Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nat Immunol ; 21(5): 555-566, 2020 05.
Article in English | MEDLINE | ID: mdl-32327756

ABSTRACT

Regulatory myeloid immune cells, such as myeloid-derived suppressor cells (MDSCs), populate inflamed or cancerous tissue and block immune cell effector functions. The lack of mechanistic insight into MDSC suppressive activity and a marker for their identification has hampered attempts to overcome T cell inhibition and unleash anti-cancer immunity. Here, we report that human MDSCs were characterized by strongly reduced metabolism and conferred this compromised metabolic state to CD8+ T cells, thereby paralyzing their effector functions. We identified accumulation of the dicarbonyl radical methylglyoxal, generated by semicarbazide-sensitive amine oxidase, to cause the metabolic phenotype of MDSCs and MDSC-mediated paralysis of CD8+ T cells. In a murine cancer model, neutralization of dicarbonyl activity overcame MDSC-mediated T cell suppression and, together with checkpoint inhibition, improved the efficacy of cancer immune therapy. Our results identify the dicarbonyl methylglyoxal as a marker metabolite for MDSCs that mediates T cell paralysis and can serve as a target to improve cancer immune therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Melanoma/immunology , Myeloid-Derived Suppressor Cells/immunology , Pyruvaldehyde/metabolism , Amine Oxidase (Copper-Containing)/metabolism , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Communication , Cell Proliferation , Humans , Immune Tolerance , Lymphocyte Activation , Melanoma, Experimental , Mice , Mice, Transgenic , Neoplasms, Experimental , Programmed Cell Death 1 Receptor/metabolism
2.
Nature ; 592(7854): 444-449, 2021 04.
Article in English | MEDLINE | ID: mdl-33762736

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a manifestation of systemic metabolic disease related to obesity, and causes liver disease and cancer1,2. The accumulation of metabolites leads to cell stress and inflammation in the liver3, but mechanistic understandings of liver damage in NASH are incomplete. Here, using a preclinical mouse model that displays key features of human NASH (hereafter, NASH mice), we found an indispensable role for T cells in liver immunopathology. We detected the hepatic accumulation of CD8 T cells with phenotypes that combined tissue residency (CXCR6) with effector (granzyme) and exhaustion (PD1) characteristics. Liver CXCR6+ CD8 T cells were characterized by low activity of the FOXO1 transcription factor, and were abundant in NASH mice and in patients with NASH. Mechanistically, IL-15 induced FOXO1 downregulation and CXCR6 upregulation, which together rendered liver-resident CXCR6+ CD8 T cells susceptible to metabolic stimuli (including acetate and extracellular ATP) and collectively triggered auto-aggression. CXCR6+ CD8 T cells from the livers of NASH mice or of patients with NASH had similar transcriptional signatures, and showed auto-aggressive killing of cells in an MHC-class-I-independent fashion after signalling through P2X7 purinergic receptors. This killing by auto-aggressive CD8 T cells fundamentally differed from that by antigen-specific cells, which mechanistically distinguishes auto-aggressive and protective T cell immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Liver/immunology , Liver/pathology , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/pathology , Receptors, CXCR6/immunology , Acetates/pharmacology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Cell Death/drug effects , Cell Death/immunology , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Interleukin-15/immunology , Interleukin-15/pharmacology , Liver/drug effects , Male , Mice , Mice, Inbred C57BL
3.
J Hepatol ; 78(4): 820-835, 2023 04.
Article in English | MEDLINE | ID: mdl-36681162

ABSTRACT

BACKGROUND & AIMS: Hepatocyte growth and proliferation depends on membrane phospholipid biosynthesis. Short-chain fatty acids (SCFAs) generated by bacterial fermentation, delivered through the gut-liver axis, significantly contribute to lipid biosynthesis. We therefore hypothesized that dysbiotic insults like antibiotic treatment not only affect gut microbiota, but also impair hepatic lipid synthesis and liver regeneration. METHODS: Stable isotope labeling and 70% partial hepatectomy (PHx) was carried out in C57Bl/6J wild-type mice, in mice treated with broad-spectrum antibiotics, in germ-free mice and mice colonized with minimal microbiota. The microbiome was analyzed by 16S rRNA gene sequencing and microbial culture. Gut content, liver, blood and primary hepatocyte organoids were tested by mass spectrometry-based lipidomics, quantitative reverse-transcription PCR (qRT-PCR), immunoblot and immunohistochemistry for expression of proliferative and lipogenic markers. Matched biopsies from hyperplastic and hypoplastic liver tissue of patients subjected to surgical intervention to induce hyperplasia were analyzed by qRT-PCR for lipogenic enzymes. RESULTS: Three days of antibiotic treatment induced persistent dysbiosis with significantly decreased beta-diversity and richness, but a massive increase of Proteobacteria, accompanied by decreased colonic SCFAs. After PHx, antibiotic-treated mice showed delayed liver regeneration, increased mortality, impaired hepatocyte proliferation and decreased hepatic phospholipid synthesis. Expression of the lipogenic enzyme SCD1 was upregulated after PHx but delayed by antibiotic treatment. Germ-free mice essentially recapitulated the phenotype of antibiotic treatment. Phospholipid biosynthesis, hepatocyte proliferation, liver regeneration and survival were rescued in gnotobiotic mice colonized with a minimal SCFA-producing microbial community. SCFAs induced the growth of murine hepatocyte organoids and hepatic SCD1 expression in mice. Further, SCD1 was required for proliferation of human hepatoma cells and was associated with liver regeneration in human patients. CONCLUSION: Gut microbiota are pivotal for hepatic membrane phospholipid biosynthesis and liver regeneration. IMPACT AND IMPLICATIONS: Gut microbiota affect hepatic lipid metabolism through the gut-liver axis, but the underlying mechanisms are poorly understood. Perturbations of the gut microbiome, e.g. by antibiotics, impair the production of bacterial metabolites, which normally serve as building blocks for membrane lipids in liver cells. As a consequence, liver regeneration and survival after liver surgery is severely impaired. Even though this study is preclinical, its results might allow physicians in the future to improve patient outcomes after liver surgery, by modulation of gut microbiota or their metabolites.


Subject(s)
Cell Membrane , Gastrointestinal Microbiome , Hepatocytes , Liver Regeneration , Phospholipids , Animals , Humans , Mice , Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Hyperplasia/metabolism , Hyperplasia/pathology , Liver/pathology , Liver Regeneration/physiology , Mice, Inbred C57BL , Phospholipids/biosynthesis , Phospholipids/metabolism , RNA, Ribosomal, 16S , Hepatocytes/metabolism , Cell Membrane/metabolism
5.
HPB (Oxford) ; 24(8): 1362-1364, 2022 08.
Article in English | MEDLINE | ID: mdl-35289281

ABSTRACT

BACKGROUND: The first-line therapy for liver malignancies is a radical extended liver resection. This high-risk operation has a high incidence of post-hepatectomy liver failure (PHLF) due to a small future liver remnant (FLR). One of the procedures to increase the FLR is the associating liver partition and portal vein ligation for staged hepatectomy (ALPPS) which is still associated with high morbidity and mortality. Here, we present a new, less invasive ALPPS variant that may be associated with lower morbidity. METHODS: SoftALPPS is characterized by reduced trauma to the liver tissue and individual adaptation to the patient's health constitution. In softALPPS, portal vein embolization (PVE) is performed instead of portal vein ligation (PVL) after complete recovery of liver function. In addition, a non-absorbable foil was avoided in order to be able to extend the interval to step two or skip step two when required. RESULTS: Four patients successfully underwent softALPPS. Two of these patients have been followed-up for over a year (one patient with Klatskin tumor, one patient with extensive HCC). Both patients show no evidence of recurrence after 12 months and are in good medical condition. The other two patients who recently had surgery are also doing well. CONCLUSION: SoftALPPS offers the chance to curatively resect patients with high tumor burden of the liver even when the FLR is inadequate. This individual therapy method can give patients the possibility of complete tumor resection and can help to reduce perioperative morbidity.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/surgery , Hepatectomy , Humans , Ligation/methods , Liver/pathology , Portal Vein/pathology , Portal Vein/surgery , Treatment Outcome
6.
FASEB J ; 34(8): 10387-10397, 2020 08.
Article in English | MEDLINE | ID: mdl-32539223

ABSTRACT

TLR3 is implicated in anti-viral immune responses, but may also act as a sensor of tissue damage in the absence of infection. Here, we provide evidence for an essential role of TLR3 in liver regeneration after an acute loss of tissue due to partial hepatectomy. Mice lacking TLR3 had a severe and sustained defect in the restoration of liver tissue with reduced liver-to-body weight ratios even after an extended recovery period of 2 weeks. Hepatocyte cell cycle progression into S phase was impaired in TLR3-deficient mice. Mechanistic analyses revealed that TLR3-deficient mice had markedly reduced systemic levels of active HGF, but had increased amounts of inactive tissue-bound HGF. Importantly, expression of uPA, which orchestrates the processing and release of HGF from the hepatic extracellular matrix, was reduced in regenerating livers of TLR3-deficient mice. In addition, expression of the HGF maturation factor HGFAC was transiently diminished in TLR3-deficient mice. In vitro, engagement of TLR3 directly stimulated expression of uPA by hepatic stellate cells. Thus, TLR3 supports liver regeneration through upregulation of uPA, which promotes the release of preformed HGF from extracellular matrix stores.


Subject(s)
Cell Proliferation/physiology , Hepatocyte Growth Factor/metabolism , Hepatocytes/metabolism , Toll-Like Receptor 3/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Hepatectomy/methods , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/physiology , Liver/metabolism , Liver Regeneration/physiology , Male , Mice , Mice, Inbred C57BL , Organogenesis/physiology
7.
FASEB J ; 34(6): 8125-8138, 2020 06.
Article in English | MEDLINE | ID: mdl-32329113

ABSTRACT

The effectiveness of liver regeneration limits surgical therapies of hepatic disorders and determines patient outcome. Here, we investigated the role of the neuropeptide calcitonin gene-related peptide (CGRP) for liver regeneration after acute or chronic injury. Mice deficient for the CGRP receptor component receptor activity-modifying protein 1 (RAMP1) were subjected to a 70% partial hepatectomy or repeated intraperitoneal injections of carbon tetrachloride. RAMP1 deficiency severely impaired recovery of organ mass and hepatocyte proliferation after both acute and chronic liver injury. Mechanistically, protein expression of the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) was decreased in regenerating livers of RAMP1-deficient mice. Lack of RAMP1 was associated with hyperphosphorylation of YAP on Ser127 and Ser397, which regulates YAP functional activity and protein levels. Consequently, expression of various YAP-controlled cell cycle regulators and hepatocyte proliferation were severely reduced in the absence of RAMP1. In vitro, CGRP treatment caused increased YAP protein expression and a concomitant decline of YAP phosphorylation in liver tissue slice cultures of mouse and human origin and in primary human hepatocytes. Thus, our results indicate that sensory nerves represent a crucial control element of liver regeneration after acute and chronic injury acting through the CGRP-RAMP1 pathway, which stimulates YAP/TAZ expression and activity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Liver Regeneration/physiology , Receptor Activity-Modifying Protein 1/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Cell Cycle/physiology , Cell Proliferation/physiology , Hepatectomy/methods , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation/physiology , Signal Transduction/physiology , YAP-Signaling Proteins
8.
Int J Cancer ; 147(6): 1715-1731, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32147820

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a cell plasticity process required for metastasis and chemoresistance of carcinoma cells. We report a crucial role of the signal adaptor proteins CRK and CRKL in promoting EMT and tumor aggressiveness, as well as resistance against chemotherapy in colorectal and pancreatic carcinoma. Genetic loss of either CRKL or CRK partially counteracted EMT in three independent cancer cell lines. Strikingly, complete loss of the CRK family shifted cells strongly toward the epithelial phenotype. Cells exhibited greatly increased E-cadherin and grew as large, densely packed clusters, completely lacked invasiveness and the ability to undergo EMT induced by cytokines or genetic activation of SRC. Furthermore, CRK family-deficiency significantly reduced cell survival, proliferation and chemoresistance, as well as ERK1/2 phosphorylation and c-MYC protein levels. In accordance, MYC-target gene expression was identified as novel hallmark process positively regulated by CRK family proteins. Mechanistically, CRK proteins were identified as pivotal amplifiers of SRC/FAK signaling at focal adhesions, mediated through a novel positive feedback loop depending on RAP1. Expression of the CRK family and the EMT regulator ZEB1 was significantly correlated in samples from colorectal cancer patients, especially in invasive regions. Further, high expression of CRK family genes was significantly associated with reduced survival in locally advanced colorectal cancer, as well as in pan-cancer datasets from the TCGA project. Thus, CRK family adaptor proteins are promising therapeutic targets to counteract EMT, chemoresistance, metastasis formation and minimal residual disease. As proof of concept, CRK family-mediated oncogenic signaling was successfully inhibited by a peptide-based inhibitor.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-crk/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colon/pathology , Colon/surgery , Colorectal Neoplasms/therapy , Datasets as Topic , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition/drug effects , Female , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/pathology , Humans , Male , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-crk/antagonists & inhibitors , RNA-Seq , Rectum/pathology , Rectum/surgery , Signal Transduction/drug effects , Zinc Finger E-box-Binding Homeobox 1/metabolism , src-Family Kinases/metabolism
9.
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
10.
Gut ; 65(6): 1001-14, 2016 06.
Article in English | MEDLINE | ID: mdl-26762195

ABSTRACT

OBJECTIVE: The impact of glia cells during GI carcinogenesis and in cancer pain is unknown. Here, we demonstrate a novel mechanism how Schwann cells (SCs) become activated in the pancreatic cancer (PCa) microenvironment and influence spinal activity and pain sensation. DESIGN: Human SCs were exposed to hypoxia, to pancreatic cancer cells (PCCs) and/or to T-lymphocytes. Both SC and intrapancreatic nerves of patients with PCa with known pain severity were assessed for glial intermediate filament and hypoxia marker expression, proliferation and for transcriptional alterations of pain-related targets. In conditional PCa mouse models with selective in vivo blockade of interleukin (IL)-6 signalling (Ptf1a-Cre;LSL-Kras(G12D)/KC interbred with IL6(-/-) or sgp130(tg) mice), SC reactivity, abdominal mechanosensitivity and spinal glial/neuronal activity were quantified. RESULTS: Tumour hypoxia, PCC and/or T-lymphocytes activated SC via IL-6-signalling in vitro. Blockade of the IL-6-signalling suppressed SC activation around PCa precursor lesions (pancreatic intraepithelial neoplasia (PanIN)) in KC;IL6(-/-) (32.06%±5.25% of PanINs) and KC;sgp130(tg) (55.84%±5.51%) mouse models compared with KC mice (78.27%±3.91%). Activated SCs were associated with less pain in human PCa and with decreased abdominal mechanosensitivity in KC mice (von Frey score of KC: 3.9±0.5 vs KC;IL6(-/-) mice: 5.9±0.9; and KC;sgp130(tg): 10.21±1.4) parallel to attenuation of spinal astroglial and/or microglial activity. Activated SC exhibited a transcriptomic profile with anti-inflammatory and anti-nociceptive features. CONCLUSIONS: Activated SC in PCa recapitulate the hallmarks of 'reactive gliosis' and contribute to analgesia due to suppression of spinal glia. Our findings propose a mechanism for how cancer might remain pain-free via the SC-central glia interplay during cancer progression.


Subject(s)
Analgesia , Astrocytes , Microglia , Pancreatic Neoplasms/genetics , Schwann Cells/metabolism , Tumor Hypoxia/genetics , Animals , Astrocytes/metabolism , Disease Models, Animal , Humans , In Vitro Techniques , Interleukin-6/genetics , Mice , Mice, Transgenic , Microglia/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , T-Lymphocytes/metabolism
11.
J Immunol ; 188(9): 4590-601, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22450813

ABSTRACT

Recently, the mammalian actin-binding protein 1 (mAbp1; Hip-55, SH3P7, debrin-like protein) was identified as a novel component of the ß(2) integrin-mediated signaling cascade during complement-mediated phagocytosis and firm adhesion of polymorphonuclear neutrophils (PMN) under physiological shear stress conditions. In this study, we found that the genetic ablation of mAbp1 severely compromised not only the induction of adhesion, but also subsequent spreading of leukocytes to the endothelium as assessed by intravital microscopy of inflamed vessels of the cremaster muscle of mice. In vitro studies using murine PMN confirmed that mAbp1 was required for ß(2) integrin-mediated spreading under shear stress conditions, whereas mAbp1 was dispensable for spreading under static conditions. Upon ß(2) integrin-mediated adhesion and chemotactic migration of human neutrophil-like differentiated HL-60 cells, mAbp1 was enriched at the leading edge of the polarized cell. Total internal reflection fluorescence microscopy revealed that mAbp1 formed propagating waves toward the front of the lamellipodium, which are characteristic for dynamic reorganization of the cytoskeleton. Accordingly, binding of mAbp1 to actin was increased upon ß(2) integrin-mediated adhesion, as shown by coimmunoprecipitation experiments. However, chemotactic migration under static conditions was unaffected in the absence of mAbp1. In contrast, the downregulation of mAbp1 by RNA interference technique in neutrophil-like differentiated HL-60 cells or the genetic ablation of mAbp1 in leukocytes led to defective migration under flow conditions in vitro and in inflamed cremaster muscle venules in the situation in vivo. In conclusion, mAbp1 is of fundamental importance for spreading and migration under shear stress conditions, which are critical prerequisites for efficient PMN extravasation during inflammation.


Subject(s)
Cell Movement/immunology , Endothelium, Vascular/immunology , Microfilament Proteins/immunology , Neutrophils/immunology , Pseudopodia/immunology , src Homology Domains/immunology , Animals , CD18 Antigens/genetics , CD18 Antigens/immunology , CD18 Antigens/metabolism , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Movement/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , HL-60 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neutrophils/metabolism , Pseudopodia/genetics , Pseudopodia/metabolism , Pseudopodia/pathology , RNA Interference , Shear Strength , src Homology Domains/genetics
12.
J Clin Invest ; 133(21)2023 11 01.
Article in English | MEDLINE | ID: mdl-37607005

ABSTRACT

Solid cancers like pancreatic ductal adenocarcinoma (PDAC), a type of pancreatic cancer, frequently exploit nerves for rapid dissemination. This neural invasion (NI) is an independent prognostic factor in PDAC, but insufficiently modeled in genetically engineered mouse models (GEMM) of PDAC. Here, we systematically screened for human-like NI in Europe's largest repository of GEMM of PDAC, comprising 295 different genotypes. This phenotype screen uncovered 2 GEMMs of PDAC with human-like NI, which are both characterized by pancreas-specific overexpression of transforming growth factor α (TGF-α) and conditional depletion of p53. Mechanistically, cancer-cell-derived TGF-α upregulated CCL2 secretion from sensory neurons, which induced hyperphosphorylation of the cytoskeletal protein paxillin via CCR4 on cancer cells. This activated the cancer migration machinery and filopodia formation toward neurons. Disrupting CCR4 or paxillin activity limited NI and dampened tumor size and tumor innervation. In human PDAC, phospho-paxillin and TGF-α-expression constituted strong prognostic factors. Therefore, we believe that the TGF-α-CCL2-CCR4-p-paxillin axis is a clinically actionable target for constraining NI and tumor progression in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Animals , Mice , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism , Paxillin/genetics , Paxillin/metabolism , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/metabolism , Phenotype , Cell Line, Tumor , Pancreatic Neoplasms
13.
Blood ; 116(11): 1885-94, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20530790

ABSTRACT

A key event in the successful induction of adaptive immune responses is the antigen-specific activation of T cells by dendritic cells (DCs). Although LFA-1 (lymphocyte function-associated antigen 1) on T cells is considered to be important for antigen-specific T-cell activation, the role for LFA-1 on DCs remains elusive. Using 2 different approaches to activate LFA-1 on DCs, either by deletion of the αL-integrin cytoplasmic GFFKR sequence or by silencing cytohesin-1-interacting protein, we now provide evidence that DCs are able to make use of active LFA-1 and can thereby control the contact duration with naive T cells. Enhanced duration of DC/T-cell interaction correlates inversely with antigen-specific T-cell proliferation, generation of T-helper 1 cells, and immune responses leading to delayed-type hypersensitivity. We could revert normal interaction time and T-cell proliferation to wild-type levels by inhibition of active LFA-1 on DCs. Our data further suggest that cytohesin-1-interacting protein might be responsible for controlling LFA-1 deactivation on mature DCs. In summary, our findings indicate that LFA-1 on DCs needs to be in an inactive state to ensure optimal T-cell activation and suggest that regulation of LFA-1 activity allows DCs to actively control antigen-driven T-cell proliferation and effective immune responses.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Lymphocyte Function-Associated Antigen-1/immunology , T-Lymphocytes/immunology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion/immunology , Cell Proliferation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Flow Cytometry , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/metabolism , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA Interference , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Time Factors
14.
JHEP Rep ; 4(5): 100465, 2022 May.
Article in English | MEDLINE | ID: mdl-35462860

ABSTRACT

Background & Aims: Increased sensitivity towards tumor necrosis factor (TNF)-induced cell death in virus-infected hepatocytes has revealed a so far unrecognized hepatocyte-intrinsic antiviral immune surveillance mechanism, for which no in vitro or ex vivo model is available. We aimed to establish precision-cut liver slices (PCLS) as a model system to study hepatocyte-intrinsic regulation of apoptosis. Methods: Preparation of PCLS from mouse and human liver tissue was optimized for minimal procedure-associated apoptosis. Functionality of liver cells in PCLS was characterized using extracellular flux analysis to determine mitochondrial respiration, and viral infection with recombinant adenovirus and lymphocytic choriomeningitis virus (LCMV) was used to probe for hepatocyte-intrinsic sensitivity towards apoptosis in PCLS. Apoptosis was detected by immunohistochemical staining for cleaved-caspase 3 and quantified by detection of effector caspase activity in PCLS. Results: We established an optimized protocol for preparation of PCLS from human and mouse models using agarose-embedding of liver tissue to improve precision cutting and using organ-protective buffer solutions to minimize procedure-associated cell death. PCLS prepared from virus-infected livers showed preserved functional metabolic properties. Importantly, in PCLS from adenovirus- and LCMV-infected livers we detected increased induction of apoptosis after TNF challenge ex vivo. Conclusion: We conclude that PCLS can be used as model system to ex vivo characterize hepatocyte-intrinsic sensitivity to cell death. This may also enable researchers to characterize human hepatocyte sensitivity to apoptosis in PCLS prepared from patients with acute or chronic liver diseases. Lay summary: Virus-infected hepatocytes in vivo show an increased sensitivity towards induction of cell death signaling through the TNF receptor. Studying this hepatocyte-intrinsic antiviral immune surveillance mechanism has been hampered by the absence of model systems that reciprocate the in vivo finding of increased apoptosis of virus-infected hepatocytes challenged with TNF. Herein, we report that an optimized protocol for generation of precision-cut liver slices can be used to study this hepatocyte-intrinsic surveillance mechanism ex vivo.

15.
J Exp Med ; 201(12): 1987-98, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15955836

ABSTRACT

The dynamic regulation of ligand binding is considered crucial for integrin function. However, the importance of activity regulation for integrin function in vivo is largely unknown. Here, we have applied gene targeting to delete the GFFKR sequence of the lymphocyte function-associated antigen-1 (LFA-1) alpha(L) subunit cytoplasmic domain in mouse germline. Lymphocytes from Lfa-1(d/d) mutant mice showed constitutive activation of LFA-1-mediated cell adhesion and impaired de-adhesion from intercellular adhesion molecule-1 that resulted in defective cell migration. In contrast, signaling through LFA-1 was not affected in Lfa-1(d/d) cells. T cell activation by superantigen-loaded and allogeneic APCs, cytotoxic T cell activity, T-dependent humoral immune responses, and neutrophil recruitment during aseptic peritonitis were impaired in Lfa-1(d/d) mice. Thus, deactivation of LFA-1 and disassembly of LFA-1-mediated cell contacts seem to be vital for the generation of normal immune responses.


Subject(s)
Antibody Formation/immunology , Cell Adhesion/immunology , Immunity, Cellular/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Protein Subunits/metabolism , Animals , Antibodies/immunology , Chromosomes, Artificial, Bacterial , DNA Primers , Gene Targeting , Humans , Intercellular Adhesion Molecule-1/metabolism , Lymphatic System/physiology , Lymphocyte Function-Associated Antigen-1/genetics , Mice , Microscopy, Video , Mutation/genetics , Organogenesis/genetics , Organogenesis/physiology , Polymerase Chain Reaction , Protein Structure, Tertiary , Protein Subunits/immunology
16.
Int Immunol ; 22(1): 35-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19946016

ABSTRACT

Leucocyte function-associated antigen-1 (LFA-1) is known to be involved in immune reactions leading to allograft rejection. The role of deactivating LFA-1 in this context has not been investigated yet, although it is accepted that regulating LFA-1 activity is essential for T-cell function. Expressing LFA-1 locked in an active state in mice (LFA-1(d/d)) allowed us to investigate the in vivo function of LFA-1 deactivation for allograft rejection in a model of heterotopic cardiac transplantation. We provide in vivo evidence that regulating LFA-1 activity from an active to an inactive state controls antigen-specific priming and proliferation of T cells in response to allogeneic stimuli. Consequently, defective LFA-1 deactivation significantly prolonged cardiac allograft survival. Furthermore, reduced numbers of alloantigen-specific T cells and non-allo-specific innate immune cells within allografts of LFA-1(d/d) recipients indicate that expression of active LFA-1 impairs inflammatory responses involving all major leucocyte subpopulations. Taken together, our in vivo data suggest that LFA-1 deactivation is important for the formation of inflammatory lesions and rejection of cardiac allografts. Thus, the dynamic regulation of LFA-1 activity, rather than the mere presence of LFA-1, appears to contribute to the control of immune reactions inducing allogeneic transplant rejection.


Subject(s)
Graft Rejection/immunology , Heart Transplantation , Lymphocyte Function-Associated Antigen-1/metabolism , T-Lymphocytes/metabolism , Animals , Cell Movement/genetics , Cell Movement/immunology , Cell Proliferation , Graft Rejection/genetics , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival/genetics , Graft Survival/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/pathology
17.
BMC Cancer ; 9: 463, 2009 Dec 26.
Article in English | MEDLINE | ID: mdl-20035626

ABSTRACT

BACKGROUND: Costimulatory signaling has been implicated as a potential regulator of antitumor immunity in various human cancers. In contrast to the negative prognostic value of aberrant B7-H1 expression by pancreatic cancer cells, the role of B7-H3 is still unknown. Therefore, we investigated the expression pattern and clinical significance of B7-H3 expression in human pancreatic cancer. METHODS: B7-H3 expression was evaluated by immunohistochemistry in 68 patients with pancreatic cancer who underwent surgical tumor resection. Expression data was correlated with clinicopathologic features and with the number of tumor-infiltrating T cells. RESULTS: B7-H3 expression was significantly upregulated in pancreatic cancer compared to normal pancreas (p < 0.05). In 60 of 68 examined tumors B7-H3 protein was detectable in pancreatic cancer cells. Patients with high tumor B7-H3 levels had a significantly better postoperative prognosis than patients with low tumor B7-H3 levels (p = 0.0067). Furthermore, tumor B7-H3 expression significantly correlated with the number of tumor-infiltrating CD8+ T cells (p = 0.018). CONCLUSION: We demonstrate for the first time that B7-H3 is abundantly expressed in pancreatic cancer and that tumor-associated B7-H3 expression significantly correlates with prolonged postoperative survival. Our findings suggest that B7-H3 might play an important role as a potential stimulator of antitumor immune response in pancreatic cancer.


Subject(s)
Antigens, CD/genetics , Carcinoma/mortality , Pancreatic Neoplasms/mortality , Receptors, Immunologic/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , B7 Antigens , Carcinoma/diagnosis , Carcinoma/genetics , Carcinoma/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Receptors, Immunologic/metabolism , Survival Analysis , Tumor Cells, Cultured , Young Adult
18.
Hum Gene Ther ; 30(1): 44-56, 2019 01.
Article in English | MEDLINE | ID: mdl-29916265

ABSTRACT

Muscle-invasive bladder cancer represents approximately 25% of diagnosed bladder cancer cases and carries a significant risk of death. Oncolytic viruses are novel antitumor agents with the ability to selectively replicate and lyse tumor cells while sparing healthy tissue. We explored the efficiency of the oncolytic YB-1-selective adenovirus XVir-N-31 in vitro and in an orthotopic mouse model for bladder cancer by intramural injection under ultrasound guidance. We demonstrated that XVir-N-31 replicated in bladder cancer cells and induced a stronger immunogenic cell death than wild-type adenovirus by facilitating enhanced release of HMGB1 and exosomal Hsp70. The intratumoral delivery of XVir-N-31 by ultrasound guidance delayed tumor growth in an immunodeficient model, demonstrating the feasibility of this approach to deliver oncolytic viruses directly into the tumor.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Genetic Vectors/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Animals , Cell Death/genetics , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Mice , Oncolytic Virotherapy/methods , Transgenes , Tumor Burden , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
19.
Cancer Lett ; 268(1): 98-109, 2008 Sep 08.
Article in English | MEDLINE | ID: mdl-18486325

ABSTRACT

We investigated the expression pattern and clinical significance of the costimulatory ligands B7-1, B7-2, B7-H1, and B7-DC, and their counter-receptors CTLA-4 and PD-1 in pancreatic cancer. Gene expression of all examined costimulatory molecules was significantly upregulated in pancreatic cancer tissues. B7-1, B7-2, B7-H1, and B7-DC protein was detectable in pancreatic cancer cells. Only the expression of B7-H1 significantly correlated with postoperative survival (p<0.0001). B7-H1 was inducible in cultured pancreatic cancer cells by IFN-gamma and significantly correlated with the level of IFN-gamma expression in human pancreatic cancer tissues (Spearman rho=0.4536,p=0.0029). B7-H1 positive tumors showed an increased prevalence of tumor-infiltrating regulatory T cells (Tregs) compared to B7-H1 negative tumors. Among the investigated costimulatory molecules only tumor-associated B7-H1 seems to be of prognostic relevance in pancreatic cancer. B7-H1 might, therefore, be involved in the downregulation of antitumor responses through regulation of Tregs in pancreatic cancer. Our findings also suggest a dual role of IFN-gamma in antitumor response. Through induction of B7-H1 in pancreatic cancer cells IFN-gamma might contribute to the evasion of antitumor immunity.


Subject(s)
B7-1 Antigen/metabolism , Interferon-gamma/metabolism , Pancreatic Neoplasms/metabolism , Antigens, CD/metabolism , B7-2 Antigen/metabolism , CTLA-4 Antigen , Cell Line, Tumor , Cytokines/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Programmed Cell Death 1 Ligand 2 Protein , Survival Analysis , T-Lymphocytes, Regulatory/immunology , Up-Regulation
20.
Sci Rep ; 8(1): 12271, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30115978

ABSTRACT

Stimulation of cytosolic nucleic acid sensors of innate immunity by pathogen-derived nucleic acids is important for antimicrobial defence, but stimulation through self-derived nucleic acids may contribute to autoinflammation and cancer. DNA sensing in the cytosol requires the stimulator of interferon genes (STING), while cytosolic RNA sensors use mitochondrial antiviral-signalling protein (MAVS). In a murine model of two-thirds hepatectomy, combined deficiency of MAVS and STING resulted in strongly impaired hepatocyte proliferation and delayed recovery of liver mass. Whereas lack of MAVS and STING did not influence upregulation of the G1-phase cyclins D1 and E1, it substantially reduced the hyperphosphorylation of retinoblastoma protein, attenuated the activation of cyclin-dependent kinase (CDK)-2, delayed upregulation of CDK1 and cyclins A2 and B1, and impaired S-phase entry of hepatocytes. Mechanistically, lack of cytosolic nucleic acid sensors strongly upregulated the anti-proliferative mediators TGF-ß2 and activin A, which was associated with an increased expression of the cell cycle inhibitors p15 and p21. Partial hepatectomy was followed by the release of exosomes with abundant nucleic acid cargo, which may provide ligands for the MAVS and STING pathways. Together, these findings identify a previously unrecognised function of cytosolic nucleic acid sensors of innate immunity for promoting liver regeneration.


Subject(s)
Cytosol/metabolism , DNA/metabolism , Hepatectomy , Immunity, Innate , Liver Regeneration/immunology , Adaptor Proteins, Signal Transducing/deficiency , Animals , Cell Cycle , Cell Proliferation , Hepatocytes/cytology , Hepatocytes/metabolism , Interleukin-6/biosynthesis , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , Signal Transduction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL