proceeded independently of recycling endosomes, and interferon-γ remained excluded from late endosomes. Our results suggest that Sortilin modulates systemic immune responses through exocytic sorting of immunological effector molecules.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Granzymes/metabolism , Interferon-gamma/metabolism , T-Lymphocytes/metabolism , Adaptor Proteins, Vesicular Transport/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , CD4 Antigens/immunology , CD4 Antigens/metabolism , Cell Membrane/immunology , Cell Membrane/metabolism , Colitis/immunology , Colitis/metabolism , Endosomes/immunology , Endosomes/metabolism , Exocytosis/immunology , Golgi Apparatus/immunology , Golgi Apparatus/metabolism , Granzymes/immunology , Interferon-gamma/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Protein Transport , R-SNARE Proteins/immunology , R-SNARE Proteins/metabolism , T-Lymphocytes/immunology , Transport Vesicles/immunology , Transport Vesicles/metabolism
7.
Mol Ther ; 26(8): 1906-1920, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30078440

ABSTRACT

Autologous T cells genetically modified with a chimeric antigen receptor (CAR) redirected at CD19 have potent activity in the treatment of B cell leukemia and B cell non-Hodgkin's lymphoma (B-NHL). Immunotherapies to treat multiple myeloma (MM) targeted the B cell maturation antigen (BCMA), which is expressed in most cases of MM. We developed a humanized CAR with specificity for BCMA based on our previously generated anti-BCMA monoclonal antibody. The targeting single-chain variable fragment (scFv) domain exhibited a binding affinity in the low nanomolar range, conferring T cells with high functional avidity. Redirecting T cells by this CAR allowed us to explore BCMA as an alternative target for mature B-NHLs. We validated BCMA expression in diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma, and chronic lymphocytic leukemia. BCMA CAR T cells triggered target cell lysis with an activation threshold in the range of 100 BCMA molecules, which allowed for an efficient eradication of B-NHL cells in vitro and in vivo. Our data corroborate BCMA is a suitable target in B cell tumors beyond MM, providing a novel therapeutic option for patients where BCMA is expressed at low abundance or where anti-CD19 immunotherapies have failed due to antigen loss.


Subject(s)
B-Cell Maturation Antigen/immunology , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/transplantation , Animals , Cell Line, Tumor , Humans , Immunotherapy, Adoptive , Jurkat Cells , Lymphoma, B-Cell/immunology , Mice , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
8.
FASEB J ; 30(2): 761-74, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26499265

ABSTRACT

Interleukin 17-producing helper T (Th17) cells have been widely defined by the lineage transcription factor retinoid-related orphan receptor (ROR)γt. Pathophysiologically, these cells play a crucial role in autoimmune diseases and have been linked to dysregulated germinal center (GC) reactions and autoantibody production. In this study, we used gene expression and flow cytometric analyses for the characterization of Rorγt(-/-) and Rorγt(-/-)Il21(RFP/+) mice to demonstrate a previously unknown transcriptional flexibility in the development of IL-17-producing Th-cell subsets. We found an accumulation of follicular Th (Tfh) cells by 5.2-fold, spontaneous 13-fold higher GC formation, decreased frequency of follicular Foxp3(+) T-regulatory (Treg) cells (50%), and a 3.4-fold increase in the number of proliferating follicular B cells in RORγt-deficient vs. wild-type mice. Dysregulated B-cell responses were associated with enhanced production of IL-17 (6.4-fold), IL-21 (2.2-fold), and B-cell-activating factor (BAFF) (2-fold) and were partially rescued by adoptive transfer of Treg cells. In an unexpected finding, we detected RORγt-independent IL-17 expression in ICOS(+)CXCR5(+)Tfh and in ICOS(+)CXCR5(-)Th cells. Based on the observed high Irf4 and Batf gene expression, we suggest that CD4(+) T-cell transcription factors other than RORγt can cooperatively induce differentiation of IL-17-producing Th cells, including Th17-like Tfh-cell subsets. We conclude that the occurrence of aberrant Tfh and follicular Treg cells support spontaneous GC formation and dysregulated B-cell responses in RORγt-deficient mice.


Subject(s)
Cell Differentiation/immunology , Germinal Center/immunology , Interleukin-17/immunology , Interleukins/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , T-Lymphocytes, Regulatory/immunology , Animals , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/genetics , Germinal Center/cytology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukin-17/genetics , Interleukins/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , T-Lymphocytes, Regulatory/cytology
9.
Front Immunol ; 15: 1383136, 2024.
Article in English | MEDLINE | ID: mdl-38979422

ABSTRACT

Multiple myeloma (MM) is a plasma cell disease with a preferential bone marrow (BM) tropism. Enforced expression of tissue-specific chemokine receptors has been shown to successfully guide adoptively-transferred CAR NK cells towards the malignant milieu in solid cancers, but also to BM-resident AML and MM. For redirection towards BM-associated chemokine CXCL12, we armored BCMA CAR-NK-92 as well as primary NK cells with ectopic expression of either wildtype CXCR4 or a gain-of-function mutant CXCR4R334X. Our data showed that BCMA CAR-NK-92 and -primary NK cells equipped with CXCR4 gained an improved ability to migrate towards CXCL12 in vitro. Beyond its classical role coordinating chemotaxis, CXCR4 has been shown to participate in T cell co-stimulation, which prompted us to examine the functionality of CXCR4-cotransduced BCMA-CAR NK cells. Ectopic CXCR4 expression enhanced the cytotoxic capacity of BCMA CAR-NK cells, as evidenced by the ability to eliminate BCMA-expressing target cell lines and primary MM cells in vitro and through accelerated cytolytic granule release. We show that CXCR4 co-modification prolonged BCMA CAR surface deposition, augmented ZAP-70 recruitment following CAR-engagement, and accelerated distal signal transduction kinetics. BCMA CAR sensitivity towards antigen was enhanced by virtue of an enhanced ZAP-70 recruitment to the immunological synapse, revealing an increased propensity of CARs to become triggered upon CXCR4 overexpression. Unexpectedly, co-stimulation via CXCR4 occurred in the absence of CXCL12 ligand-stimulation. Collectively, our findings imply that co-modification of CAR-NK cells with tissue-relevant chemokine receptors affect adoptive NK cell therapy beyond improved trafficking and retention within tumor sites.


Subject(s)
B-Cell Maturation Antigen , Chemokine CXCL12 , Immunotherapy, Adoptive , Killer Cells, Natural , Multiple Myeloma , Receptors, CXCR4 , Receptors, Chimeric Antigen , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Humans , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , B-Cell Maturation Antigen/immunology , B-Cell Maturation Antigen/metabolism , B-Cell Maturation Antigen/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Immunotherapy, Adoptive/methods , Chemokine CXCL12/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic
10.
J Autoimmun ; 47: 58-72, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24075646

ABSTRACT

Autoimmunity is associated with a strong genetic component, but onset and persistence of clinically apparent autoimmune diseases often require an additional environmental trigger. The balance between immunity and tolerance is regulated by numerous molecular factors including nuclear hormone and homeostatic chemokine receptors. The nuclear hormone receptor RORγt and the chemokine receptor CCR7 are both essentially involved in functional lymphoid organogenesis and maintenance of lymphocyte homeostasis. Lack of one or the other impairs thymic T cell development and alters T cell homeostasis. Mice deficient for both, Ccr7(-/-)Rorγt(-/-), succumbed early to acute destructive inflammation, characterized by massive recruitment of inflammatory leukocytes, pro-inflammatory cytokine and autoantibody production, and wasting disease. Antibiotic-treatment of mice before disease onset reduced the overall gut microflora and abrogated the development of fatal mucosal inflammation. Hence, commensal bacteria and a confined tissue-specific inflammatory milieu serve as complementary trigger to initiate the lethal pathophysiologic process in Ccr7(-/-)Rorγt(-/-) mice.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/immunology , Intestinal Mucosa/microbiology , Microbiota , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptors, CCR7/genetics , Ampicillin/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Autoantibodies/immunology , Autoimmune Diseases/microbiology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Chimera/immunology , Colistin/therapeutic use , Inflammation/immunology , Intestinal Mucosa/immunology , Leukocytes/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Streptomycin/therapeutic use
11.
Blood ; 118(4): 1020-33, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21586747

ABSTRACT

Lymphoma cell survival and progression are putatively dependent on a specific microanatomic localization within secondary lymphoid organs. Despite compelling data correlating homeostatic chemokine receptor expression and human lymphoma pathogenesis, genetic models that either mimic lymphoma dissemination or dissect a crosstalk of lymphoma and stromal cells are missing. Applying the genetically tractable Eµ-Myc transgenic mouse model, we show that the chemokine receptor CCR7 regulates Eµ-Myc lymphoma homing to lymph nodes and distinctive microanatomic sites of the spleen. CCR7-controlled access of lymphoma cells to the splenic T-cell zone led to a significant survival advantage compared with CCR7-deficient lymphoma cells, which were excluded from this zone. Within the niche, lymphoma cells stimulated a reciprocal cross-talk with gp38(+) fibroblastic reticular cells. This reciprocal cooperation program was mediated by lymphoma B cell-presented lymphotoxin, which acted on lymphotoxin-ß-receptor-bearing stromal cells followed by alteration of stromal cellular composition. Cross-talk inhibition by lymphotoxin-α deletion and using a lymphotoxin-ß receptor-immunoglobulin fusion protein impaired lymphoma growth. Thus, abrogation of CCR7-governed migration and of sustained lymphotoxin signaling could provide new targets in lymphoma therapy.


Subject(s)
Lymphoid Tissue/pathology , Lymphoma, B-Cell/pathology , Lymphotoxin-alpha/metabolism , Receptors, CCR7/metabolism , Tumor Microenvironment/physiology , Adoptive Transfer , Animals , Cell Movement , Cell Separation , Disease Progression , Flow Cytometry , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Nick-End Labeling , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Lymphotoxin-alpha/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Receptor Cross-Talk/physiology , Receptors, CCR7/immunology , Receptors, Lymphocyte Homing/immunology , Receptors, Lymphocyte Homing/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
12.
Eur J Immunol ; 41(9): 2774-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21630249

ABSTRACT

Coxsackievirus B3 (CVB3)-infection is a frequent cause of acute myocarditis, which may result in chronic myocarditis and virus persistence. Investigation of the initial immune responses to CVB3 may shed light on the mechanisms that contribute to ongoing disease. DCs, as key professional APCs, were investigated in two MHC-matched hosts: while C57BL/6 mice are resistant to chronic CVB3-myocarditis, the A.BY/SnJ mouse strain exhibits susceptibility. DC maturation and activation were critically impaired in A.BY/SnJ mice, as reflected by the failure of DCs to induce co-stimulatory molecules and cytokine/chemokine responses. MHC class I-restricted antigen presentation via the cross-presentation pathway was also affected in DCs from A.BY/SnJ mice. DC maturation involves the accumulation of DC aggresome-like induced structures (DALISs) and the transient storage of defective ribosomal products (DRiPs). DCs from A.BY/SnJ mice showed permanent DALIS accumulation; the detection of poly-ubiquitinylated CVB3 proteins in these DALISs suggested a limitation in the MHC class I antigenic supply in this host. In conclusion, ongoing chronic disease in A.BY/SnJ mice due to a failure to clear the virus may be attributed to defects in DC maturation/activation and DC MHC class I antigen processing.


Subject(s)
Coxsackievirus Infections/immunology , Cross-Priming , Dendritic Cells/metabolism , Enterovirus/immunology , Histocompatibility Antigens Class I/metabolism , Animals , Cell Differentiation , Cells, Cultured , Coxsackievirus Infections/complications , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Enterovirus/pathogenicity , Histocompatibility Antigens Class I/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocarditis/etiology , Ubiquitination , Virulence
13.
Am J Pathol ; 179(2): 754-65, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21801869

ABSTRACT

Autoimmune gastritis is a common autoimmune disorder characterized by chronic inflammatory cell infiltrates, atrophy of the corpus and fundus, and the occurrence of autoantibodies to parietal cell antigen. In CCR7-deficient mice, autoimmune gastritis developed spontaneously and was accompanied by metaplasia of the gastric mucosa and by the formation of tertiary lymphoid organs at gastric mucosal sites. T cells of CCR7-deficient mice showed an activated phenotype in the gastric mucosa, mesenteric lymph nodes, and peripheral blood. In addition, elevated serum IgG levels specific to gastric parietal cell antigen were detected. Because the role of organized lymphocytic aggregates at this inflammatory site is not completely understood, we first analyzed the cellular requirements for the formation of these structures. Autoreactive CD4(+) T cells were pivotal for tertiary lymphoid follicle formation, most likely in cooperation with dendritic cells, macrophages, and B cells. Second, we analyzed the necessity of secondary lymph nodes and tertiary lymphoid organs for the development of autoimmune gastritis using CCR7 single- and CCR7/lymphotoxin α double-deficient mice. Strikingly, manifestation of autoimmune gastritis was observed in the absence of secondary lymph nodes and preceded the development of tertiary lymphoid organs. Taken together, these findings identify an inflammatory process where gastric autoreactive T cells independent of organized tertiary lymphoid organs and classic lymph nodes can induce and maintain autoimmune gastritis.


Subject(s)
Autoimmune Diseases/pathology , Gastritis/pathology , Receptors, CCR7/genetics , Animals , Bone Marrow Cells/cytology , CD11c Antigen/biosynthesis , CD4-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Flow Cytometry/methods , Gastric Mucosa/metabolism , Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Interferon-gamma/metabolism , Interleukin-17/metabolism , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
STAR Protoc ; 3(2): 101267, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35403006

ABSTRACT

Blood endothelial cells (BECs) in lymph nodes are distinct stromal cells with a transcriptional profile allowing fast and specific adaptation to the functional requirements. Here, we describe a step-by-step protocol for the enzymatic digestion of lymph nodes, the enrichment of stromal cells, the sorting of BECs, and the processing of BEC-related data for modern analysis approaches as spectral flow cytometry and single-cell RNA sequencing (scRNA-seq). For complete details on the use and execution of this protocol, please refer to Menzel et al. (2021).


Subject(s)
Endothelial Cells , Single-Cell Analysis , Animals , Flow Cytometry/methods , Lymph Nodes/pathology , Mice , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
15.
Mol Ther Methods Clin Dev ; 24: 181-198, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35118163

ABSTRACT

The advent of CAR T cells targeting CD19 or BCMA on B cell neoplasm demonstrated remarkable efficacy, but rapid relapses and primary refractoriness remains challenging. A leading cause of CAR T cell failure is their lack of expansion and limited persistence. Long-lived, self-renewing multipotent T memory stem cells (TSCM) and T central memory cells (TCM) likely sustain superior tumor regression, but their low frequencies in blood from cancer patients impose a major hurdle for clinical CAR T production. We designed a clinically compliant protocol for generating BCMA CAR T cells starting with increased TSCM/TCM cell input. A CliniMACS Prodigy process was combined with flow cytometry-based enrichment of CD62L+CD95+ T cells. Although starting with only 15% of standard T cell input, the selected TSCM/TCM material was efficiently activated and transduced with a BCMA CAR-encoding retrovirus. Cultivation in the presence of IL-7/IL-15 enabled the harvest of CAR T cells containing an increased CD4+ TSCM fraction and 70% TSCM cells amongst CD8+. Strong cell proliferation yielded cell numbers sufficient for clinical application, while effector functions were maintained. Together, adaptation of a standard CliniMACS Prodigy protocol to low input numbers resulted in efficient retroviral transduction with a high CAR T cell yield.

16.
JCI Insight ; 7(11)2022 06 08.
Article in English | MEDLINE | ID: mdl-35482418

ABSTRACT

Insight into processes that determine CD8+ T cell memory formation has been obtained from infection models. These models are biased toward an inflammatory milieu and often use high-avidity CD8+ T cells in adoptive-transfer procedures. It is unclear whether these conditions mimic the differentiation processes of an endogenous repertoire that proceed upon noninflammatory conditions prevailing in premalignant tumor lesions. We examined the role of cytolytic capacity on CD8+ T cell fate decisions when primed by tumor cells or by minor histocompatibility antigen-mismatched leukocytes. CD8+ memory commitment was analyzed in Ebag9-deficient mice that exhibited enhanced tumor cell lysis. This property endowed Ebag9-/- mice with extended control of Tcl-1 oncogene-induced chronic lymphocytic leukemia progression. In Ebag9-/- mice, an expanded memory population was obtained for anti-HY and anti-SV-40 T antigen-specific T cells, despite unchanged effector frequencies in the primary response. By comparing the single-cell transcriptomes of CD8+ T cells responding to tumor cell vaccination, we found differential distribution of subpopulations between Ebag9+/+ and Ebag9-/- T cells. In Ebag9-/- cells, these larger clusters contained genes encoding transcription factors regulating memory cell differentiation and anti-apoptotic gene functions. Our findings link EBAG9-controlled cytolytic activity and the commitment to the CD8+ memory lineage.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Adoptive Transfer , Animals , Mice , Minor Histocompatibility Antigens
17.
Mol Ther Methods Clin Dev ; 25: 311-330, 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35573047

ABSTRACT

Chimeric antigen receptor (CAR) redirected T cells are potent therapeutic options against hematological malignancies. The current dominant manufacturing approach for CAR T cells depends on retroviral transduction. With the advent of gene editing, insertion of a CD19-CAR into the T cell receptor (TCR) alpha constant (TRAC) locus using adeno-associated viruses for gene transfer was demonstrated, and these CD19-CAR T cells showed improved functionality over their retrovirally transduced counterparts. However, clinical-grade production of viruses is complex and associated with extensive costs. Here, we optimized a virus-free genome-editing method for efficient CAR insertion into the TRAC locus of primary human T cells via nuclease-assisted homology-directed repair (HDR) using CRISPR-Cas and double-stranded template DNA (dsDNA). We evaluated DNA-sensor inhibition and HDR enhancement as two pharmacological interventions to improve cell viability and relative CAR knockin rates, respectively. While the toxicity of transfected dsDNA was not fully prevented, the combination of both interventions significantly increased CAR knockin rates and CAR T cell yield. Resulting TRAC-replaced CD19-CAR T cells showed antigen-specific cytotoxicity and cytokine production in vitro and slowed leukemia progression in a xenograft mouse model. Amplicon sequencing did not reveal significant indel formation at potential off-target sites with or without exposure to DNA-repair-modulating small molecules. With TRAC-integrated CAR+ T cell frequencies exceeding 50%, this study opens new perspectives to exploit pharmacological interventions to improve non-viral gene editing in T cells.

18.
Nat Commun ; 13(1): 7341, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36446823

ABSTRACT

Allogeneic natural killer (NK) cell adoptive transfer is a promising treatment for several cancers but is less effective for the treatment of multiple myeloma. In this study, we report on quadruple gene-engineered induced pluripotent stem cell (iPSC)-derived NK cells designed for mass production from a renewable source and for dual targeting against multiple myeloma through the introduction of an NK cell-optimized chimeric antigen receptor (CAR) specific for B cell maturation antigen (BCMA) and a high affinity, non-cleavable CD16 to augment antibody-dependent cellular cytotoxicity when combined with therapeutic anti-CD38 antibodies. Additionally, these cells express a membrane-bound interleukin-15 fusion molecule to enhance function and persistence along with knock out of CD38 to prevent antibody-mediated fratricide and enhance NK cell metabolic fitness. In various preclinical models, including xenogeneic adoptive transfer models, quadruple gene-engineered NK cells consistently demonstrate durable antitumor activity independent of exogenous cytokine support. Results presented here support clinical translation of this off-the-shelf strategy for effective treatment of multiple myeloma.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Killer Cells, Natural , B-Cell Maturation Antigen , Receptors, Natural Killer Cell , NK Cell Lectin-Like Receptor Subfamily D
19.
FASEB J ; 24(10): 4000-19, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20570965

ABSTRACT

Many proteins mature within the secretory pathway by the acquisition of glycans. Failure to maintain the proper distribution of the glycosylation machinery might lead to disease. High expression levels of the ubiquitous Golgi protein estrogen receptor-binding fragment-associated gene 9 (EBAG9) in human tumors correlate with poor clinical prognosis, and EBAG9 overexpression in epithelial cell lines induces truncated glycans, typical of many carcinomas. Here, we addressed the pathogenetic link between EBAG9 expression and the alteration of the cellular glycome. We applied confocal microscopy, live imaging, pulse-chase labeling in conjunction with immunoprecipitation, and enzymatic activity assays in a variety of EBAG9-overexpressing or depleted epithelial tumor cell lines. EBAG9 shuttles between the ER-Golgi intermediate compartment and the cis-Golgi, and we demonstrate association of EBAG9 with coat protein complex I (COPI)-coated transport vesicles. EBAG9 overexpression imposes delay of endoplasmic reticulum-to-Golgi transport and mislocalizes components of the ER quality control and glycosylation machinery. Conversely, EBAG9 down-regulation accelerates glycoprotein transport through the Golgi and enhances mannosidase activity. Thus, EBAG9 acts as a negative regulator of a COPI-dependent ER-to-Golgi transport pathway in epithelial cells and represents a novel pathogenetic principle in which interference with intracellular membrane trafficking results in the emergence of a tumor-associated glycome.


Subject(s)
Antigens, Neoplasm/physiology , Coat Protein Complex I/physiology , Glycoproteins/metabolism , Blotting, Western , Cell Line, Tumor , Fluorescent Antibody Technique , Humans
20.
Cell Rep ; 37(4): 109878, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706240

ABSTRACT

Blood endothelial cells display remarkable plasticity depending on the demands of a malignant microenvironment. While studies in solid tumors focus on their role in metabolic adaptations, formation of high endothelial venules (HEVs) in lymph nodes extends their role to the organization of immune cell interactions. As a response to lymphoma growth, blood vessel density increases; however, the fate of HEVs remains elusive. Here, we report that lymphoma causes severe HEV regression in mouse models that phenocopies aggressive human B cell lymphomas. HEV dedifferentiation occurrs as a consequence of a disrupted lymph-carrying conduit system. Mechanosensitive fibroblastic reticular cells then deregulate CCL21 migration paths, followed by deterioration of dendritic cell proximity to HEVs. Loss of this crosstalk deprives HEVs of lymphotoxin-ß-receptor (LTßR) signaling, which is indispensable for their differentiation and lymphocyte transmigration. Collectively, this study reveals a remodeling cascade of the lymph node microenvironment that is detrimental for immune cell trafficking in lymphoma.


Subject(s)
Cell Movement , Endothelial Cells/metabolism , Lymphocytes/metabolism , Lymphoma, B-Cell/metabolism , Animals , Endothelial Cells/pathology , Humans , Jurkat Cells , Lymphocytes/pathology , Lymphoma, B-Cell/pathology , Mice , Mice, Transgenic , Venules
SELECTION OF CITATIONS
SEARCH DETAIL