Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Immunity ; 55(3): 423-441.e9, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35139355

ABSTRACT

Cell death plays an important role during pathogen infections. Here, we report that interferon-γ (IFNγ) sensitizes macrophages to Toll-like receptor (TLR)-induced death that requires macrophage-intrinsic death ligands and caspase-8 enzymatic activity, which trigger the mitochondrial apoptotic effectors, BAX and BAK. The pro-apoptotic caspase-8 substrate BID was dispensable for BAX and BAK activation. Instead, caspase-8 reduced pro-survival BCL-2 transcription and increased inducible nitric oxide synthase (iNOS), thus facilitating BAX and BAK signaling. IFNγ-primed, TLR-induced macrophage killing required iNOS, which licensed apoptotic caspase-8 activity and reduced the BAX and BAK inhibitors, A1 and MCL-1. The deletion of iNOS or caspase-8 limited SARS-CoV-2-induced disease in mice, while caspase-8 caused lethality independent of iNOS in a model of hemophagocytic lymphohistiocytosis. These findings reveal that iNOS selectively licenses programmed cell death, which may explain how nitric oxide impacts disease severity in SARS-CoV-2 infection and other iNOS-associated inflammatory conditions.


Subject(s)
COVID-19/immunology , Caspase 8/metabolism , Interferon-gamma/metabolism , Lymphohistiocytosis, Hemophagocytic/immunology , Macrophages/immunology , Mitochondria/metabolism , SARS-CoV-2/physiology , Animals , Caspase 8/genetics , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Interferon-gamma/genetics , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Signal Transduction , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
2.
Nature ; 618(7967): 1033-1040, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37316667

ABSTRACT

Most clinically applied cancer immunotherapies rely on the ability of CD8+ cytolytic T cells to directly recognize and kill tumour cells1-3. These strategies are limited by the emergence of major histocompatibility complex (MHC)-deficient tumour cells and the formation of an immunosuppressive tumour microenvironment4-6. The ability of CD4+ effector cells to contribute to antitumour immunity independently of CD8+ T cells is increasingly recognized, but strategies to unleash their full potential remain to be identified7-10. Here, we describe a mechanism whereby a small number of CD4+ T cells is sufficient to eradicate MHC-deficient tumours that escape direct CD8+ T cell targeting. The CD4+ effector T cells preferentially cluster at tumour invasive margins where they interact with MHC-II+CD11c+ antigen-presenting cells. We show that T helper type 1 cell-directed CD4+ T cells and innate immune stimulation reprogramme the tumour-associated myeloid cell network towards interferon-activated antigen-presenting and iNOS-expressing tumouricidal effector phenotypes. Together, CD4+ T cells and tumouricidal myeloid cells orchestrate the induction of remote inflammatory cell death that indirectly eradicates interferon-unresponsive and MHC-deficient tumours. These results warrant the clinical exploitation of this ability of CD4+ T cells and innate immune stimulators in a strategy to complement the direct cytolytic activity of CD8+ T cells and natural killer cells and advance cancer immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Death , Immunotherapy , Inflammation , Neoplasms , Tumor Microenvironment , Humans , Antigen-Presenting Cells/immunology , CD11c Antigen/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Death/immunology , Histocompatibility Antigens Class II/immunology , Immunity, Innate , Inflammation/immunology , Interferons/immunology , Major Histocompatibility Complex/immunology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Tumor Microenvironment/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Th1 Cells/cytology , Th1 Cells/immunology
3.
EMBO J ; 42(22): e113614, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37789765

ABSTRACT

Cellular inhibitor of apoptosis proteins (cIAPs) are RING-containing E3 ubiquitin ligases that ubiquitylate receptor-interacting protein kinase 1 (RIPK1) to regulate TNF signalling. Here, we established mice simultaneously expressing enzymatically inactive cIAP1/2 variants, bearing mutations in the RING domains of cIAP1/2 (cIAP1/2 mutant RING, cIAP1/2MutR ). cIap1/2MutR/MutR mice died during embryonic development due to RIPK1-mediated apoptosis. While expression of kinase-inactive RIPK1D138N rescued embryonic development, Ripk1D138N/D138N /cIap1/2MutR/MutR mice developed systemic inflammation and died postweaning. Cells expressing cIAP1/2MutR and RIPK1D138N were still susceptible to TNF-induced apoptosis and necroptosis, implying additional kinase-independent RIPK1 activities in regulating TNF signalling. Although further ablation of Ripk3 did not lead to any phenotypic improvement, Tnfr1 gene knock-out prevented early onset of systemic inflammation and premature mortality, indicating that cIAPs control TNFR1-mediated toxicity independent of RIPK1 and RIPK3. Beyond providing novel molecular insights into TNF-signalling, the mouse model established in this study can serve as a useful tool to further evaluate ongoing therapeutic protocols using inhibitors of TNF, cIAPs and RIPK1.


Subject(s)
Inhibitor of Apoptosis Proteins , Receptors, Tumor Necrosis Factor, Type I , Animals , Mice , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Cell Death , Apoptosis , Inflammation/genetics , Inflammation/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology
4.
EMBO J ; 41(2): e108690, 2022 12 17.
Article in English | MEDLINE | ID: mdl-34931711

ABSTRACT

During apoptosis, the BCL-2-family protein tBID promotes mitochondrial permeabilization by activating BAX and BAK and by blocking anti-apoptotic BCL-2 members. Here, we report that tBID can also mediate mitochondrial permeabilization by itself, resulting in release of cytochrome c and mitochondrial DNA, caspase activation and apoptosis even in absence of BAX and BAK. This previously unrecognized activity of tBID depends on helix 6, homologous to the pore-forming regions of BAX and BAK, and can be blocked by pro-survival BCL-2 proteins. Importantly, tBID-mediated mitochondrial permeabilization independent of BAX and BAK is physiologically relevant for SMAC release in the immune response against Shigella infection. Furthermore, it can be exploited to kill leukaemia cells with acquired venetoclax resistance due to lack of active BAX and BAK. Our findings define tBID as an effector of mitochondrial permeabilization in apoptosis and provide a new paradigm for BCL-2 proteins, with implications for anti-bacterial immunity and cancer therapy.


Subject(s)
Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/chemistry , BH3 Interacting Domain Death Agonist Protein/genetics , HCT116 Cells , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Domains , Proteolysis , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
5.
Nature ; 575(7784): 683-687, 2019 11.
Article in English | MEDLINE | ID: mdl-31748744

ABSTRACT

Caspase-8 is the initiator caspase of extrinsic apoptosis1,2 and inhibits necroptosis mediated by RIPK3 and MLKL. Accordingly, caspase-8 deficiency in mice causes embryonic lethality3, which can be rescued by deletion of either Ripk3 or Mlkl4-6. Here we show that the expression of enzymatically inactive CASP8(C362S) causes embryonic lethality in mice by inducing necroptosis and pyroptosis. Similar to Casp8-/- mice3,7, Casp8C362S/C362S mouse embryos died after endothelial cell necroptosis leading to cardiovascular defects. MLKL deficiency rescued the cardiovascular phenotype but unexpectedly caused perinatal lethality in Casp8C362S/C362S mice, indicating that CASP8(C362S) causes necroptosis-independent death at later stages of embryonic development. Specific loss of the catalytic activity of caspase-8 in intestinal epithelial cells induced intestinal inflammation similar to intestinal epithelial cell-specific Casp8 knockout mice8. Inhibition of necroptosis by additional deletion of Mlkl severely aggravated intestinal inflammation and caused premature lethality in Mlkl knockout mice with specific loss of caspase-8 catalytic activity in intestinal epithelial cells. Expression of CASP8(C362S) triggered the formation of ASC specks, activation of caspase-1 and secretion of IL-1ß. Both embryonic lethality and premature death were completely rescued in Casp8C362S/C362SMlkl-/-Asc-/- or Casp8C362S/C362SMlkl-/-Casp1-/- mice, indicating that the activation of the inflammasome promotes CASP8(C362S)-mediated tissue pathology when necroptosis is blocked. Therefore, caspase-8 represents the molecular switch that controls apoptosis, necroptosis and pyroptosis, and prevents tissue damage during embryonic development and adulthood.


Subject(s)
Apoptosis/genetics , Caspase 8/genetics , Caspase 8/metabolism , Necroptosis/genetics , Pyroptosis/genetics , Animals , Cell Line , Cells, Cultured , Enzyme Activation/genetics , Gene Expression Profiling , Gene Knockout Techniques , HEK293 Cells , Humans , Inflammasomes/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Keratinocytes/cytology , Keratinocytes/pathology , Mice , Mutation , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism
6.
Blood ; 140(10): 1119-1131, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35759728

ABSTRACT

Unique molecular vulnerabilities have been identified in the aggressive MCD/C5 genetic subclass of diffuse large B-cell lymphoma (DLBCL). However, the premalignant cell-of-origin exhibiting MCD-like dependencies remains elusive. In this study, we examined animals carrying up to 4 hallmark genetic lesions found in MCD consisting of gain-of-function mutations in Myd88 and Cd79b, loss of Prdm1, and overexpression of BCL2. We discovered that expression of combinations of these alleles in vivo promoted a cell-intrinsic accumulation of B cells in spontaneous splenic germinal centers (GCs). As with MCD, these premalignant B cells were enriched for B-cell receptors (BCRs) with evidence of self-reactivity, displayed a de novo dependence on Tlr9, and were more sensitive to inhibition of Bruton's tyrosine kinase. Mutant spontaneous splenic GC B cells (GCB) showed increased proliferation and IRF4 expression. Mice carrying all 4 genetic lesions showed a >50-fold expansion of spontaneous splenic GCs exhibiting aberrant histologic features with a dark zone immunophenotype and went on to develop DLBCL in the spleen with age. Thus, by combining multiple hallmark genetic alterations associated with MCD, our study identifies aberrant spontaneous splenic GCBs as a likely cell-of-origin for this aggressive genetic subtype of lymphoma.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Spleen , Animals , B-Lymphocytes/pathology , Germinal Center/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mutation , Spleen/pathology
7.
EMBO Rep ; 23(6): e53608, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35437868

ABSTRACT

Elevated expression of the X-linked inhibitor of apoptosis protein (XIAP) has been frequently reported in malignant melanoma suggesting that XIAP renders apoptosis resistance and thereby supports melanoma progression. Independent of its anti-apoptotic function, XIAP mediates cellular inflammatory signalling and promotes immunity against bacterial infection. The pro-inflammatory function of XIAP has not yet been considered in cancer. By providing detailed in vitro analyses, utilising two independent mouse melanoma models and including human melanoma samples, we show here that XIAP is an important mediator of melanoma neutrophil infiltration. Neutrophils represent a major driver of melanoma progression and are increasingly considered as a valuable therapeutic target in solid cancer. Our data reveal that XIAP ubiquitylates RIPK2, involve TAB1/RIPK2 complex and induce the transcriptional up-regulation and secretion of chemokines such as IL8, that are responsible for intra-tumour neutrophil accumulation. Alteration of the XIAP-RIPK2-TAB1 inflammatory axis or the depletion of neutrophils in mice reduced melanoma growth. Our data shed new light on how XIAP contributes to tumour growth and provides important insights for novel XIAP targeting strategies in cancer.


Subject(s)
Inhibitor of Apoptosis Proteins , Melanoma , Neutrophil Infiltration , Skin Neoplasms , X-Linked Inhibitor of Apoptosis Protein , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Disease Models, Animal , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/immunology , Interleukin-8/biosynthesis , Melanoma/genetics , Melanoma/immunology , Mice , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/immunology , X-Linked Inhibitor of Apoptosis Protein/metabolism
8.
EMBO Rep ; 22(10): e52301, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34342114

ABSTRACT

Maintaining the architecture, size and composition of an intact stem cell (SC) compartment is crucial for tissue homeostasis and regeneration throughout life. In mammalian skin, elevated expression of the anti-apoptotic Bcl-2 protein has been reported in hair follicle (HF) bulge SCs (BSCs), but its impact on SC function is unknown. Here, we show that systemic exposure of mice to the Bcl-2 antagonist ABT-199/venetoclax leads to the selective loss of suprabasal BSCs (sbBSCs), thereby disrupting cyclic HF regeneration. RNAseq analysis shows that the pro-apoptotic BH3-only proteins BIM and Bmf are upregulated in sbBSCs, explaining their addiction to Bcl-2 and the marked susceptibility to Bcl-2 antagonism. In line with these observations, conditional knockout of Bcl-2 in mouse epidermis elevates apoptosis in BSCs. In contrast, ectopic Bcl-2 expression blocks apoptosis during HF regression, resulting in the accumulation of quiescent SCs and delaying HF growth in mice. Strikingly, Bcl-2-induced changes in size and composition of the HF bulge accelerate tumour formation. Our study identifies a niche-instructive mechanism of Bcl-2-regulated apoptosis response that is required for SC homeostasis and tissue regeneration, and may suppress carcinogenesis.


Subject(s)
Apoptosis Regulatory Proteins , Hair Follicle , Animals , Apoptosis/genetics , Mice , Skin , Stem Cells
9.
Proc Natl Acad Sci U S A ; 117(40): 24957-24963, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32963096

ABSTRACT

B lymphocytes acquire self-reactivity as an unavoidable byproduct of antibody gene diversification in the bone marrow and in germinal centers (GCs). Autoreactive B cells emerging from the bone marrow are silenced in a series of well-defined checkpoints, but less is known about how self-reactivity that develops by somatic mutation in GCs is controlled. Here, we report the existence of an apoptosis-dependent tolerance checkpoint in post-GC B cells. Whereas defective GC B cell apoptosis has no measurable effect on autoantibody development, disruption of post-GC apoptosis results in accumulation of autoreactive memory B cells and plasma cells, antinuclear antibody production, and autoimmunity. The data presented shed light on mechanisms that regulate immune tolerance and the development of autoantibodies.


Subject(s)
Apoptosis/genetics , Autoimmunity/genetics , Genes, Immunoglobulin/genetics , Immune Tolerance/genetics , Animals , Antibodies, Antinuclear/immunology , Apoptosis/immunology , Autoantibodies/immunology , Autoimmunity/immunology , B-Lymphocytes/immunology , Genes, Immunoglobulin/immunology , Germinal Center/immunology , Humans , Immunologic Memory/genetics , Immunologic Memory/immunology , Mice , Plasma Cells/immunology
10.
Nat Immunol ; 10(7): 761-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19525969

ABSTRACT

Granule-mediated cytotoxicity is the main effector mechanism of cytotoxic CD8+ T cells. We report that CD8+ T cells from acid sphingomyelinase (ASMase)-deficient (ASMase-KO) mice are defective in exocytosis of cytolytic effector molecules; this defect resulted in attenuated cytotoxic activity of ASMase-KO CD8+ T cells and delayed elimination of lymphocytic choriomeningitis virus from ASMase-KO mice. Cytolytic granules of ASMase-KO and wild-type CD8+ T cells were equally loaded with granzymes and perforin, and correctly directed to the immunological synapse. In wild-type CD8+ T cells, secretory granules underwent shrinkage by 82% after fusion with the plasma membrane. In ASMase-KO CD8+ T cells, the contraction of secretory granules was markedly impaired. Thus, ASMase is required for contraction of secretory granules and expulsion of cytotoxic effector molecules.


Subject(s)
Cytotoxicity, Immunologic/immunology , Secretory Vesicles/metabolism , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes/immunology , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/metabolism , Arenaviridae Infections/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CCL5/metabolism , Female , Granzymes/genetics , Granzymes/metabolism , Immunoblotting , Immunological Synapses/immunology , Lymphocytic choriomeningitis virus/physiology , Male , Mice , Mice, Knockout , Microscopy, Fluorescence , Perforin/genetics , Perforin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Secretory Vesicles/enzymology , Sphingomyelin Phosphodiesterase/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
11.
Carcinogenesis ; 41(12): 1735-1745, 2020 12 31.
Article in English | MEDLINE | ID: mdl-32255484

ABSTRACT

Functioning mitochondria are crucial for cancer metabolism, but aerobic glycolysis is still considered to be an important pathway for energy production in many tumor cells. Here we show that two well established, classic Hodgkin lymphoma (cHL) cell lines harbor deleterious variants within mitochondrial DNA (mtDNA) and thus exhibit reduced steady-state levels of respiratory chain complexes. However, instead of resulting in the expected bioenergetic defect, these mtDNA variants evoke a retrograde signaling response that induces mitochondrial biogenesis and ultimately results in increased mitochondrial mass as well as function and enhances proliferation in vitro as well as tumor growth in mice in vivo. When complex I assembly was impaired by knockdown of one of its subunits, this led to further increased mitochondrial mass and function and, consequently, further accelerated tumor growth in vivo. In contrast, inhibition of mitochondrial respiration in vivo by the mitochondrial complex I inhibitor metformin efficiently slowed down growth. We conclude that, as a new mechanism, mildly deleterious mtDNA variants in cHL cancer cells cause an increase of mitochondrial mass and enhanced function as a compensatory effect using a retrograde signaling pathway, which provides an obvious advantage for tumor growth.


Subject(s)
Carcinogenesis/pathology , DNA, Mitochondrial/genetics , Hodgkin Disease/pathology , Mutation , Organelle Biogenesis , Animals , Apoptosis , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Hodgkin Disease/genetics , Hodgkin Disease/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oxidative Phosphorylation , Reed-Sternberg Cells , Xenograft Model Antitumor Assays
12.
J Hepatol ; 73(6): 1347-1359, 2020 12.
Article in English | MEDLINE | ID: mdl-32598967

ABSTRACT

BACKGROUND & AIMS: Selective elimination of virus-infected hepatocytes occurs through virus-specific CD8 T cells recognizing peptide-loaded MHC molecules. Herein, we report that virus-infected hepatocytes are also selectively eliminated through a cell-autonomous mechanism. METHODS: We generated recombinant adenoviruses and genetically modified mouse models to identify the molecular mechanisms determining TNF-induced hepatocyte apoptosis in vivo and used in vivo bioluminescence imaging, immunohistochemistry, immunoblot analysis, RNAseq/proteome/phosphoproteome analyses, bioinformatic analyses, mitochondrial function tests. RESULTS: We found that TNF precisely eliminated only virus-infected hepatocytes independently of local inflammation and activation of immune sensory receptors. TNF receptor I was equally relevant for NF-kB activation in healthy and infected hepatocytes, but selectively mediated apoptosis in infected hepatocytes. Caspase 8 activation downstream of TNF receptor signaling was dispensable for apoptosis in virus-infected hepatocytes, indicating an unknown non-canonical cell-intrinsic pathway promoting apoptosis in hepatocytes. We identified a unique state of mitochondrial vulnerability in virus-infected hepatocytes as the cause for this non-canonical induction of apoptosis through TNF. Mitochondria from virus-infected hepatocytes showed normal biophysical and bioenergetic functions but were characterized by reduced resilience to calcium challenge. In the presence of unchanged TNF-induced signaling, reactive oxygen species-mediated calcium release from the endoplasmic reticulum caused mitochondrial permeability transition and apoptosis, which identified a link between extrinsic death receptor signaling and cell-intrinsic mitochondrial-mediated caspase activation. CONCLUSION: Our findings reveal a novel concept in immune surveillance by identifying a cell-autonomous defense mechanism that selectively eliminates virus-infected hepatocytes through mitochondrial permeability transition. LAY SUMMARY: The liver is known for its unique immune functions. Herein, we identify a novel mechanism by which virus-infected hepatocytes can selectively eliminate themselves through reduced mitochondrial resilience to calcium challenge.


Subject(s)
Caspase 8/metabolism , Hepatocytes , Mitochondria, Liver , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Apoptosis/immunology , Calcium Signaling , Cells, Cultured , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Mice , Mitochondria, Liver/immunology , Mitochondria, Liver/metabolism , Mitochondrial Transmembrane Permeability-Driven Necrosis , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
13.
Br J Cancer ; 120(1): 69-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30377339

ABSTRACT

BACKGROUND: Immune infiltration is implicated in the development of acquired resistance to anti-angiogenic cancer therapy. We therefore investigated the correlation between neutrophil infiltration in metastasis of colorectal cancer (CRC) patients and survival after treatment with bevacizumab. Our study identifies CD177+ tumour neutrophil infiltration as an adverse prognostic factor for bevacizumab treatment. We further demonstrate that a novel anti-VEGF/anti-Ang2 compound (BI-880) can overcome resistance to VEGF inhibition in experimental tumour models. METHODS: A total of 85 metastatic CRC patients were stratified into cohorts that had either received chemotherapy alone (n = 39) or combined with bevacizumab (n = 46). Tumour CD177+ neutrophil infiltration was correlated to clinical outcome. The impact of neutrophil infiltration on anti-VEGF or anti-VEGF/anti-Ang2 therapy was studied in both xenograft and syngeneic tumour models by immunohistochemistry. RESULTS: The survival of bevacizumab-treated CRC patients in the presence of CD177+ infiltrates was significantly reduced compared to patients harbouring CD177- metastases. BI-880 treatment reduced the development of hypoxia associated with bevacizumab treatment and improved vascular normalisation in xenografts. Furthermore, neutrophil depletion or BI-880 treatment restored treatment sensitivity in a syngeneic tumour model of anti-VEGF resistance. CONCLUSIONS: Our findings implicate CD177 as a biomarker for bevacizumab and suggest VEGF/Ang2 inhibition as a strategy to overcome neutrophil associated resistance to anti-angiogenic treatment.


Subject(s)
Colorectal Neoplasms/drug therapy , Isoantigens/genetics , Neovascularization, Pathologic/drug therapy , Receptors, Cell Surface/genetics , Vascular Endothelial Growth Factor A/genetics , Vesicular Transport Proteins/genetics , Aged , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab/administration & dosage , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Female , GPI-Linked Proteins/genetics , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neutrophils/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays
14.
BMC Cancer ; 19(1): 531, 2019 May 31.
Article in English | MEDLINE | ID: mdl-31151416

ABSTRACT

BACKGROUND: Molecular markers predicting survival in esophageal adenocarcinoma (EAC) are rare. Specifically, in favorable oncologic situations, e.g. nodal negativity or major neoadjuvant therapy response, there is a lack of additional risk factors that serve to predict patients' outcome more precisely. This study evaluated X-linked inhibitor of apoptosis protein (XIAP) as a potential marker improving outcome prediction. METHODS: Tissue microarrays from 362 patients that were diagnosed with resectable EAC were included in the study. XIAP was stained by immunohistochemistry and correlated to clinical outcome, molecular markers and markers of the cellular tumor microenvironment. RESULTS: XIAP did not impact on overall survival (OS) in the whole study collective. Subgroup analyses stratifying for common genetic markers (TP53, ERBB2, ARID1A/SWI/SNF) did not disclose any impact of XIAP expression on survival. Detailed subgroup analyses of [1] nodal negative patients, [2] highly T-cell infiltrated tumors and [3] therapy responders to neoadjuvant treatment revealed a significant inverse role of high XIAP expression in these specific oncologic situations; elevated XIAP expression detrimentally affected patients' outcome in these subgroups. [1]: OS XIAP low: 202 months (m) vs. XIAP high: 38 m; [2]: OS 116 m vs. 28.2 m; [3]: OS 31 m vs. 4 m). CONCLUSIONS: Our data suggest XIAP expression in EAC as a worthy tool to improve outcome prediction in specific oncologic settings that might directly impact on clinical diagnosis and treatment of EAC in the future.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Lymphocytes, Tumor-Infiltrating/metabolism , Neoadjuvant Therapy , T-Lymphocyte Subsets/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Aged , Biomarkers, Tumor/metabolism , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophagectomy , Female , Humans , Immunohistochemistry , Lymph Nodes/pathology , Male , Neoadjuvant Therapy/methods , Prognosis , Retrospective Studies , Survival Rate
15.
EMBO J ; 33(19): 2171-87, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25056906

ABSTRACT

The X-linked inhibitor of apoptosis protein (XIAP) is a potent caspase inhibitor, best known for its anti-apoptotic function in cancer. During apoptosis, XIAP is antagonized by SMAC, which is released from the mitochondria upon caspase-mediated activation of BID. Recent studies suggest that XIAP is involved in immune signaling. Here, we explore XIAP as an important mediator of an immune response against the enteroinvasive bacterium Shigella flexneri, both in vitro and in vivo. Our data demonstrate for the first time that Shigella evades the XIAP-mediated immune response by inducing the BID-dependent release of SMAC from the mitochondria. Unlike apoptotic stimuli, Shigella activates the calpain-dependent cleavage of BID to trigger the release of SMAC, which antagonizes the inflammatory action of XIAP without inducing apoptosis. Our results demonstrate how the cellular death machinery can be subverted by an invasive pathogen to ensure bacterial colonization.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/physiology , Carrier Proteins/metabolism , Dysentery, Bacillary/immunology , Mitochondria/immunology , Mitochondrial Proteins/metabolism , Shigella/immunology , X-Linked Inhibitor of Apoptosis Protein/physiology , Animals , Apoptosis , Apoptosis Regulatory Proteins , Blotting, Western , Caspases/metabolism , Cell Proliferation , Cells, Cultured , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/pathology , Female , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Immunoenzyme Techniques , Integrases/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/immunology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Shigella/pathogenicity , Signal Transduction , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
16.
Blood ; 127(22): 2732-41, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27048211

ABSTRACT

The adaptor protein MYD88 is critical for relaying activation of Toll-like receptor signaling to NF-κB activation. MYD88 mutations, particularly the p.L265P mutation, have been described in numerous distinct B-cell malignancies, including diffuse large B-cell lymphoma (DLBCL). Twenty-nine percent of activated B-cell-type DLBCL (ABC-DLBCL), which is characterized by constitutive activation of the NF-κB pathway, carry the p.L265P mutation. In addition, ABC-DLBCL frequently displays focal copy number gains affecting BCL2 Here, we generated a novel mouse model in which Cre-mediated recombination, specifically in B cells, leads to the conditional expression of Myd88(p.L252P) (the orthologous position of the human MYD88(p.L265P) mutation) from the endogenous locus. These mice develop a lymphoproliferative disease and occasional transformation into clonal lymphomas. The clonal disease displays the morphologic and immunophenotypical characteristics of ABC-DLBCL. Lymphomagenesis can be accelerated by crossing in a further novel allele, which mediates conditional overexpression of BCL2 Cross-validation experiments in human DLBCL samples revealed that both MYD88 and CD79B mutations are substantially enriched in ABC-DLBCL compared with germinal center B-cell DLBCL. Furthermore, analyses of human DLBCL genome sequencing data confirmed that BCL2 amplifications frequently co-occurred with MYD88 mutations, further validating our approach. Finally, in silico experiments revealed that MYD88-mutant ABC-DLBCL cells in particular display an actionable addiction to BCL2. Altogether, we generated a novel autochthonous mouse model of ABC-DLBCL that could be used as a preclinical platform for the development and validation of novel therapeutic approaches for the treatment of ABC-DLBCL.


Subject(s)
B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Mutation, Missense , Myeloid Differentiation Factor 88/biosynthesis , Neoplasms, Experimental/metabolism , Animals , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics
17.
EMBO Rep ; 16(6): 719-27, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25825408

ABSTRACT

In this study, we show for the first time that the therapeutic antagonization of inhibitor of apoptosis proteins (IAPs) inhibits B16 melanoma growth by disrupting tumor vasculature. Specifically, the treatment of mice bearing B16 melanoma with an IAP antagonist compound A (Comp A) inhibits tumor growth not by inducing direct cytotoxicity against B16 cells but rather by a hitherto unrecognized antiangiogenic activity against tumor vessels. Our detailed analysis showed that Comp A treatment induces NF-κB activity in B16 tumor cells and facilitates the production of TNF. In the presence of Comp A, endothelial cells (ECs) become highly susceptible to TNF and undergo apoptotic cell death. Accordingly, the antiangiogenic and growth-attenuating effects of Comp A treatment were completely abolished in TNF-R knockout mice. This novel targeting approach could be of clinical value in controlling pathological neoangiogenesis under inflammatory condition while sparing blood vessels under normal condition.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/pathology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Animals , Apoptosis/drug effects , Inflammation/physiopathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Neovascularization, Pathologic , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
PLoS Pathog ; 10(9): e1004351, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25187968

ABSTRACT

NOD1 is an intracellular pathogen recognition receptor that contributes to anti-bacterial innate immune responses, adaptive immunity and tissue homeostasis. NOD1-induced signaling relies on actin remodeling, however, the details of the connection of NOD1 and the actin cytoskeleton remained elusive. Here, we identified in a druggable-genome wide siRNA screen the cofilin phosphatase SSH1 as a specific and essential component of the NOD1 pathway. We show that depletion of SSH1 impaired pathogen induced NOD1 signaling evident from diminished NF-κB activation and cytokine release. Chemical inhibition of actin polymerization using cytochalasin D rescued the loss of SSH1. We further demonstrate that NOD1 directly interacted with SSH1 at F-actin rich sites. Finally, we show that enhanced cofilin activity is intimately linked to NOD1 signaling. Our data thus provide evidence that NOD1 requires the SSH1/cofilin network for signaling and to detect bacterial induced changes in actin dynamics leading to NF-κB activation and innate immune responses.


Subject(s)
Actins/metabolism , Cofilin 1/metabolism , Dysentery, Bacillary/microbiology , Nod1 Signaling Adaptor Protein/metabolism , Phosphoprotein Phosphatases/metabolism , Shigella flexneri/physiology , Actins/chemistry , Blotting, Western , Cells, Cultured , Cofilin 1/genetics , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation , HeLa Cells , High-Throughput Screening Assays , Humans , Immunoenzyme Techniques , Immunoprecipitation , Inflammation , Inflammation Mediators/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod1 Signaling Adaptor Protein/genetics , Phosphoprotein Phosphatases/genetics , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
19.
Nature ; 460(7259): 1159-63, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19641494

ABSTRACT

Reactive oxygen species (ROS) produced by NADPH oxidase function as defence and signalling molecules related to innate immunity and various cellular responses. The activation of NADPH oxidase in response to plasma membrane receptor activation depends on the phosphorylation of cytoplasmic oxidase subunits, their translocation to membranes and the assembly of all NADPH oxidase components. Tumour necrosis factor (TNF) is a prominent stimulus of ROS production, but the molecular mechanisms by which TNF activates NADPH oxidase are poorly understood. Here we identify riboflavin kinase (RFK, formerly known as flavokinase) as a previously unrecognized TNF-receptor-1 (TNFR1)-binding protein that physically and functionally couples TNFR1 to NADPH oxidase. In mouse and human cells, RFK binds to both the TNFR1-death domain and to p22(phox), the common subunit of NADPH oxidase isoforms. RFK-mediated bridging of TNFR1 and p22(phox) is a prerequisite for TNF-induced but not for Toll-like-receptor-induced ROS production. Exogenous flavin mononucleotide or FAD was able to substitute fully for TNF stimulation of NADPH oxidase in RFK-deficient cells. RFK is rate-limiting in the synthesis of FAD, an essential prosthetic group of NADPH oxidase. The results suggest that TNF, through the activation of RFK, enhances the incorporation of FAD in NADPH oxidase enzymes, a critical step for the assembly and activation of NADPH oxidase.


Subject(s)
NADPH Oxidases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Cell Line , Cytochrome b Group/metabolism , Enzyme Activation , Fibroblasts , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/biosynthesis , Flavin-Adenine Dinucleotide/metabolism , HeLa Cells , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Membrane Glycoproteins/metabolism , Mice , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/chemistry , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Binding , Protein Structure, Tertiary , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor, Type I/chemistry
20.
J Immunol ; 189(4): 1559-66, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22802420

ABSTRACT

Factor associated with neutral sphingomyelinase activity (FAN) is an adaptor protein that specifically binds to the p55 receptor for TNF (TNF-RI). Our previous investigations demonstrated that FAN plays a role in TNF-induced actin reorganization by connecting the plasma membrane with actin cytoskeleton, suggesting that FAN may impact on cellular motility in response to TNF and in the context of immune inflammatory conditions. In this study, we used the translucent zebrafish larvae for in vivo analysis of leukocyte migration after morpholino knockdown of FAN. FAN-deficient zebrafish leukocytes were impaired in their migration toward tail fin wounds, leading to a reduced number of cells reaching the wound. Furthermore, FAN-deficient leukocytes show an impaired response to bacterial infections, suggesting that FAN is generally required for the directed chemotactic response of immune cells independent of the nature of the stimulus. Cell-tracking analysis up to 3 h after injury revealed that the reduced number of leukocytes is not due to a reduction in random motility or speed of movement. Leukocytes from FAN-deficient embryos protrude pseudopodia in all directions instead of having one clear leading edge. Our results suggest that FAN-deficient leukocytes exhibit an impaired navigational capacity, leading to a disrupted chemotactic response.


Subject(s)
Bacterial Infections/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Leukocytes/immunology , Sphingomyelin Phosphodiesterase/metabolism , Wound Healing/physiology , Amino Acid Sequence , Animals , Bacterial Infections/metabolism , Chemotaxis, Leukocyte , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/immunology , Larva , Leukocytes/cytology , Microscopy, Confocal , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sphingomyelin Phosphodiesterase/immunology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL