Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Hepatol ; 79(2): 296-313, 2023 08.
Article in English | MEDLINE | ID: mdl-37224925

ABSTRACT

BACKGROUND & AIMS: The progression of non-alcoholic steatohepatitis (NASH) to fibrosis and hepatocellular carcinoma (HCC) is aggravated by auto-aggressive T cells. The gut-liver axis contributes to NASH, but the mechanisms involved and the consequences for NASH-induced fibrosis and liver cancer remain unknown. We investigated the role of gastrointestinal B cells in the development of NASH, fibrosis and NASH-induced HCC. METHODS: C57BL/6J wild-type (WT), B cell-deficient and different immunoglobulin-deficient or transgenic mice were fed distinct NASH-inducing diets or standard chow for 6 or 12 months, whereafter NASH, fibrosis, and NASH-induced HCC were assessed and analysed. Specific pathogen-free/germ-free WT and µMT mice (containing B cells only in the gastrointestinal tract) were fed a choline-deficient high-fat diet, and treated with an anti-CD20 antibody, whereafter NASH and fibrosis were assessed. Tissue biopsy samples from patients with simple steatosis, NASH and cirrhosis were analysed to correlate the secretion of immunoglobulins to clinicopathological features. Flow cytometry, immunohistochemistry and single-cell RNA-sequencing analysis were performed in liver and gastrointestinal tissue to characterise immune cells in mice and humans. RESULTS: Activated intestinal B cells were increased in mouse and human NASH samples and licensed metabolic T-cell activation to induce NASH independently of antigen specificity and gut microbiota. Genetic or therapeutic depletion of systemic or gastrointestinal B cells prevented or reverted NASH and liver fibrosis. IgA secretion was necessary for fibrosis induction by activating CD11b+CCR2+F4/80+CD11c-FCGR1+ hepatic myeloid cells through an IgA-FcR signalling axis. Similarly, patients with NASH had increased numbers of activated intestinal B cells; additionally, we observed a positive correlation between IgA levels and activated FcRg+ hepatic myeloid cells, as well the extent of liver fibrosis. CONCLUSIONS: Intestinal B cells and the IgA-FcR signalling axis represent potential therapeutic targets for the treatment of NASH. IMPACT AND IMPLICATIONS: There is currently no effective treatment for non-alcoholic steatohepatitis (NASH), which is associated with a substantial healthcare burden and is a growing risk factor for hepatocellular carcinoma (HCC). We have previously shown that NASH is an auto-aggressive condition aggravated, amongst others, by T cells. Therefore, we hypothesized that B cells might have a role in disease induction and progression. Our present work highlights that B cells have a dual role in NASH pathogenesis, being implicated in the activation of auto-aggressive T cells and the development of fibrosis via activation of monocyte-derived macrophages by secreted immunoglobulins (e.g., IgA). Furthermore, we show that the absence of B cells prevented HCC development. B cell-intrinsic signalling pathways, secreted immunoglobulins, and interactions of B cells with other immune cells are potential targets for combinatorial NASH therapies against inflammation and fibrosis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Microbiota , Non-alcoholic Fatty Liver Disease , Humans , Mice , Animals , Non-alcoholic Fatty Liver Disease/complications , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Mice, Inbred C57BL , Liver/pathology , Fibrosis , Liver Cirrhosis/complications , Mice, Transgenic , Immunoglobulin A/metabolism , Immunoglobulin A/pharmacology , Disease Models, Animal , Diet, High-Fat/adverse effects
3.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article in English | MEDLINE | ID: mdl-33798093

ABSTRACT

The c-Jun N-terminal kinase (JNK) signaling pathway mediates adaptation to stress signals and has been associated with cell death, cell proliferation, and malignant transformation in the liver. However, up to now, its function was experimentally studied mainly in young mice. By generating mice with combined conditional ablation of Jnk1 and Jnk2 in liver parenchymal cells (LPCs) (JNK1/2LPC-KO mice; KO, knockout), we unraveled a function of the JNK pathway in the regulation of liver homeostasis during aging. Aging JNK1/2LPC-KO mice spontaneously developed large biliary cysts that originated from the biliary cell compartment. Mechanistically, we could show that cyst formation in livers of JNK1/2LPC-KO mice was dependent on receptor-interacting protein kinase 1 (RIPK1), a known regulator of cell survival, apoptosis, and necroptosis. In line with this, we showed that RIPK1 was overexpressed in the human cyst epithelium of a subset of patients with polycystic liver disease. Collectively, these data reveal a functional interaction between JNK signaling and RIPK1 in age-related progressive cyst development. Thus, they provide a functional linkage between stress adaptation and programmed cell death (PCD) in the maintenance of liver homeostasis during aging.


Subject(s)
Aging/metabolism , Bile Duct Diseases/etiology , Bile Duct Diseases/metabolism , Caspase 8/metabolism , Cysts/etiology , Cysts/metabolism , MAP Kinase Signaling System , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Apoptosis , Biopsy , Disease Models, Animal , Disease Susceptibility , Immunohistochemistry , Immunophenotyping , Liver Diseases/etiology , Liver Diseases/metabolism , Mice , Mitogen-Activated Protein Kinase 8/deficiency , Necroptosis
4.
Gastroenterology ; 159(1): 183-199, 2020 07.
Article in English | MEDLINE | ID: mdl-32179094

ABSTRACT

BACKGROUND & AIMS: Intestinal epithelial homeostasis depends on a tightly regulated balance between intestinal epithelial cell (IEC) death and proliferation. While the disruption of several IEC death regulating factors result in intestinal inflammation, the loss of the anti-apoptotic BCL2 family members BCL2 and BCL2L1 has no effect on intestinal homeostasis in mice. We investigated the functions of the antiapoptotic protein MCL1, another member of the BCL2 family, in intestinal homeostasis in mice. METHODS: We generated mice with IEC-specific disruption of Mcl1 (Mcl1ΔIEC mice) or tamoxifen-inducible IEC-specific disruption of Mcl1 (i-Mcl1ΔIEC mice); these mice and mice with full-length Mcl1 (controls) were raised under normal or germ-free conditions. Mice were analyzed by endoscopy and for intestinal epithelial barrier permeability. Intestinal tissues were analyzed by histology, in situ hybridization, proliferation assays, and immunoblots. Levels of calprotectin, a marker of intestinal inflammation, were measured in intestinal tissues and feces. RESULTS: Mcl1ΔIEC mice spontaneously developed apoptotic enterocolopathy, characterized by increased IEC apoptosis, hyperproliferative crypts, epithelial barrier dysfunction, and chronic inflammation. Loss of MCL1 retained intestinal crypts in a hyperproliferated state and prevented the differentiation of intestinal stem cells. Proliferation of intestinal stem cells in MCL1-deficient mice required WNT signaling and was associated with DNA damage accumulation. By 1 year of age, Mcl1ΔIEC mice developed intestinal tumors with morphologic and genetic features of human adenomas and carcinomas. Germ-free housing of Mcl1ΔIEC mice reduced markers of microbiota-induced intestinal inflammation but not tumor development. CONCLUSION: The antiapoptotic protein MCL1, a member of the BCL2 family, is required for maintenance of intestinal homeostasis and prevention of carcinogenesis in mice. Loss of MCL1 results in development of intestinal carcinomas, even under germ-free conditions, and therefore does not involve microbe-induced chronic inflammation. Mcl1ΔIEC mice might be used to study apoptotic enterocolopathy and inflammatory bowel diseases.


Subject(s)
Carcinoma/pathology , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinoma/diagnosis , Carcinoma/genetics , Disease Models, Animal , Endoscopy , Epithelial Cells/pathology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/diagnostic imaging , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/genetics , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/genetics
6.
Cell ; 180(1): 50-63.e12, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31923399

ABSTRACT

Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.


Subject(s)
Immunity, Mucosal/immunology , Interleukin-18/immunology , Intestinal Mucosa/immunology , Animals , Cytokines/immunology , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Epithelial Cells/immunology , Female , Goblet Cells/immunology , Interleukin-18/biosynthesis , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Male , Mice , Mice, Inbred C57BL , Neurons/immunology , Rats , Rats, Sprague-Dawley , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology
7.
Nat Med ; 25(4): 641-655, 2019 04.
Article in English | MEDLINE | ID: mdl-30936549

ABSTRACT

Non-alcoholic fatty liver disease ranges from steatosis to non-alcoholic steatohepatitis (NASH), potentially progressing to cirrhosis and hepatocellular carcinoma (HCC). Here, we show that platelet number, platelet activation and platelet aggregation are increased in NASH but not in steatosis or insulin resistance. Antiplatelet therapy (APT; aspirin/clopidogrel, ticagrelor) but not nonsteroidal anti-inflammatory drug (NSAID) treatment with sulindac prevented NASH and subsequent HCC development. Intravital microscopy showed that liver colonization by platelets depended primarily on Kupffer cells at early and late stages of NASH, involving hyaluronan-CD44 binding. APT reduced intrahepatic platelet accumulation and the frequency of platelet-immune cell interaction, thereby limiting hepatic immune cell trafficking. Consequently, intrahepatic cytokine and chemokine release, macrovesicular steatosis and liver damage were attenuated. Platelet cargo, platelet adhesion and platelet activation but not platelet aggregation were identified as pivotal for NASH and subsequent hepatocarcinogenesis. In particular, platelet-derived GPIbα proved critical for development of NASH and subsequent HCC, independent of its reported cognate ligands vWF, P-selectin or Mac-1, offering a potential target against NASH.


Subject(s)
Blood Platelets/metabolism , Liver Neoplasms/blood , Liver Neoplasms/drug therapy , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/drug therapy , Platelet Glycoprotein GPIb-IX Complex/metabolism , Animals , Blood Platelets/drug effects , Body Weight/drug effects , Cytokines/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Endothelium/drug effects , Endothelium/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice, Transgenic , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Count
8.
J Pathol ; 248(3): 377-389, 2019 07.
Article in English | MEDLINE | ID: mdl-30883740

ABSTRACT

Recent studies suggest that clear cell renal cell carcinoma (ccRCC) possesses a rare population of cancer stem cells (CSCs) that might contribute to tumor heterogeneity, metastasis and therapeutic resistance. Nevertheless, their relevance for renal cancer is still unclear. In this study, we successfully isolated CSCs from established human ccRCC cell lines. CSCs displayed high expression of the chemokine IL-8 and its receptor CXCR1. While recombinant IL-8 significantly increased CSC number and properties in vitro, CXCR1 inhibition using an anti-CXCR1 antibody or repertaxin significantly reduced these features. After injection into immune-deficient mice, CSCs formed primary tumors that metastasized to the lung and liver. All xenografted tumors in mice expressed high levels of IL-8 and CXCR1. Furthermore, IL-8/CXCR1 expression significantly correlated with decreased overall survival in ccRCC patients. These results suggest that the IL-8/CXCR1 phenotype is associated with CSC-like properties in renal cancer. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma, Renal Cell/genetics , Interleukin-8/genetics , Kidney Neoplasms/genetics , Neoplastic Stem Cells/pathology , Receptors, Interleukin-8A/genetics , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/genetics , Humans , Kidney Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Receptors, Interleukin-8B/antagonists & inhibitors
9.
Cancer Cell ; 32(3): 342-359.e10, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28898696

ABSTRACT

Concomitant hepatocyte apoptosis and regeneration is a hallmark of chronic liver diseases (CLDs) predisposing to hepatocellular carcinoma (HCC). Here, we mechanistically link caspase-8-dependent apoptosis to HCC development via proliferation- and replication-associated DNA damage. Proliferation-associated replication stress, DNA damage, and genetic instability are detectable in CLDs before any neoplastic changes occur. Accumulated levels of hepatocyte apoptosis determine and predict subsequent hepatocarcinogenesis. Proliferation-associated DNA damage is sensed by a complex comprising caspase-8, FADD, c-FLIP, and a kinase-dependent function of RIPK1. This platform requires a non-apoptotic function of caspase-8, but no caspase-3 or caspase-8 cleavage. It may represent a DNA damage-sensing mechanism in hepatocytes that can act via JNK and subsequent phosphorylation of the histone variant H2AX.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Caspase 8/metabolism , DNA Damage , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Cellular Senescence , Chronic Disease , Crosses, Genetic , DNA Repair , Fas-Associated Death Domain Protein/metabolism , Female , Genomic Instability , Hepatectomy , Hepatocytes/pathology , Histones/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/metabolism , Liver/pathology , Liver Regeneration , Male , Mice , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Risk Factors
10.
Cell ; 167(3): 843-857.e14, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27720451

ABSTRACT

Glucagon and thyroid hormone (T3) exhibit therapeutic potential for metabolic disease but also exhibit undesired effects. We achieved synergistic effects of these two hormones and mitigation of their adverse effects by engineering chemical conjugates enabling delivery of both activities within one precisely targeted molecule. Coordinated glucagon and T3 actions synergize to correct hyperlipidemia, steatohepatitis, atherosclerosis, glucose intolerance, and obesity in metabolically compromised mice. We demonstrate that each hormonal constituent mutually enriches cellular processes in hepatocytes and adipocytes via enhanced hepatic cholesterol metabolism and white fat browning. Synchronized signaling driven by glucagon and T3 reciprocally minimizes the inherent harmful effects of each hormone. Liver-directed T3 action offsets the diabetogenic liability of glucagon, and glucagon-mediated delivery spares the cardiovascular system from adverse T3 action. Our findings support the therapeutic utility of integrating these hormones into a single molecular entity that offers unique potential for treatment of obesity, type 2 diabetes, and cardiovascular disease.


Subject(s)
Glucagon/therapeutic use , Metabolic Diseases/drug therapy , Triiodothyronine/drug effects , Animals , Atherosclerosis/drug therapy , Body Weight/drug effects , Bone and Bones/drug effects , Chemical Engineering/methods , Cholesterol/metabolism , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Drug Combinations , Drug Delivery Systems , Drug Synergism , Glucagon/adverse effects , Glucagon/chemistry , Glucagon/pharmacology , Hyperglycemia/drug therapy , Liver/drug effects , Liver/metabolism , Mice , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Triiodothyronine/adverse effects , Triiodothyronine/chemistry , Triiodothyronine/pharmacology
11.
Infect Immun ; 84(5): 1424-1437, 2016 05.
Article in English | MEDLINE | ID: mdl-26902727

ABSTRACT

Members of the Burkholderia cepacia complex (Bcc) cause chronic opportunistic lung infections in people with cystic fibrosis (CF), resulting in a gradual lung function decline and, ultimately, patient death. The Bcc is a complex of 20 species and is rarely eradicated once a patient is colonized; therefore, vaccination may represent a better therapeutic option. We developed a new proteomics approach to identify bacterial proteins that are involved in the attachment of Bcc bacteria to lung epithelial cells. Fourteen proteins were reproducibly identified by two-dimensional gel electrophoresis from four Bcc strains representative of two Bcc species: Burkholderia cenocepacia, the most virulent, and B. multivorans, the most frequently acquired. Seven proteins were identified in both species, but only two were common to all four strains, linocin and OmpW. Both proteins were selected based on previously reported data on these proteins in other species. Escherichia coli strains expressing recombinant linocin and OmpW showed enhanced attachment (4.2- and 3.9-fold) to lung cells compared to the control, confirming that both proteins are involved in host cell attachment. Immunoproteomic analysis using serum from Bcc-colonized CF patients confirmed that both proteins elicit potent humoral responses in vivo Mice immunized with either recombinant linocin or OmpW were protected from B. cenocepacia and B. multivorans challenge. Both antigens induced potent antigen-specific antibody responses and stimulated strong cytokine responses. In conclusion, our approach identified adhesins that induced excellent protection against two Bcc species and are promising vaccine candidates for a multisubunit vaccine. Furthermore, this study highlights the potential of our proteomics approach to identify potent antigens against other difficult pathogens.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , Bacterial Outer Membrane Proteins/metabolism , Bacteriocins/metabolism , Burkholderia Infections/prevention & control , Burkholderia cepacia complex/physiology , Epithelial Cells/microbiology , Adhesins, Bacterial/immunology , Animals , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Bacteriocins/immunology , Burkholderia Infections/immunology , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/physiology , Female , Gene Expression , Humans , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Treatment Outcome
12.
Stem Cells Dev ; 24(20): 2391-402, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26076727

ABSTRACT

The immune suppressive and anti-inflammatory capabilities of bone marrow-derived mesenchymal stromal cells (MSCs) represent an innovative new tool in regenerative medicine and immune regulation. The potent immune suppressive ability of MSC over T cells, dendritic cells, and natural killer cells has been extensively characterized, however, the effect of MSC on B cell function has not yet been clarified. In this study, the direct effect of MSC on peripheral blood B cell function is defined and the mechanism utilized by MSC in enhancing B cell survival in vitro identified. Human MSC supported the activation, proliferation, and survival of purified CD19(+) B cells through a cell contact-dependent mechanism. These effects were not mediated through B cell activating factor or notch signaling. However, cell contact between MSC and B cells resulted in increased production of vascular endothelial growth factor (VEGF) by MSC facilitating AKT phosphorylation within the B cell and inhibiting caspase 3-mediated apoptosis. Blocking studies demonstrated that this cell contact-dependent effect was not dependent on signaling through CXCR4-CXCL12 or through the epidermal growth factor receptor (EGFR). These results suggest that direct cell contact between MSC and B cells supports B cell viability and function, suggesting that MSC may not represent a suitable therapy for B cell-mediated disease.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/cytology , Caspase 3/metabolism , Mesenchymal Stem Cells/cytology , Vascular Endothelial Growth Factor A/metabolism , Antigens, CD19/immunology , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Cell Proliferation/physiology , Humans , Killer Cells, Natural/immunology , Mesenchymal Stem Cell Transplantation , T-Lymphocytes/cytology , Transcriptional Activation/immunology , Up-Regulation
13.
Nat Immunol ; 14(9): 927-36, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23892723

ABSTRACT

Mutations that result in loss of function of Nod2, an intracellular receptor for bacterial peptidoglycan, are associated with Crohn's disease. Here we found that the E3 ubiquitin ligase Pellino3 was an important mediator in the Nod2 signaling pathway. Pellino3-deficient mice had less induction of cytokines after engagement of Nod2 and had exacerbated disease in various experimental models of colitis. Furthermore, expression of Pellino3 was lower in the colons of patients with Crohn's disease. Pellino3 directly bound to the kinase RIP2 and catalyzed its ubiquitination. Loss of Pellino3 led to attenuation of Nod2-induced ubiquitination of RIP2 and less activation of the transcription factor NF-κB and mitogen-activated protein kinases (MAPKs). Our findings identify RIP2 as a substrate for Pellino3 and Pellino3 as an important mediator in the Nod2 pathway and regulator of intestinal inflammation.


Subject(s)
Colitis/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Citrobacter rodentium/immunology , Colitis/genetics , Colitis/immunology , Colitis/microbiology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Male , Mice , Mice, Knockout , Middle Aged , Protein Binding , Protein Interaction Domains and Motifs , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...