Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
Blood ; 142(9): 794-805, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37217183

ABSTRACT

Targeted therapies for cutaneous T-cell lymphoma (CTCL) are limited and curative approaches are lacking. Furthermore, relapses and drug induced side effects are major challenges in the therapeutic management of patients with CTCL, creating an urgent need for new and effective therapies. Pathologic constitutive NF-κB activity leads to apoptosis resistance in CTCL cells and, thus, represents a promising therapeutic target in CTCL. In a preclinical study we showed the potential of dimethyl fumarate (DMF) to block NF-κB and, specifically, kill CTCL cells. To translate these findings to applications in a clinical setting, we performed a multicentric phase 2 study evaluating oral DMF therapy in 25 patients with CTCL stages Ib to IV over 24 weeks (EudraCT number 2014-000924-11/NCT number NCT02546440). End points were safety and efficacy. We evaluated skin involvement (using a modified severity weighted assessment tool [mSWAT]), pruritus, quality of life, and blood involvement, if applicable, as well as translational data. Upon skin analysis, 7 of 23 (30.4%) patients showed a response with >50% reduction in the mSWAT score. Patients with high tumor burden in the skin and blood responded best to DMF therapy. Although not generally significant, DMF also improved pruritus in several patients. Response in the blood was mixed, but we confirmed the NF-κB-inhibiting mechanism of DMF in the blood. The overall tolerability of the DMF therapy was very favorable, with mostly mild side effects. In conclusion, our study presents DMF as an effective and excellently tolerable therapeutic option in CTCL to be further evaluated in a phase 3 study or real-life patient care as well as in combination therapies. This trial was registered at www.clinicaltrials.gov as #NCT02546440.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Skin Neoplasms , Humans , Dimethyl Fumarate/therapeutic use , NF-kappa B , Quality of Life , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Neoplasm Recurrence, Local/drug therapy , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/pathology , Pruritus/drug therapy
2.
BMC Cancer ; 22(1): 994, 2022 Sep 19.
Article in English | MEDLINE | ID: mdl-36123610

ABSTRACT

BACKGROUND: Uptake of apoptotic cells induces a tolerogenic phenotype in phagocytes and promotes peripheral tolerance. The highly conserved Annexin core domain, present in all members of the Annexin family, becomes exposed on the apoptotic cell-surface and triggers tolerogenic signalling in phagocytes via the Dectin-1 receptor. Consequently, Annexins exposed on tumour cells upon cell death are expected to induce tolerance towards tumour antigens, inhibiting tumour rejection. METHODS: Expression analysis for all Annexin family members was conducted in cancer cell lines of diverse origins. Presentation of Annexins on the cell surface during apoptosis of cancer cell lines was investigated using surface washes and immunoblotting. Expression data from the GEO database was analysed to compare Annexin levels between malignant and healthy tissue. RESULTS: Six Annexins at least were consistently detected on mRNA and protein level for each investigated cell line. AnxA1, AnxA2 and AnxA5 constituted the major part of total Annexin expression. All expressed Annexins translocated to the cell surface upon apoptosis induction in all cell lines. Human expression data indicate a correlation between immune infiltration and overall Annexin expression in malignant compared to healthy tissue. CONCLUSIONS: This study is the first comprehensive analysis of expression, distribution and presentation of Annexins in cancer.


Subject(s)
Annexins , Neoplasms , Annexin A5 , Annexins/genetics , Annexins/metabolism , Antigens, Neoplasm , Humans , Neoplasms/genetics , RNA, Messenger
4.
Cancer Med ; 10(19): 6807-6822, 2021 10.
Article in English | MEDLINE | ID: mdl-34546000

ABSTRACT

Rocaglates are natural compounds that have been extensively studied for their ability to inhibit translation initiation. Rocaglates represent promising drug candidates for tumor treatment due to their growth-inhibitory effects on neoplastic cells. In contrast to natural rocaglates, synthetic analogues of rocaglates have been less comprehensively characterized, but were also shown to have similar effects on the process of protein translation. Here, we demonstrate an enhanced growth-inhibitory effect of synthetic rocaglates when combined with glucose anti-metabolite 2-deoxy-D-glucose (2DG) in different cancer cell lines. Moreover, we unravel a new aspect in the mechanism of action of synthetic rocaglates involving reduction of glucose uptake mediated by downregulation or abrogation of glucose transporter GLUT-1 expression. Importantly, cells with genetically induced resistance to synthetic rocaglates showed substantially less pronounced treatment effect on glucose metabolism and did not demonstrate GLUT-1 downregulation, pointing at the crucial role of this mechanism for the anti-tumor activity of the synthetic rocaglates. Transcriptome profiling revealed glycolysis as one of the major pathways differentially regulated in sensitive and resistant cells. Analysis of synthetic rocaglate efficacy in a 3D tissue context with a co-culture of tumor and normal cells demonstrated a selective effect on tumor cells and substantiated the mechanistic observations obtained in cancer cell lines. Increased glucose uptake and metabolism is a universal feature across different tumor types. Therefore, targeting this feature by synthetic rocaglates could represent a promising direction for exploitation of rocaglates in novel anti-tumor therapies.


Subject(s)
Benzofurans/therapeutic use , Glucose Transporter Type 1/metabolism , Glucose/metabolism , Neoplasms/drug therapy , Benzofurans/pharmacology , Cell Proliferation , Humans
5.
Autoimmunity ; 53(2): 86-94, 2020 03.
Article in English | MEDLINE | ID: mdl-31933381

ABSTRACT

Apoptotic cells mediate the development of tolerogenic dendritic cells (DC) and thus facilitate induction and maintenance of peripheral tolerance. Following the identification of the evolutionary conserved annexin core domain (Anx) as a specific signal on apoptotic cells which antagonises Toll-like receptor (TLR) signalling, we examined whether the tolerogenic capacity of Anx can be exploited to downregulate antigen-specific immune responses. The treatment of bone marrow-derived dendritic cells (BMDC) with particles harbouring Anx as well as the model antigen ovalbumin (OVA) attenuated the response of OVA-specific OT-II T cells. The co-culture of Anx-particle-treated DC and T cells resulted in an anergy-like phenotype characterized by reduced proliferation and cytokine secretion. Here we demonstrate that the anti-inflammatory effects of Anx which are mediated through DC can be used as a tool to generate a particle-based antigen delivery system that promotes antigen-specific immunosuppression. Such Anx-particles may be a new therapeutic approach for the treatment of autoimmune diseases.


Subject(s)
Annexins/pharmacology , Antigens/administration & dosage , Dendritic Cells/drug effects , Drug Delivery Systems/methods , Immune Tolerance/drug effects , Animals , Annexins/immunology , Apoptosis/drug effects , Apoptosis/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Humans , Mice , Microspheres , Ovalbumin/immunology , Primary Cell Culture/methods , Protein Domains/immunology , T-Lymphocytes/immunology
6.
Bio Protoc ; 10(17): e3737, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-33659398

ABSTRACT

Depending on its concentration and cellular origin the production of reactive oxygen species (ROS) in the organism serves a variety of functions. While high concentrations during an oxidative burst are used to fight pathogens, low to moderate amounts of ROS act as signaling molecules important for several physiological processes such as regulation of immune responses. The ROS-sensitive dye 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) is an inexpensive and well-established tool for measuring intracellular ROS levels. However, it needs to be carefully controlled to be able to draw firm conclusions on the nature of ROS species produced and the cellular source of ROS generation such as the enzyme complex NADPH-oxidase 2 (NOX-2). In this protocol, a robust method to determine low intracellular ROS production using H2DCFDA was validated by several ROS-specific as well as NOX-2-specific inhibitors. Cells were treated with inhibitors or control substances prior to treatment with the ROS-inducer of interest. H2DCFDA was added only for the last 30 min of the treatment schedule. To terminate its conversion, we used a ROS-specific inhibitor until analysis by flow cytometry within the FITC-channel (Ex: 488 nm/Em: 519 nm). In summary, this protocol allows the detection of signaling-relevant intracellular ROS production in cell lines and primary immune cells (e.g., Mono Mac 6 cells and Bone marrow-derived dendritic cells, respectively). Using this method in combination with specific inhibitors, we were able to validate even exceptionally low amounts of ROS produced by NOX-2 and relevant for immune-regulatory signaling.

7.
Cell Rep ; 29(13): 4435-4446.e9, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875551

ABSTRACT

Uptake of apoptotic cells (ACs) by dendritic cells (DCs) and induction of a tolerogenic DC phenotype is an important mechanism for establishing peripheral tolerance to self-antigens. The receptors involved and underlying signaling pathways are not fully understood. Here, we identify Dectin-1 as a crucial tolerogenic receptor binding with nanomolar affinity to the core domain of several annexins (annexin A1, A5, and A13) exposed on ACs. Annexins bind to Dectin-1 on a site distinct from the interaction site of pathogen-derived ß-glucans. Subsequent tolerogenic signaling induces selective phosphorylation of spleen tyrosine kinase (SYK), causing activation of NADPH oxidase-2 and moderate production of reactive oxygen species. Thus, mice deficient for Dectin-1 develop autoimmune pathologies (autoantibodies and splenomegaly) and generate stronger immune responses (cytotoxic T cells) against ACs. Our data describe an important immunological checkpoint system and provide a link between immunosuppressive signals of ACs and maintenance of peripheral immune tolerance.


Subject(s)
Annexins/metabolism , Apoptosis , Lectins, C-Type/metabolism , NADPH Oxidase 2/metabolism , Peripheral Tolerance , Aging/metabolism , Animals , Annexins/chemistry , Antigens/metabolism , Autoimmunity , Binding Sites , Conserved Sequence/genetics , Drosophila , Female , Humans , Immunosuppression Therapy , Jurkat Cells , Male , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , Protein Binding , Protein Domains , Reactive Oxygen Species/metabolism , Syk Kinase/metabolism , beta-Glucans/metabolism
8.
Blood ; 134(5): 445-455, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31167801

ABSTRACT

Therapeutic options for cutaneous T-cell lymphoma (CTCL) are limited and curative treatment regimens are not available. Thus, new targeted and well-tolerated therapeutic approaches are urgently needed. In this respect, we have recently shown that dimethyl fumerate (DMF) inhibits NF-κB acting as a survival factor in CTCL. Similarly, inhibition of the antiapoptotic protein B-cell lymphoma 2 (Bcl-2) has been shown to induce cell death in CTCL especially when combined with histone deacetylase inhibitors. Therefore, we hypothesized that inhibition of Bcl-2 should potentiate NF-κB inhibition in a novel combination treatment of CTCL. We show that, in vitro, the Bcl-2 inhibitors ABT-199 and ABT-263 induced specific cell death in primary CD4+ cells from CTCL patients as well as in the CTCL cell line SeAx, but not in T cells of healthy donors nor in the CTCL cell line HH, which lacks Bcl-2. Combined treatment with ABT-199 and DMF caused synergistic cell death specifically in CTCL cells engaging 2 independent signaling pathways. To verify these findings in vivo, we performed combined ABT-199 and DMF treatment in a xenograft mouse model for CTCL. The combined treatment effectively reduced tumor growth and increased overall survival via synergistic induction of CTCL cell death and suppression of tumor cell proliferation. Essentially, the combination treatment was superior to ABT-199 monotherapy with respect to both efficacy and tolerability. To sum up, our data provide proof of principle for the therapeutic potential of combining Bcl-2 and NF-κB inhibitors in treating CTCL. Next, this potential should be explored further in a clinical study.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Lymphoma, T-Cell, Cutaneous/metabolism , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Apoptosis , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Humans , Lymphoma, T-Cell, Cutaneous/diagnosis , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/genetics , Mice , NF-kappa B/genetics , Neoplasm Staging , Protein Binding , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
9.
FEBS Lett ; 592(13): 2297-2307, 2018 07.
Article in English | MEDLINE | ID: mdl-29897613

ABSTRACT

The "free radical theory of aging" suggests that reactive oxygen species (ROS) are responsible for age-related loss of cellular functions and, therefore, represent the main cause of aging. Redox regulation by thioredoxin-1 (TRX) plays a crucial role in responses to oxidative stress. We show that thioredoxin-interacting protein (TXNIP), a negative regulator of TRX, plays a major role in maintaining the redox status and, thereby, influences aging processes. This role of TXNIP is conserved from flies to humans. Age-dependent upregulation of TXNIP results in decreased stress resistance to oxidative challenge in primary human cells and in Drosophila. Experimental overexpression of TXNIP in flies shortens lifespan due to elevated oxidative DNA damage, whereas downregulation of TXNIP enhances oxidative stress resistance and extends lifespan.


Subject(s)
Aging/genetics , Carrier Proteins/physiology , Cell Cycle Proteins/physiology , DNA Damage/genetics , Oxidative Stress/genetics , Adult , Aged , Aging/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cells, Cultured , Drosophila melanogaster , HEK293 Cells , Humans , Jurkat Cells , Longevity/genetics , Middle Aged , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism , Up-Regulation/genetics , Young Adult
10.
Haematologica ; 103(4): 688-697, 2018 04.
Article in English | MEDLINE | ID: mdl-29326123

ABSTRACT

Chronic lymphocytic leukemia is a malignancy of mature B cells that strongly depend on microenvironmental factors, and their deprivation has been identified as a promising treatment approach for this incurable disease. Cytokine array screening of 247 chronic lymphocytic leukemia serum samples revealed elevated levels of tumor necrosis factor (TNF) receptor-1 which were associated with poor clinical outcome. We detected a microenvironment-induced expression of TNF receptor-1 in chronic lymphocytic leukemia cells in vitro, and an aberrantly high expression of this receptor in the proliferation centers of patients' lymph nodes. Stimulation of TNF receptor-1 with TNF-α enhanced nuclear factor κ-light-chain-enhancer of activated B cells (NFκB) activity and viability of chronic lymphocytic leukemia cells, which was inhibited by wogonin. The therapeutic effects of wogonin were analyzed in mice after adoptive transfer of Eµ-T-cell leukemia 1 (TCL1) leukemic cells. Wogonin treatment prevented leukemia development when given early after transplantation. The treatment of full-blown leukemia resulted in the loss of the TNF receptor-1 on chronic lymphocytic leukemia cells and their mobilization to blood. Targeting TNF receptor-1 signaling is therefore proposed for the treatment of chronic lymphocytic leukemia.


Subject(s)
Flavanones/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Adoptive Transfer , Animals , Coculture Techniques , Humans , Leukemia/pathology , Leukemia/prevention & control , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymph Nodes/metabolism , Mice , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/drug effects
11.
Cell Death Dis ; 9(2): 132, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29374146

ABSTRACT

Ischemia-reperfusion injury is a common pathological process in liver surgery and transplantation, and has considerable impact on the patient outcome and survival. Death receptors are important mediators of ischemia-reperfusion injury, notably the signaling pathways of the death receptor CD95 (Apo-1/Fas) and its corresponding ligand CD95L. This study investigates, for the first time, whether the inhibition of CD95L protects the liver against ischemia-reperfusion injury. Warm ischemia was induced in the median and left liver lobes of C57BL/6 mice for 45 min. CD95Fc, a specific inhibitor of CD95L, was applied prior to ischemia. Hepatic injury was assessed via consecutive measurements of liver serum enzymes, histopathological assessment of apoptosis and necrosis and caspase assays at 3, 6, 12, 18 and 24 h after reperfusion. Serum levels of liver enzymes, as well as characteristic histopathological changes and caspase assays indicated pronounced features of apoptotic and necrotic liver damage 12 and 24 h after ischemia-reperfusion injury. Animals treated with the CD95L-blocker CD95Fc, exhibited a significant reduction in the level of serum liver enzymes and showed both decreased histopathological signs of parenchymal damage and decreased caspase activation. This study demonstrates that inhibition of CD95L with the CD95L-blocker CD95Fc, is effective in protecting mice from liver failure due to ischemia-reperfusion injury of the liver. CD95Fc could therefore emerge as a new pharmacological therapy for liver resection, transplantation surgery and acute liver failure.


Subject(s)
Fas Ligand Protein/metabolism , Liver Failure, Acute/complications , Liver Failure, Acute/prevention & control , Liver/pathology , Reperfusion Injury/complications , Reperfusion Injury/prevention & control , Animals , Caspases/metabolism , Hepatocytes/pathology , Liver/enzymology , Male , Mice, Inbred C57BL , Necrosis , Reperfusion Injury/blood , Reperfusion Injury/enzymology
12.
Mol Cell Proteomics ; 17(5): 993-1009, 2018 05.
Article in English | MEDLINE | ID: mdl-29217617

ABSTRACT

Coimmunoprecipitation (co-IP) is one of the most frequently used techniques to study protein-protein (PPIs) or protein-nucleic acid interactions (PNIs). However, the presence of coprecipitated contaminants is a well-recognized issue associated with single-step co-IPs. To overcome this limitation, we developed the two-step co-IP (TIP) strategy that enables sequential coimmunoprecipitations of endogenous protein complexes. TIP can be performed with a broad range of mono- and polyclonal antibodies targeting a single protein or different components of a given complex. TIP results in a highly selective enrichment of protein complexes and thus outperforms single-step co-IPs for downstream applications such as mass spectrometry for the identification of PPIs and quantitative PCR for the analysis of PNIs. We benchmarked TIP for the identification of CD95/FAS-interacting proteins in primary human CD4+ T cells, which recapitulated all major known interactors, but also enabled the proteomics discovery of PPM1G and IPO7 as new interaction partners. For its feasibility and high performance, we propose TIP as an advanced tool for the isolation of highly purified protein-protein and protein-nucleic acid complexes under native expression conditions.


Subject(s)
Immunoprecipitation/methods , Multiprotein Complexes/isolation & purification , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Apoptosis , Biotinylation , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Chromatin Immunoprecipitation , Gene Knockdown Techniques , Humans , Karyopherins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Phosphatase 2C/metabolism , Proteomics , Receptors, Cytoplasmic and Nuclear/metabolism , Reproducibility of Results , fas Receptor/metabolism
13.
Oncotarget ; 8(28): 45687-45697, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28537899

ABSTRACT

Therapy of cutaneous T cell lymphoma (CTCL) is complicated by a distinct resistance of the malignant T cells towards apoptosis that can be caused by NRAS mutations in late-stage patients. These mutations correlate with decreased overall survival, but sensitize the respective CTCL cells towards MEK-inhibition-induced apoptosis which represents a promising novel therapeutic target in CTCL. Here, we show that the multi-kinase inhibitor Sorafenib induces apoptosis in NRAS-mutated CTCL cells. CTCL cell lines and to a minor extent primary T cells from Sézary patients without NRAS mutations are also affected by Sorafenib-induced apoptosis suggesting a sensitizing role of NRAS mutations for Sorafenib-induced apoptosis. When combining Sorafenib with the established CTCL medication Vorinostat we detected an increase in cell death sensitivity in CTCL cells. The combination treatment acted synergistically in apoptosis induction in both non-mutant and mutant CTCL cells. Mechanistically, this synergistic apoptosis induction by Sorafenib and Vorinostat is based on the downregulation of the anti-apoptotic protein Mcl-1, but not of other Bcl-2 family members. Taken together, these findings suggest that Sorafenib in combination with Vorinostat represents a novel therapeutic approach for the treatment of CTCL patients.


Subject(s)
Antineoplastic Agents/pharmacology , GTP Phosphohydrolases/genetics , Lymphoma, T-Cell, Cutaneous/genetics , Membrane Proteins/genetics , Mutation , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Synergism , GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Molecular Targeted Therapy , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/therapeutic use , Sorafenib , Vorinostat
14.
Immunol Res ; 65(2): 551-563, 2017 04.
Article in English | MEDLINE | ID: mdl-28224362

ABSTRACT

Regulatory T cells (Tregs) are critical mediators of immune tolerance, yet their involvement in the autoimmune disease systemic lupus erythematosus (SLE) is incompletely understood. We analyzed CD4+ T cell subpopulations with Treg-related phenotypes and their association with disease activity in peripheral blood (PB) and tissues of patients with SLE. In detail, we quantified subpopulations regarding CD25, FOXP3, CD62L, CCR6, CD27, CD45RA, and CD45RO expression in PB from 31 patients with SLE divided into two disease activity groups and 32 healthy controls using flow cytometry. CD4+ and FOXP3+ T cells in skin and kidney biopsies of patients with SLE were quantified by immunohistochemistry. CD4+CD25+/++FOXP3+ and CD4+CD25+CD45RA-/CD45RO+ T cell frequencies were significantly higher in PB from patients with active compared to inactive SLE. The fraction of CD4+CD25++FOXP3+ Tregs and CD4+CD25+CD45RA+/CD45RO- naïve Tregs was not significantly different between these groups. CD4+CD25++ Tregs from active SLE patients comprised significantly less CD27+ cells and more CCR6+ cells compared to patients with inactive SLE. The percentage of CD4+FOXP3+ T cells among inflammatory infiltrates in skin and kidney biopsies of SLE patients was not different from other inflammatory skin/kidney diseases. In conclusion, although CD4+FOXP3+ T cell frequencies in the inflamed tissues of SLE patients were comparable to other inflammatory diseases, distinct T cell subpopulations appeared misbalanced in PB of patients with active SLE. Here, cells phenotypically resembling activated T cells, but not Tregs, were increased compared to patients with inactive SLE. Within Tregs of patients with active SLE, markers related to Treg function and homing were altered.


Subject(s)
Kidney/immunology , Lupus Erythematosus, Systemic/immunology , Skin/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Antigens, CD/metabolism , Cell Separation , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Humans , Immunophenotyping , Lupus Erythematosus, Systemic/diagnosis , Male , Middle Aged
16.
Sci Rep ; 7: 43168, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28233787

ABSTRACT

Constitutively active NFκB promotes survival of many cancers, especially T-cell lymphomas and leukemias by upregulating antiapoptotic proteins such as inhibitors of apoptosis (IAPs) and FLICE-like inhibitory proteins (cFLIPs). IAPs and cFLIPs negatively regulate the ripoptosome, which mediates cell death in an apoptotic or necroptotic manner. Here, we demonstrate for the first time, that DMF antagonizes NFκB by suppressing Thioredoxin-1 (Trx1), a major regulator of NFκB transcriptional activity. DMF-mediated inhibition of NFκB causes ripoptosome formation via downregulation of IAPs and cFLIPs. In addition, DMF promotes mitochondrial Smac release and subsequent degradation of IAPs, further enhancing cell death in tumor cells displaying constitutive NFκB activity. Significantly, CTCL patients treated with DMF display substantial ripoptosome formation and caspase-3 cleavage in T-cells. DMF induces cell death predominantly in malignant or activated T-cells. Further, we show that malignant T-cells can die by both apoptosis and necroptosis, in contrast to resting T-cells, which are restricted to apoptosis upon DMF administration. In summary, our data provide new mechanistic insight in the regulation of cell death by targeting NFκB via Trx1 in cancer. Thus, interference with Trx1 activity is a novel approach for treatment of NFκB-dependent tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Dimethyl Fumarate/pharmacology , NF-kappa B/antagonists & inhibitors , Thioredoxins/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Dimethyl Fumarate/administration & dosage , Humans , Sezary Syndrome/drug therapy
17.
Cancer Lett ; 389: 70-77, 2017 03 28.
Article in English | MEDLINE | ID: mdl-27998762

ABSTRACT

Multiple myeloma (MM) is an incurable malignancy by the presently known therapies. TRAIL is a promising anticancer agent that virtually not shows any toxicity to normal cells. We have recently carried out clinical trials with a human circularly permuted TRAIL, CPT, against MM saw a partial response in approximate 20-30% of patients. In the current study, we investigated the cause of CPT resistance and revealed that the majority of the MM patients express elevated levels of c-FLIP. Knockdown of c-FLIP expression by siRNA alone was sufficient to increase CPT-mediated apoptosis in a CPT-resistant human MM cell line U266. To overcome CPT resistance, we investigated the combination of CPT with Rocaglamides(s) in MM which has been shown to inhibit c-FLIP expression in vitro. We show that Rocaglamide(s) overcomes CPT resistance in U266 in vitro and significant increases in anti-tumor efficacies of CPT in mice xenografted with U266. Similar results were also obtained in mice xenografted with the CPT-resistant human acute T-cell leukemia cell line Molt-4. Our study suggests that the combination of Rocaglamide(s) with CPT may provide a more efficient treatment against myeloma and leukemia.


Subject(s)
Benzofurans/therapeutic use , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Multiple Myeloma/drug therapy , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/analysis , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Mice , Recombinant Proteins/therapeutic use , Xenograft Model Antitumor Assays
18.
Blood ; 128(6): 805-15, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27268084

ABSTRACT

Despite intensive efforts in recent years, a curative therapy for cutaneous T-cell lymphoma (CTCL) has not yet been developed. Therefore, the establishment of new therapeutic approaches with higher efficacy rates and milder side effects is strongly desired. A characteristic feature of the malignant T-cell population in CTCL is resistance toward cell death resulting from constitutive NF-κB activation. Therefore, NF-κB-dependent cell death resistance represents an interesting therapeutic target in CTCL because an NF-κB-directed therapy would leave bystander T cells widely unaffected. We investigated the effects of dimethyl fumarate (DMF) on CTCL cells in vitro and in vivo. DMF induced cell death in primary patient-derived CD4(+) cells and CTCL cell lines, but hardly in T cells from healthy donors. DMF-induced cell death was linked specifically to NF-κB inhibition. To study the impact of DMF in vivo, we developed 2 CTCL xenograft mouse models with different cutaneous localizations of the T-cell infiltrate. DMF treatment delayed the growth of CTCL tumors and prevented formation of distant metastases. In addition, DMF induced increased cell death in primary CTCL tumors and in liver metastases. In summary, DMF treatment represents a remarkable therapeutic option in CTCL because it restores CTCL apoptosis in vitro and in preclinical models in vivo and prevents spreading of the disease to distant sites. DMF treatment is of particular promise in CTCL because DMF is already in successful clinical use in the treatment of psoriasis and multiple sclerosis allowing fast translation into clinical studies in CTCL.


Subject(s)
Apoptosis/drug effects , Dimethyl Fumarate/therapeutic use , Immunosuppressive Agents/therapeutic use , Lymphoma, T-Cell, Cutaneous/drug therapy , NF-kappa B/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin/drug effects , Animals , Humans , Lymphoma, T-Cell, Cutaneous/immunology , Lymphoma, T-Cell, Cutaneous/pathology , Mice , NF-kappa B/immunology , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Signal Transduction/drug effects , Skin/immunology , Skin/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Cells, Cultured
19.
Oncotarget ; 7(32): 51908-51921, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27340868

ABSTRACT

Chemotherapy is one of the pillars of anti-cancer therapy. Although chemotherapeutics cause regression of the primary tumor, many chemotherapeutics are often shown to induce or accelerate metastasis formation. Moreover, metastatic tumors are largely resistant against chemotherapy. As more than 90% of cancer patients die due to metastases and not due to primary tumor formation, novel drugs are needed to overcome these shortcomings. In this study, we identified the anticancer phytochemical Rocaglamide (Roc-A) to be an inhibitor of cancer cell migration, a crucial event in metastasis formation. We show that Roc-A inhibits cellular migration and invasion independently of its anti-proliferative and cytotoxic effects in different types of human cancer cells. Mechanistically, Roc-A treatment induces F-actin-based morphological changes in membrane protrusions. Further investigation of the molecular mechanisms revealed that Roc-A inhibits the activities of the small GTPases RhoA, Rac1 and Cdc42, the master regulators of cellular migration. Taken together, our results provide evidence that Roc-A may be a lead candidate for a new class of anticancer drugs that inhibit metastasis formation.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Cell Movement/drug effects , rho GTP-Binding Proteins/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , rho GTP-Binding Proteins/drug effects
20.
Mol Ther ; 24(2): 298-305, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26581163

ABSTRACT

Monoclonal antibodies directed to the B-cell-specific CD20-antigen are successfully used for the treatment of lymphomas and autoimmune diseases. Here, we compare the anti-B-cell activity of three different antibodies directed to CD20: (i) a chimeric, monospecific antibody, (ii) an Fc-optimized variant thereof, and (iii) a bispecific CD20×CD95-antibody in a newly developed recombinant format, termed Fabsc. The bispecific antibody specifically triggers the CD95 death receptor on malignant, as well as activated, normal B-cells. We found that the capability of this antibody to suppress the growth of malignant B-cells in vitro and in vivo and to specifically deplete normal, activated B-cells from peripheral blood mononuclear cell (PBMC) cultures was superior to that of the Fc-optimized monospecific antibody. This antibody in turn was more effective than its nonoptimized variant. Moreover, the bispecific antibody was the only reagent capable of significantly suppressing antibody production in vitro. Our findings imply that the bispecific CD20×CD95-antibody might become a new, prototypical reagent for the treatment of B-cell-mediated autoimmune disease.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antigens, CD20/immunology , B-Lymphocytes/drug effects , Lymphoma, B-Cell/drug therapy , fas Receptor/immunology , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation/drug effects , Lymphoma, B-Cell/immunology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...