Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Glia ; 70(12): 2361-2377, 2022 12.
Article in English | MEDLINE | ID: mdl-36054432

ABSTRACT

The plastic potential of Schwann cells (SCs) is increasingly recognized to play a role after nerve injury and in diseases of the peripheral nervous system. Reports on the interaction between immune cells and SCs indicate their involvement in inflammatory processes. However, the immunocompetence of human SCs has been primarily deduced from neuropathies, but whether after nerve injury SCs directly regulate an adaptive immune response is unknown. Here, we performed comprehensive analysis of immunomodulatory capacities of human repair-related SCs (hrSCs), which recapitulate SC response to nerve injury in vitro. We used our well-established culture model of primary hrSCs from human peripheral nerves and analyzed the transcriptome, secretome, and cell surface proteins for pathways and markers relevant in innate and adaptive immunity, performed phagocytosis assays, and monitored T-cell subset activation in allogeneic co-cultures. Our findings show that hrSCs are phagocytic, which is in line with high MHCII expression. Furthermore, hrSCs express co-regulatory proteins, such as CD40, CD80, B7H3, CD58, CD86, and HVEM, release a plethora of chemoattractants, matrix remodeling proteins and pro- as well as anti-inflammatory cytokines, and upregulate the T-cell inhibiting PD-L1 molecule upon pro-inflammatory stimulation with IFNγ. In contrast to monocytes, hrSC alone are not sufficient to trigger allogenic CD4+ and CD8+ T-cells, but limit number and activation status of exogenously activated T-cells. This study demonstrates that hrSCs possess features and functions typical for professional antigen-presenting cells in vitro, and suggest a new role of these cells as negative regulators of T-cell immunity during nerve regeneration.


Subject(s)
B7-H1 Antigen , CD8-Positive T-Lymphocytes , Antigen-Presenting Cells/metabolism , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Chemotactic Factors/metabolism , Cytokines/metabolism , Humans , Nerve Regeneration/physiology , Plastics/metabolism , Schwann Cells/metabolism
2.
EMBO Mol Med ; 14(8): e15230, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35781796

ABSTRACT

The recent emergence of multiple SARS-CoV-2 variants has caused considerable concern due to both reduced vaccine efficacy and escape from neutralizing antibody therapeutics. It is, therefore, paramount to develop therapeutic strategies that inhibit all known and future SARS-CoV-2 variants. Here, we report that all SARS-CoV-2 variants analyzed, including variants of concern (VOC) Alpha, Beta, Gamma, Delta, and Omicron, exhibit enhanced binding affinity to clinical grade and phase 2 tested recombinant human soluble ACE2 (APN01). Importantly, soluble ACE2 neutralized infection of VeroE6 cells and human lung epithelial cells by all current VOC strains with markedly enhanced potency when compared to reference SARS-CoV-2 isolates. Effective inhibition of infections with SARS-CoV-2 variants was validated and confirmed in two independent laboratories. These data show that SARS-CoV-2 variants that have emerged around the world, including current VOC and several variants of interest, can be inhibited by soluble ACE2, providing proof of principle of a pan-SARS-CoV-2 therapeutic.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 Drug Treatment , Humans , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2
3.
Cell Rep ; 38(8): 110420, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35196494

ABSTRACT

Dendritic cells (DCs) induce peripheral T cell tolerance, but cell-intrinsic signaling cascades governing their stable tolerogenesis remain poorly defined. Janus Kinase 1 (JAK1) transduces cytokine-receptor signaling, and JAK inhibitors (Jakinibs), including JAK1-specific filgotinib, break inflammatory cycles in autoimmunity. Here, we report in heterogeneous DC populations of multiple secondary lymphoid organs that JAK1 promotes peripheral T cell tolerance during experimental autoimmune encephalomyelitis (EAE). Mice harboring DC-specific JAK1 deletion exhibit elevated peripheral CD4+ T cell expansion, less regulatory T cells (Tregs), and worse EAE outcomes, whereas adoptive DC transfer ameliorates EAE pathogenesis by inducing peripheral Tregs, programmed cell death ligand 1 (PD-L1) dependently. This tolerogenic program is substantially reduced upon the transfer of JAK1-deficient DCs. DC-intrinsic IFN-γ-JAK1-STAT1 signaling induces PD-L1, which is required for DCs to convert CD4+ T cells into Tregs in vitro and attenuated upon JAK1 deficiency and filgotinib treatment. Thus, DC-intrinsic JAK1 promotes peripheral tolerance, suggesting potential unwarranted DC-mediated effects of Jakinibs in autoimmune diseases.


Subject(s)
B7-H1 Antigen , Encephalomyelitis, Autoimmune, Experimental , Janus Kinase 1 , T-Lymphocytes, Regulatory , Animals , Autoimmunity , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Dendritic Cells/metabolism , Immune Tolerance , Janus Kinase 1/immunology , Janus Kinase 1/metabolism , Mice , Peripheral Tolerance
4.
bioRxiv ; 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34545368

ABSTRACT

The recent emergence of multiple SARS-CoV-2 variants has caused considerable concern due to reduced vaccine efficacy and escape from neutralizing antibody therapeutics. It is therefore paramount to develop therapeutic strategies that inhibit all known and future SARS-CoV-2 variants. Here we report that all SARS-CoV-2 variants analyzed, including variants of concern (VOC) Alpha, Beta, Gamma, and Delta, exhibit enhanced binding affinity to clinical grade and phase 2 tested recombinant human soluble ACE2 (APN01). Importantly, soluble ACE2 neutralized infection of VeroE6 cells and human lung epithelial cells by multiple VOC strains with markedly enhanced potency when compared to reference SARS-CoV-2 isolates. Effective inhibition of infections with SARS-CoV-2 variants was validated and confirmed in two independent laboratories. These data show that SARS-CoV-2 variants that have emerged around the world, including current VOC and several variants of interest, can be inhibited by soluble ACE2, providing proof of principle of a pan-SARS-CoV-2 therapeutic.

5.
Front Immunol ; 11: 2189, 2020.
Article in English | MEDLINE | ID: mdl-33042133

ABSTRACT

Natural killer (NK) cells are important components of the innate immune defense against infections and cancers. Signal transducer and activator of transcription 1 (STAT1) is a transcription factor that is essential for NK cell maturation and NK cell-dependent tumor surveillance. Two alternatively spliced isoforms of STAT1 exist: a full-length STAT1α and a C-terminally truncated STAT1ß isoform. Aberrant splicing is frequently observed in cancer cells and several anti-cancer drugs interfere with the cellular splicing machinery. To investigate whether NK cell-mediated tumor surveillance is affected by a switch in STAT1 splicing, we made use of knock-in mice expressing either only the STAT1α (Stat1α/α) or the STAT1ß (Stat1ß/ß ) isoform. NK cells from Stat1α/α mice matured normally and controlled transplanted tumor cells as efficiently as NK cells from wild-type mice. In contrast, NK cells from Stat1ß/ß mice showed impaired maturation and effector functions, albeit less severe than NK cells from mice that completely lack STAT1 (Stat1-/- ). Mechanistically, we show that NK cell maturation requires the presence of STAT1α in the niche rather than in NK cells themselves and that NK cell maturation depends on IFNγ signaling under homeostatic conditions. The impaired NK cell maturation in Stat1ß/ß mice was paralleled by decreased IL-15 receptor alpha (IL-15Rα) surface levels on dendritic cells, macrophages and monocytes. Treatment of Stat1ß/ß mice with exogenous IL-15/IL-15Rα complexes rescued NK cell maturation but not their effector functions. Collectively, our findings provide evidence that STAT1 isoforms are not functionally redundant in regulating NK cell activity and that the absence of STAT1α severely impairs, but does not abolish, NK cell-dependent tumor surveillance.


Subject(s)
Killer Cells, Natural/cytology , Lymphopoiesis/physiology , STAT1 Transcription Factor/immunology , Animals , Bone Marrow Transplantation , Cell Line, Tumor , Cytotoxicity, Immunologic , Immunologic Surveillance/drug effects , Immunologic Surveillance/immunology , Interferon-Stimulated Gene Factor 3/deficiency , Interferon-Stimulated Gene Factor 3/genetics , Interferon-Stimulated Gene Factor 3/immunology , Interleukin-15/pharmacology , Interleukin-15 Receptor alpha Subunit , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Depletion , Lymphoid Tissue/cytology , Lymphoma/immunology , Lymphoma/pathology , Lymphopoiesis/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Interferon/deficiency , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , Specific Pathogen-Free Organisms , Spleen/cytology , Interferon gamma Receptor
6.
NPJ Vaccines ; 5(1): 5, 2020.
Article in English | MEDLINE | ID: mdl-31969991

ABSTRACT

Glioblastoma is the most prevalent and aggressive brain cancer. With a median overall survival of ~15-20 months under standard therapy, novel treatment approaches are desperately needed. A recent phase II clinical trial with a personalized immunotherapy based on tumor lysate-charged dendritic cell (DC) vaccination, however, failed to prolong survival. Here, we investigated tumor tissue from trial patients to explore glioblastoma survival-related factors. We followed an innovative approach of combining mass spectrometry-based quantitative proteomics (n = 36) with microRNA sequencing plus RT-qPCR (n = 38). Protein quantification identified, e.g., huntingtin interacting protein 1 (HIP1), retinol-binding protein 1 (RBP1), ferritin heavy chain (FTH1) and focal adhesion kinase 2 (FAK2) as factor candidates correlated with a dismal prognosis. MicroRNA analysis identified miR-216b, miR-216a, miR-708 and let-7i as molecules potentially associated with favorable tissue characteristics as they were enriched in patients with a comparably longer survival. To illustrate the utility of integrated miRNomics and proteomics findings, focal adhesion was studied further as one example for a pathway of potential general interest. Taken together, we here mapped possible drivers of glioblastoma outcome under immunotherapy in one of the largest DC vaccination tissue analysis cohorts so far-demonstrating usefulness and feasibility of combined proteomics/miRNomics approaches. Future research should investigate agents that sensitize glioblastoma to (immuno)therapy-potentially building on insights generated here.

7.
Cytotherapy ; 21(6): 643-658, 2019 06.
Article in English | MEDLINE | ID: mdl-30975602

ABSTRACT

BACKGROUND: Glioblastoma is the most aggressive type of brain cancer. Dendritic cell (DC)-based immunotherapy against glioblastoma depends on the effectiveness of loaded antigens. Sphere-inducing culture conditions are being studied by many as a potential antigen source. Here, we investigated two different in vitro conditions (spheroid culture versus adherent culture) in relation to DC immunotherapy: (1) We studied the specific spheroid-culture proteome and assessed the clinical importance of spheroid proteins. (2) We evaluated the immunogenicity of spheroid lysate - both compared to adherent conditions. METHODS: We used seven spheroid culture systems, three of them patient-derived. Stemness-related markers were studied in those three via immunofluorescence. Spheroid-specific protein expression was measured via quantitative proteomics. The Cancer Genome Atlas (TCGA) survival data was used to investigate the clinical impact of spheroid proteins. Immunogenicity of spheroid versus adherent cell lysate was explored in autologous ELISPOT systems (DCs and T cells from the three patients). RESULTS: (1) The differential proteome of spheroid versus adherent glioblastoma culture conditions could successfully be established. The top 10 identified spheroid-specific proteins were associated with significantly decreased overall survival (TCGA MIT/Harvard cohort; n = 350, P = 0.014). (2) In exploratory experiments, immunogenicity of spheroid lysate vis-á-vis interferon (IFN)γ production was lower than that of adherent cell lysate (IFNγ ELISPOT; P = 0.034). CONCLUSIONS: Spheroid culture proteins seem to represent survival-relevant targets, supporting the use of spheroid culture conditions as an antigen source for DC immunotherapy. However, immunogenicity enhancement should be considered for future research. Transferability of our findings in terms of clinical impact and regarding different spheroid-generation techniques needs further validation.


Subject(s)
Brain Neoplasms/immunology , Cell Culture Techniques/methods , Dendritic Cells/immunology , Glioblastoma/immunology , Neoplasm Proteins/immunology , Antigens, Neoplasm/immunology , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Glioblastoma/pathology , Humans , Immunotherapy/methods , Interferon-gamma/immunology , Interferon-gamma/metabolism , Neoplasm Proteins/metabolism , Spheroids, Cellular/pathology , T-Lymphocytes/immunology , Tumor Cells, Cultured
8.
J Cell Mol Med ; 23(1): 281-292, 2019 01.
Article in English | MEDLINE | ID: mdl-30467961

ABSTRACT

Glioblastoma is the most dangerous brain cancer. One reason for glioblastoma's aggressiveness are glioblastoma stem-like cells. To target them, a number of markers have been proposed (CD133, CD44, CD15, A2B5, CD36, CXCR4, IL6R, L1CAM, and ITGA6). A comprehensive study of co-expression patterns of them has, however, not been performed so far. Here, we mapped the multidimensional co-expression profile of these stemness-associated molecules. Gliomaspheres - an established model of glioblastoma stem-like cells - were used. Seven different gliomasphere systems were subjected to multicolor flow cytometry measuring the nine markers CD133, CD44, CD15, A2B5, CD36, CXCR4, IL6R, L1CAM, and ITGA6 all simultaneously based on a novel 9-marker multicolor panel developed for this study. The viSNE dimensionality reduction algorithm was applied for analysis. All gliomaspheres were found to express at least five different glioblastoma stem-like cell markers. Multi-dimensional analysis showed that all studied gliomaspheres consistently harbored a cell population positive for the molecular signature CD44+/CD133+/ITGA6+/CD36+. Glioblastoma patients with an enrichment of this combination had a significantly worse survival outcome when analyzing the two largest available The Cancer Genome Atlas datasets (MIT/Harvard Affymetrix: P = 0.0015, University of North Carolina Agilent: P = 0.0322). In sum, we detected a previously unknown marker combination - demonstrating feasibility, usefulness, and importance of high-dimensional gliomasphere marker combinatorics.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/pathology , Flow Cytometry/methods , Glioblastoma/pathology , AC133 Antigen/analysis , Algorithms , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , CD36 Antigens/analysis , Cell Adhesion/physiology , Cell Line, Tumor , Computer Simulation , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Hyaluronan Receptors/analysis , Integrin alpha6/analysis , Kaplan-Meier Estimate , Neoplastic Stem Cells/metabolism
9.
Acta Neuropathol Commun ; 6(1): 135, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30518425

ABSTRACT

Audencel is a dendritic cell (DC)-based cellular cancer immunotherapy against glioblastoma multiforme (GBM). It is characterized by loading of DCs with autologous whole tumor lysate and in vitro maturation via "danger signals". The recent phase II "GBM-Vax" trial showed no clinical efficacy for Audencel as assessed with progression-free and overall survival in all patients. Here we present immunological research accompanying the trial with a focus on immune system factors related to outcome and Audencel's effect on the immune system. Methodologically, peripheral blood samples (from apheresis before Audencel or venipuncture during Audencel) were subjected to functional characterization via enzyme-linked immunospot (ELISPOT) assays connected with cytokine bead assays (CBAs) as well as phenotypical characterization via flow cytometry and mRNA quantification. GBM tissue samples (from surgery) were subjected to T cell receptor sequencing and immunohistochemistry. As results we found: Patients with favorable pre-existing anti-tumor characteristics lived longer under Audencel than Audencel patients without them. Pre-vaccination blood CD8+ T cell count and ELISPOT Granzyme B production capacity in vitro upon tumor antigen exposure were significantly correlated with overall survival. Despite Audencel's general failure to induce a significant clinical response, it nevertheless seemed to have an effect on the immune system. For instance, Audencel led to a significant up-regulation of the Th1-related immunovariables ELISPOT IFNγ, the transcription factor T-bet in the blood and ELISPOT IL-2 in a dose-dependent manner upon vaccination. Post-vaccination levels of ELISPOT IFNγ and CD8+ cells in the blood were indicative of a significantly better survival. In summary, Audencel failed to reach an improvement of survival in the recent phase II clinical trial. No clinical efficacy was registered. Our concomitant immunological work presented here indicates that outcome under Audencel was influenced by the state of the immune system. On the other hand, Audencel also seemed to have stimulated the immune system. Overall, these immunological considerations suggest that DC immunotherapy against glioblastoma should be studied further - with the goal of translating an apparent immunological response into a clinical response. Future research should concentrate on investigating augmentation of immune reactions through combination therapies or on developing meaningful biomarkers.


Subject(s)
Brain Neoplasms/therapy , CD8-Positive T-Lymphocytes/physiology , Cancer Vaccines/therapeutic use , Dendritic Cells/physiology , Glioblastoma/therapy , Antigens, CD/metabolism , Boron Compounds/metabolism , Brain Neoplasms/blood , Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes/drug effects , Female , Glioblastoma/blood , Glioblastoma/immunology , Humans , Kaplan-Meier Estimate , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Longitudinal Studies , Male , Treatment Outcome , Up-Regulation
10.
Cell Immunol ; 325: 33-40, 2018 03.
Article in English | MEDLINE | ID: mdl-29402391

ABSTRACT

Gangliosides shed by tumors into their microenvironment (TME) are immunoinhibitory. Interferon-γ (IFN-γ) may boost antitumor immune responses. Thus we wondered whether IFN-γ would counteract tumor ganglioside-mediated immune suppression. To test this hypothesis, we exposed human monocyte-derived LPS-activated dendritic cells (DC) to IFN-γ and to a highly purified ganglioside, GD1a. DC ganglioside exposure decreased TLR-dependent p38 signaling, explaining the previously observed ganglioside-induced down-modulation of pro-inflammatory surface markers and cytokines. Strikingly, while increasing LPS-dependent DC responses, IFN-γ unexpectedly did not counteract the inhibitory effects of GD1a. Rather, induction of indoleamine 2,3-dioxygenase (IDO1), and expression of STAT1/IRF-1 and programmed cell death ligand (PD-L1), indicated that the immunoinhibitory, not an immune stimulatory, IFN-γ-signaling axis, was active. The combination, IFN-γ and DC ganglioside enrichment, markedly impaired DC stimulatory potential of CD8+ T-cells. We suggest that gangliosides and IFN-γ may act in concert as immunosuppressive mediators in the TME, possibly promoting tumor progression.


Subject(s)
Gangliosides/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Apoptosis/immunology , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/immunology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/immunology , Gangliosides/metabolism , Healthy Volunteers , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Neoplasms/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology
11.
Front Immunol ; 9: 3108, 2018.
Article in English | MEDLINE | ID: mdl-30671064

ABSTRACT

The Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling pathway is critical in tuning immune responses and its dysregulation is tightly associated with cancer and immune disorders. Disruption of interleukin (IL)-15/STAT5 signaling pathway due to the loss of IL-15 receptor chains, JAK3 or STAT5 leads to immune deficiencies with natural killer (NK) cell abnormalities. JAK1, together with JAK3 transmits signals downstream of IL-15, but the exact contribution of JAK1 to NK cell biology remains to be elucidated. To study the consequences of JAK1 deficiency in NK cells, we generated mice with conditional deletion of JAK1 in NKp46+ cells (Jak1fl/flNcr1Cre). We show here that deletion of NK cell-intrinsic JAK1 significantly reduced NK cell numbers in the bone marrow and impaired their development. In line, we observed almost a complete loss of NK cells in the spleen, blood, and liver, proving a crucial role of JAK1 in peripheral NK cells. In line, Jak1fl/+Ncr1Cre mice showed significantly impaired NK cell-mediated tumor surveillance. Our data suggest that JAK2 is not able to compensate for the loss of JAK1 in NK cells. Importantly, conditional deletion of JAK2 in NKp46+ cells had no effect on peripheral NK cells revealing that NK cell-intrinsic JAK2 is dispensable for NK cell survival. In summary, we identified that loss of JAK1 in NK cells drives innate immune deficiency, whereas JAK2 deficiency leaves NK cell numbers and maturation unaltered. We thus propose that in contrast to currently used JAK1/JAK2 inhibitors, the use of JAK2-specific inhibitors would be advantageous for the patients by leaving NK cells intact.


Subject(s)
Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Killer Cells, Natural/metabolism , Lymphoma/enzymology , Alleles , Analysis of Variance , Animals , CD11b Antigen/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Cell Survival/physiology , Disease Models, Animal , Immunity, Innate , Janus Kinase 1/genetics , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphoma/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Tumor Burden , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
12.
Front Immunol ; 8: 1152, 2017.
Article in English | MEDLINE | ID: mdl-28979262

ABSTRACT

Donor T-cells contribute to reconstitution of protective immunity after allogeneic hematopoietic stem cell transplantation (HSCT) but must acquire specific tolerance against recipient alloantigens to avoid life-threatening graft-versus-host disease (GvHD). Systemic immunosuppressive drugs may abrogate severe GvHD, but this also impedes memory responses to invading pathogens. Here, we tested whether ex vivo blockade of CD28 co-stimulation can enable selective T-cell tolerization to alloantigens by facilitating CD80/86-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling. Treatment of human allogeneic dendritic cell/T-cell co-cultures with a human CD28 blocking antibody fragment (α-huCD28) significantly abrogated subsequent allospecific immune responses, seen by decreased T-cell proliferation and of type 1 cytokine (IFN-γ and IL-2) expression. Allo-tolerization persisted after discontinuation of CD28 blockade and secondary alloantigen stimulation, as confirmed by enhanced CTLA-4 and PD-1 immune checkpoint signaling. However, T-cells retained reactivity to pathogens, supported by clonotyping of neo-primed and cross-reactive T-cells specific for Candida albicans or third-party antigens using deep sequencing analysis. In an MHC-mismatched murine model, we tolerized C57BL/6 T-cells by ex vivo exposure to a murine single chain Fv specific for CD28 (α-muCD28). Infusion of these cells, after α-muCD28 washout, into bone marrow-transplanted BALB/c mice caused allo-tolerance and did not induce GvHD-associated hepatic pathology. We conclude that selective CD28 blockade ex vivo can allow the generation of stably allo-tolerized T-cells that in turn do not induce graft-versus-host reactions while maintaining pathogen reactivity. Hence, CD28 co-stimulation blockade of donor T-cells may be a useful therapeutic approach to support the immune system after HSCT.

13.
Sci Rep ; 7(1): 11746, 2017 09 18.
Article in English | MEDLINE | ID: mdl-28924177

ABSTRACT

Maintaining dendritic cells (DC) in a state of dysfunction represents a key mechanism by which tumour cells evade recognition and elimination by the immune system. Limited knowledge about the intracellular mediators of DC dysfunction restricts success of therapies aimed at reactivating a DC-driven anti-tumour immune response. Using a cell type-specific murine knock-out model, we have identified MAPK-activated protein kinase 2 (MK2) as a major guardian of a suppressive DC phenotype in the melanoma tumour microenvironment. MK2 deletion in CD11c+ cells led to an expansion of stimulatory CD103+ DCs, mounting a potent CD8+ T cell response that resulted in elimination of highly aggressive B16-F10 tumours upon toll-like receptor (TLR) activation in the presence of tumour antigen. Moreover, tumour infiltration by suppressive myeloid cells was strongly diminished. These insights into the regulation of DC functionality reveal MK2 as a targetable pathway for DC-centred immunomodulatory cancer therapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunity, Cellular , Intracellular Signaling Peptides and Proteins/deficiency , Melanoma, Experimental/immunology , Protein Serine-Threonine Kinases/deficiency , Tumor Microenvironment , Animals , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Dendritic Cells/enzymology , Dendritic Cells/pathology , Intracellular Signaling Peptides and Proteins/immunology , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
14.
Chemistry ; 23(70): 17721-17726, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-28758266

ABSTRACT

The chemical synthesis and biological activity of novel functionalized imidazoquinoline derivatives (ImQ) to generate Toll-like receptor (TLR) 7/8 specific prodrugs are presented. In vivo activity of ImQs to induce inflammation was confirmed in zebrafish larvae. After covalent ligation to fully biodegradable polyphosphazenes (ImQ-polymer), the macromolecular prodrugs were designed to undergo intracellular pH-sensitive release of ImQs to induce inflammation through binding to endosomal TLR7/8 (danger signal). We showed ImQ dissociation from prodrugs at a pH 5 pointing towards endosomal prodrug degradability. ImQ-polymers strongly activated ovalbumin-specific T cells in murine splenocytes as shown by increased proliferation and expression of the IL-2 receptor (CD25) on CD8+ T cells accompanied by strong IFN-γ release. ImQ prodrugs presented here are suggested to form the basis of novel nanovaccines, for example, for intravenous or intratumoral cancer immunotherapeutic applications to trigger physiological antitumor immune responses.


Subject(s)
Prodrugs/chemistry , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 8/antagonists & inhibitors , Animals , Animals, Genetically Modified/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Hydrogen-Ion Concentration , Inflammation/etiology , Interferon-gamma/metabolism , Larva/drug effects , Larva/metabolism , Mice , Microscopy, Confocal , NF-kappa B/metabolism , Prodrugs/chemical synthesis , Prodrugs/toxicity , Quinolines/chemical synthesis , Quinolines/chemistry , Quinolines/toxicity , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Zebrafish/growth & development
15.
PLoS One ; 11(11): e0163661, 2016.
Article in English | MEDLINE | ID: mdl-27893746

ABSTRACT

BACKGROUND: Mast cells (MC) are bone marrow derived haematopoetic cells playing a crucial role not only in immune response but also in the tumor microenvironment with protumorigenic and antitumorigenic functions. The role of MC in primary cutaneous T-cell lymphomas (CTCL), a heterogeneous group of non-Hodgkin lymphomas with initial presentation in the skin, is largely unknown. OBJECTIVE: To gain more accurate information about presence, number, distribution and state of activation (degranulated vs. non-degranulated) of MC in CTCL variants and clinical stages. MATERIALS AND METHODS: We established a novel computer-aided tissue analysis method on digitized skin sections. Immunohistochemistry with an anti-MC tryptase antibody was performed on 34 biopsies of different CTCL subtypes and on control skin samples. An algorithm for the automatic detection of the epidermis and of cell density based CTCL areas was developed. Cells were stratified as being within the CTCL infiltrate, in P1 (a surrounding area 0-30 µm away from CTCL), or in P2 (30-60 µm away from CTCL) area. RESULTS: We found high MC counts within CTCL infiltrates and P1 and a decreased MC number in the surrounding dermis P2. Higher MC numbers were found in MF compared to all other CTCL subgroups. Regarding different stages of MF, we found significantly higher mast cell counts in stages IA and IB than in stages IIA and IIB. Regarding MC densities, we found a higher density of MC in MF compared to all other CTCL subgroups. More MC were non-degranulated than degranulated. CONCLUSION: Here for the first time an automated method for MC analysis on tissue sections and its use in CTCL is described. Eliminating error from investigator bias, the method allows for precise cell identification and counting. Our results provide new insights on MC distribution in CTCL reappraising their role in the pathophysiology of CTCL.


Subject(s)
Lymphoma, T-Cell, Cutaneous/pathology , Mast Cells/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Degranulation , Female , Humans , Image Processing, Computer-Assisted/methods , Immunohistochemistry/methods , Male , Mast Cells/physiology , Middle Aged , Mycosis Fungoides/pathology , Skin Neoplasms/pathology , Young Adult
16.
J Immunol ; 195(6): 2560-70, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26246144

ABSTRACT

The PI3K signaling cascade in APCs has been recognized as an essential pathway to initiate, maintain, and resolve immune responses. In this study, we demonstrate that a cell type-specific loss of the PI3K antagonist phosphatase and tensin homolog (PTEN) in myeloid cells renders APCs toward a regulatory phenotype. APCs deficient for PTEN exhibit reduced activation of p38 MAPK and reduced expression of T cell-polarizing cytokines. Furthermore, PTEN deficiency leads to upregulation of markers for alternative activation, such as Arginase 1, with concomitant downregulation of inducible NO synthase in APCs in vitro and in vivo. As a result, T cell polarization was dysfunctional in PTEN(-/-) APCs, in particular affecting the Th17 cell subset. Intriguingly, mice with cell type-specific deletions of PTEN-targeting APCs were protected from experimental autoimmune encephalomyelitis, which was accompanied by a pronounced reduction of IL-17- and IL-22-producing autoreactive T cells and reduced CNS influx of classically activated monocytes/macrophages. These observations support the notion that activation of the PI3K signaling cascade promotes regulatory APC properties and suppresses pathogenic T cell polarization, thereby reducing the clinical symptoms and pathology of experimental autoimmune encephalomyelitis.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , PTEN Phosphohydrolase/genetics , Th17 Cells/immunology , Animals , Arginase/biosynthesis , Autoimmunity/immunology , CD11c Antigen/biosynthesis , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Enzyme Activation/genetics , Enzyme Activation/immunology , Interleukin-17/biosynthesis , Interleukins/biosynthesis , Lymphocyte Activation , Macrophage Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology , Nitric Oxide Synthase Type II/biosynthesis , Peptide Fragments/immunology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Interleukin-22
17.
J Immunol ; 195(2): 541-52, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26078274

ABSTRACT

Dendritic cell (DC)-mediated inflammation induced via TLRs is promoted by MAPK-activated protein kinase (MK)-2, a substrate of p38 MAPK. In this study we show an opposing role of MK2, by which it consolidates immune regulatory functions in DCs through modulation of p38, ERK1/2-MAPK, and STAT3 signaling. During primary TLR/p38 signaling, MK2 mediates the inhibition of p38 activation and positively cross-regulates ERK1/2 activity, leading to a reduction of IL-12 and IL-1α/ß secretion. Consequently, MK2 impairs secondary autocrine IL-1α signaling in DCs, which further decreases the IL-1α/p38 but increases the anti-inflammatory IL-10/STAT3 signaling route. Therefore, the blockade of MK2 activity enables human and murine DCs to strengthen proinflammatory effector mechanisms by promoting IL-1α-mediated Th1 effector functions in vitro. Furthermore, MK2-deficient DCs trigger Th1 differentiation and Ag-specific cytotoxicity in vivo. Finally, wild-type mice immunized with LPS in the presence of an MK2 inhibitor strongly accumulate Th1 cells in their lymph nodes. These observations correlate with a severe clinical course in DC-specific MK2 knockout mice compared with wild-type littermates upon induction of experimental autoimmune encephalitis. Our data suggest that MK2 exerts a profound anti-inflammatory effect that prevents DCs from prolonging excessive Th1 effector T cell functions and autoimmunity.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Intracellular Signaling Peptides and Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Th1 Cells/immunology , Animals , Cell Differentiation , Dendritic Cells/drug effects , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Humans , Immunization , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction , Th1 Cells/drug effects , Th1 Cells/pathology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
18.
J Immunol ; 193(4): 1717-27, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015834

ABSTRACT

The activation of innate immune cells triggers numerous intracellular signaling pathways, which require tight control to mount an adequate immune response. The PI3K signaling pathway is intricately involved in innate immunity, and its activation dampens the expression and release of proinflammatory cytokines in myeloid cells. These signaling processes are strictly regulated by the PI3K antagonist, the lipid phosphatase, PTEN, a known tumor suppressor. Importantly, PTEN is responsible for the elevated production of cytokines such as IL-6 in response to TLR agonists, and deletion of PTEN results in diminished inflammatory responses. However, the mechanisms by which PI3K negatively regulates TLR signaling are only partially resolved. We observed that Arginase I expression and secretion were markedly induced by PTEN deletion, suggesting PTEN(-/-) macrophages were alternatively activated. This was mediated by increased expression and activation of the transcription factors C/EBPß and STAT3. Genetic and pharmacologic experimental approaches in vitro, as well as in vivo autoimmunity models, provide convincing evidence that PI3K/PTEN-regulated extracellular Arginase I acts as a paracrine regulator of inflammation and immunity.


Subject(s)
Arginase/metabolism , Macrophages/immunology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/immunology , Adaptive Immunity , Animals , Arginase/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/immunology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Genotype , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , HEK293 Cells , Humans , Immunity, Innate , Inflammation/genetics , Interleukin-10/biosynthesis , Interleukin-10/metabolism , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/enzymology , Myeloid Cells/immunology , Phosphoinositide-3 Kinase Inhibitors , RNA, Messenger/biosynthesis , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Toll-Like Receptors/agonists , Toll-Like Receptors/immunology , Tumor Necrosis Factor-alpha/biosynthesis
19.
PLoS One ; 8(2): e54879, 2013.
Article in English | MEDLINE | ID: mdl-23408948

ABSTRACT

The dendritic cell (DC) coordinates innate and adaptive immunity to fight infections and cancer. Our observations reveal that DCs exposed to the microbial danger signal lipopolysaccharide (LPS) in the presence of interferon-γ (IFN-γ) acquire a continuously changing activation/maturation phenotype. The DCs' initial mode of action is pro-inflammatory via up-regulation among others of the signaling molecule interleukin (IL) 12, which polarizes IFN-γ secreting type 1 helper T-cells (Th1). Within 24 hours the same DC switches from the pro- into an anti-inflammatory phenotype. This is mediated by autocrine IL-10 release and secretion of soluble IL-2 receptor alpha (sIL-2RA) molecules. T-cells, when contacted with DCs during their anti-inflammatory phase loose their proliferative capacity and develop regulatory T-cell (Treg) -like anti-inflammatory functions indicated by IL-10 secretion and elevated FoxP3 levels. Studying the kinetics of IL-12 and IL-10 expression from LPS/IFN-γ activated myeloid DCs on a single cell level confirmed these observations. When T-cells are separated from DCs within 24 hours, they are spared from the anti-inflammatory DC activity. We conclude that, in addition to differentiation of DCs into distinct subsets, the observed sequential functional phases of DC differentiation permit the fine-tuning of an immune response. A better understanding of time-kinetic DC features is required for optimally exploiting the therapeutic capacity of DCs in cancer immune therapy.


Subject(s)
Cell Differentiation/physiology , Dendritic Cells/cytology , Inflammation/physiopathology , Toll-Like Receptor 4/physiology , Animals , Cytokines/metabolism , Dendritic Cells/metabolism , Humans , Mice
20.
J Immunother ; 33(1): 40-52, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19952957

ABSTRACT

In cancer patients undergoing immune therapy with lipopolysaccharides/interferon-gamma activated interleukin (IL)-12 secreting dendritic cells (DCs) we observed enhanced proliferative capacity of pyrophosphate-responsive peripheral blood (PB) gammadelta T-cells. This was not noted before as in other clinical trials DCs were used that were not enabled for IL-12 secretion and mice do not have a corresponding subset of PB gammadelta T-cells. In vitro examination of IL-12 DC/PB gammadelta T-cell interactions revealed a potential of PB gammadelta T-cells to negatively regulate the proliferative capacity of CD4 and CD8 T-cells. We further demonstrate that IL-12 is critical in the activation of PB gammadelta T-cells. In contrast, the regulatory activity of PB gammadelta T-cells in immune responses against strong recall antigens or alloantigens did not require activated DCs, but depended on pyrophosphate activation of PB gammadelta T-cells. Depletion of PB gammadelta T-cells abrogated the regulatory activity in IL-12 DC/peripheral blood mononuclear cell cocultures; adding back graded numbers of PB gammadelta T-cells restored it. Our observations revealed a potential PB gammadelta T cell-mediated negative regulatory feedback mechanism triggered by IL-12 DCs, which may critically impact on the design of DC cancer vaccines.


Subject(s)
Dendritic Cells/transplantation , Interleukin-12/immunology , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Cancer Vaccines/immunology , Clinical Trials, Phase I as Topic , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Interleukin-12/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...