Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Nat Immunol ; 20(12): 1668-1680, 2019 12.
Article in English | MEDLINE | ID: mdl-31636464

ABSTRACT

Lymph node fibroblastic reticular cells (FRCs) respond to signals from activated T cells by releasing nitric oxide, which inhibits T cell proliferation and restricts the size of the expanding T cell pool. Whether interactions with FRCs also support the function or differentiation of activated CD8+ T cells is not known. Here we report that encounters with FRCs enhanced cytokine production and remodeled chromatin accessibility in newly activated CD8+ T cells via interleukin-6. These epigenetic changes facilitated metabolic reprogramming and amplified the activity of pro-survival pathways through differential transcription factor activity. Accordingly, FRC conditioning significantly enhanced the persistence of virus-specific CD8+ T cells in vivo and augmented their differentiation into tissue-resident memory T cells. Our study demonstrates that FRCs play a role beyond restricting T cell expansion-they can also shape the fate and function of CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Fibroblasts/physiology , Lymph Nodes/immunology , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Cellular Reprogramming , Chromatin Assembly and Disassembly , Cytotoxicity, Immunologic , Epigenesis, Genetic , Gene Expression Regulation , Immunologic Memory , Interleukin-6/genetics , Interleukin-6/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism
2.
Nat Immunol ; 13(5): 499-510, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22466668

ABSTRACT

Lymph node stromal cells (LNSCs) closely regulate immunity and self-tolerance, yet key aspects of their biology remain poorly elucidated. Here, comparative transcriptomic analyses of mouse LNSC subsets demonstrated the expression of important immune mediators, growth factors and previously unknown structural components. Pairwise analyses of ligands and cognate receptors across hematopoietic and stromal subsets suggested a complex web of crosstalk. Fibroblastic reticular cells (FRCs) showed enrichment for higher expression of genes relevant to cytokine signaling, relative to their expression in skin and thymic fibroblasts. LNSCs from inflamed lymph nodes upregulated expression of genes encoding chemokines and molecules involved in the acute-phase response and the antigen-processing and antigen-presentation machinery. Poorly studied podoplanin (gp38)-negative CD31(-) LNSCs showed similarities to FRCs but lacked expression of interleukin 7 (IL-7) and were identified as myofibroblastic pericytes that expressed integrin α(7). Together our data comprehensively describe the transcriptional characteristics of LNSC subsets.


Subject(s)
Gene Expression/immunology , Inflammation/immunology , Lymph Nodes/immunology , Stromal Cells/immunology , Stromal Cells/metabolism , Transcriptome , Acute-Phase Reaction/immunology , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Cytokines/immunology , Cytokines/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Homeostasis/immunology , Inflammation/genetics , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Interleukin-7/immunology , Interleukin-7/metabolism , Lymph Nodes/cytology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Pericytes/immunology , Pericytes/metabolism , Self Tolerance/immunology , Tissue Array Analysis/methods
3.
Nat Immunol ; 12(11): 1096-104, 2011 Sep 18.
Article in English | MEDLINE | ID: mdl-21926986

ABSTRACT

Fibroblastic reticular cells (FRCs) and lymphatic endothelial cells (LECs) are nonhematopoietic stromal cells of lymphoid organs. They influence the migration and homeostasis of naive T cells; however, their influence on activated T cells remains undescribed. Here we report that FRCs and LECs inhibited T cell proliferation through a tightly regulated mechanism dependent on nitric oxide synthase 2 (NOS2). Expression of NOS2 and production of nitric oxide paralleled the activation of T cells and required a tripartite synergism of interferon-γ, tumor necrosis factor and direct contact with activated T cells. Notably, in vivo expression of NOS2 by FRCs and LECs regulated the size of the activated T cell pool. Our study elucidates an as-yet-unrecognized role for the lymph node stromal niche in controlling T cell responses.


Subject(s)
Clonal Selection, Antigen-Mediated , Endothelium, Lymphatic/metabolism , Nitric Oxide Synthase Type II/metabolism , Stromal Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cell Growth Processes/genetics , Cell Movement/genetics , Cells, Cultured , Endothelium, Lymphatic/immunology , Endothelium, Lymphatic/pathology , Intercellular Junctions/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymph Nodes/pathology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Stromal Cells/immunology , Stromal Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transgenes/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
4.
Cancer Cell Int ; 23(1): 3, 2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36609378

ABSTRACT

Hepatocellular carcinoma and intrahepatic cholangiocarcinoma are devastating primary liver cancers with increasing prevalence in many parts of the world. Despite intense investigation, many aspects of their biology are still largely obscure. For example, numerous studies have tackled the question of the cell-of-origin of primary liver cancers using different experimental approaches; they have not, however, provided a clear and undisputed answer. Here, we will review the evidence from animal models supporting the role of all major types of liver epithelial cells: hepatocytes, cholangiocytes, and their common progenitor as liver cancer cell-of-origin. Moreover, we will also propose mechanisms that promote liver cancer cell plasticity (dedifferentiation, transdifferentiation, and epithelial-to-mesenchymal transition) which may contribute to misinterpretation of the results and which make the issue of liver cancer cell-of-origin particularly complex.

5.
Immunol Invest ; 52(8): 966-984, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37846958

ABSTRACT

BACKGROUND & AIMS: Vaccination with tumor-associated antigen-pulsed dendritic cells leads to specific T-cell response against hepatocellular carcinoma. However, clinical response has been shown to be limited. High regulatory T-cell count is associated with poor prognosis and seems to mediate immune tolerance in hepatocellular carcinoma. Forkhead box P3-peptide inhibitor P60 has been shown to specifically inhibit regulatory T-cell function in murine models. Aim of this study was to investigate whether P60 can improve the immune response induced by vaccination with adenovirus-transduced dendritic cells expressing alpha-fetoprotein in subcutaneous and orthotopic murine models for hepatocellular carcinoma. METHODS: Mice developing subcutaneous or orthotopic HCC received daily treatment with P60 starting at different tumor stages. Additionally, mice were vaccinated twice with dendritic cells expressing alpha-fetoprotein. RESULTS: In a preventive setting prior to tumor engraftment, vaccination with alpha-fetoprotein-expressing dendritic cells significantly decreased tumor growth in a subcutaneous model (p = .0256), but no further effects were achieved by addition of P60. However, P60 enhanced the antitumoral effect of a vaccination with alpha-fetoprotein-expressing dendritic cells in established subcutaneous and orthotopic hepatocellular carcinoma characterized by high Treg levels (p = .011). CONCLUSION: In this study, we showed that vaccination with alpha-fetoprotein-expressing dendritic cells in combination with a specific inhibition of regulatory T-cells by using P60 leads to synergistic tumor inhibition and prolonged survival. This emphasizes the importance of regulatory T-cells inhibition for obtaining an effective antitumoral immune response in hepatocellular carcinoma.


Subject(s)
Cancer Vaccines , Carcinoma, Hepatocellular , Liver Neoplasms , T-Lymphocytes, Regulatory , Animals , Mice , Adenoviridae , alpha-Fetoproteins/genetics , Carcinoma, Hepatocellular/pathology , Dendritic Cells , Immunotherapy , Liver Neoplasms/therapy , T-Lymphocytes, Regulatory/drug effects
6.
Nat Immunol ; 11(5): 427-34, 2010 May.
Article in English | MEDLINE | ID: mdl-20305659

ABSTRACT

A major pathway for B cell acquisition of lymph-borne particulate antigens relies on antigen capture by subcapsular sinus macrophages of the lymph node. Here we tested whether this mechanism is also important for humoral immunity to inactivated influenza virus. By multiple approaches, including multiphoton intravital imaging, we found that antigen capture by sinus-lining macrophages was important for limiting the systemic spread of virus but not for the generation of influenza-specific humoral immunity. Instead, we found that dendritic cells residing in the lymph node medulla use the lectin receptor SIGN-R1 to capture lymph-borne influenza virus and promote humoral immunity. Thus, our results have important implications for the generation of durable humoral immunity to viral pathogens through vaccination.


Subject(s)
Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , Endocytosis , Influenza A virus/immunology , Lectins, C-Type/metabolism , Macrophages/metabolism , Receptors, Cell Surface/metabolism , Animals , Antibodies, Viral/blood , Antigen Presentation , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Movement , Cells, Cultured , Clodronic Acid/administration & dosage , Dendrimers/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Endocytosis/drug effects , Endocytosis/genetics , Immunity, Humoral/drug effects , Immunity, Humoral/genetics , Immunoglobulin Heavy Chains/genetics , Immunotherapy, Active , Influenza A virus/pathogenicity , Influenza Vaccines/administration & dosage , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology
7.
Nat Immunol ; 11(4): 313-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20190758

ABSTRACT

Cross-priming allows dendritic cells (DCs) to induce cytotoxic T cell (CTL) responses to extracellular antigens. DCs require cognate 'licensing' for cross-priming, classically by helper T cells. Here we demonstrate an alternative mechanism for cognate licensing by natural killer T (NKT) cells recognizing microbial or synthetic glycolipid antigens. Such licensing caused cross-priming CD8alpha(+) DCs to produce the chemokine CCL17, which attracted naive CTLs expressing the chemokine receptor CCR4. In contrast, DCs licensed by helper T cells recruited CTLs using CCR5 ligands. Thus, depending on the type of antigen they encounter, DCs can be licensed for cross-priming by NKT cells or helper T cells and use at least two independent chemokine pathways to attract naive CTLs. Because these chemokines acted synergistically, this can potentially be exploited to improve vaccinations.


Subject(s)
Chemokine CCL17/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Natural Killer T-Cells/immunology , Receptors, CCR4/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation/immunology , Cell Movement/immunology , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology
8.
Immunity ; 37(2): 276-89, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22884313

ABSTRACT

To initiate adaptive immunity, dendritic cells (DCs) move from parenchymal tissues to lymphoid organs by migrating along stromal scaffolds that display the glycoprotein podoplanin (PDPN). PDPN is expressed by lymphatic endothelial and fibroblastic reticular cells and promotes blood-lymph separation during development by activating the C-type lectin receptor, CLEC-2, on platelets. Here, we describe a role for CLEC-2 in the morphodynamic behavior and motility of DCs. CLEC-2 deficiency in DCs impaired their entry into lymphatics and trafficking to and within lymph nodes, thereby reducing T cell priming. CLEC-2 engagement of PDPN was necessary for DCs to spread and migrate along stromal surfaces and sufficient to induce membrane protrusions. CLEC-2 activation triggered cell spreading via downregulation of RhoA activity and myosin light-chain phosphorylation and triggered F-actin-rich protrusions via Vav signaling and Rac1 activation. Thus, activation of CLEC-2 by PDPN rearranges the actin cytoskeleton in DCs to promote efficient motility along stromal surfaces.


Subject(s)
Cell Movement/physiology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Actins/metabolism , Adaptive Immunity/physiology , Animals , Antigen-Presenting Cells/metabolism , Blood Platelets/metabolism , Cells, Cultured , Dendritic Cells/immunology , Embryo, Mammalian , Endothelial Cells/metabolism , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Myosin Light Chains/metabolism , Platelet Activation , Pregnancy , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction/physiology , Skin/cytology , Skin/metabolism , Tissue Culture Techniques , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
9.
Semin Liver Dis ; 39(1): 70-77, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30654391

ABSTRACT

Extracellular vesicles, comprising exosomes, microvesicles, and apoptotic bodies, represent an emerging field in disease diagnostics and prognosis. They can be isolated from peripheral blood of patients as well as from other body fluids and can therefore be considered a minimally invasive liquid biopsy screening tool. Especially their surface antigen composition can reveal information about disease backgrounds. For several liver diseases, including fatal hepatocellular and cholangiocellular carcinoma as well as other nonmalignant liver disorders such as nonalcoholic fatty liver disease, alcoholic hepatitis, or acute liver failure, it has been shown that extracellular vesicle (EV) surface profiling can be useful for disease diagnosis and prognosis. This review focuses on latest advances in these areas to improve liver disorder detection and management. Additionally, the authors will discuss possible therapeutic applications of EVs in liver diseases, which might be a potent treatment option in the future.


Subject(s)
Cell-Derived Microparticles/physiology , Exosomes/physiology , Liver Diseases/diagnosis , Animals , Biomarkers/blood , Humans , Liver Diseases/therapy , Mice
10.
Liver Int ; 39 Suppl 1: 63-78, 2019 05.
Article in English | MEDLINE | ID: mdl-30907492

ABSTRACT

Tumour microenvironment is a complex, multicellular functional compartment that, particularly when assembled as an abundant desmoplastic reaction, may profoundly affect the proliferative and invasive abilities of epithelial cancer cells. Tumour microenvironment comprises not only stromal cells, mainly cancer-associated fibroblasts, but also immune cells of both the innate and adaptive system (tumour-associated macrophages, neutrophils, natural killer cells, and T and B lymphocytes), and endothelial cells. This results in an intricate web of mutual communications regulated by an extensively remodelled extracellular matrix, where the tumour cells are centrally engaged. In this regard, cholangiocarcinoma, in particular the intrahepatic variant, has become the focus of mounting interest in the last years, largely because of the lack of effective therapies despite its rising incidence and high mortality rates worldwide. On the other hand, recent studies in pancreatic cancer, which similarly to cholangiocarcinoma, is highly desmoplastic, have argued against a tumour-promoting function of the tumour microenvironment. In this review, we will discuss recent developments concerning the role of each cellular population and their multifaceted interplay with the malignant biliary epithelial counterpart. We ultimately hope to provide the working knowledge on how their manipulation may lead to a therapeutic gain in cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms/immunology , Cholangiocarcinoma/immunology , Endothelial Cells/immunology , Fibroblasts/immunology , Tumor Microenvironment/immunology , Adaptive Immunity , Animals , Bile Duct Neoplasms/physiopathology , Cholangiocarcinoma/physiopathology , Disease Models, Animal , Fibroblasts/pathology , Humans , Immunity, Innate
12.
J Hepatol ; 66(3): 619-630, 2017 03.
Article in English | MEDLINE | ID: mdl-27826058

ABSTRACT

Liver progenitor cells (LPCs) are quiescent cells that are activated during liver injury and thought to give rise to hepatocytes and cholangiocytes in order to support liver regeneration and tissue restitution. While hepatocytes are capable of self-renewal, during most chronic injuries the proliferative capacity of hepatocytes is inhibited, thus LPCs provide main source for regeneration. Despite extensive lineage tracing studies, their role and involvement in these processes are often controversial. Additionally, increasing evidence suggests that the LPC compartment consists of heterogeneous cell populations that are actively involved in cellular interactions with myeloid and lymphoid cells during regeneration. On the other hand, LPC expansion has been associated with an increased fibrogenic response, raising concerns about the therapeutic use of these cells. This review aims to summarize the current understanding of the identity, the cellular interactions and the key pathways affecting the biology of LPCs. Understanding the regulatory circuits and the specific role of LPCs is especially important as it could provide novel therapeutic platforms for the treatment of liver inflammation, fibrosis and regeneration.


Subject(s)
Liver/cytology , Liver/injuries , Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Cell- and Tissue-Based Therapy , Hepatocytes/cytology , Hepatocytes/physiology , Humans , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Liver Regeneration/physiology , Stem Cell Niche/physiology , Stem Cells/physiology
13.
J Hepatol ; 66(6): 1241-1250, 2017 06.
Article in English | MEDLINE | ID: mdl-28108233

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic fatty liver (NAFL) is the hepatic consequence of metabolic syndrome and can progress to non-alcoholic steatohepatitis (NASH). The identification of molecular and cellular factors that determine the progression of NASH and lead to irreversible hepatocellular damage are crucial. Dendritic cells (DCs) represent a heterogeneous cell population among which CD103+ DCs play a significant role in immunity and tolerance. We aimed to clarify the role of this DC subset in the pathomechanism of NASH. METHODS: Steatosis progression towards steatohepatitis was analysed using multicolor FACS analyses, cytokine and qPCR array in high sucrose diet (HSD) and methionine and choline deficient diet (MCD) fed wild-type and basic leucine zipper transcription factor, ATF-Like-3 (Batf3) deficient animals, which lack CD103+ DCs (classical type-1 DC, cDC1s). RESULTS: Metabolic challenge of Batf3-/- animals resulted in the progression of steatosis towards steatohepatitis, manifesting by an increased influx of inflammatory cells into the liver and elevated inflammatory cytokine production of myeloid cells upon innate stimuli. However, the lack of cDC1s did not affect cellular apoptosis and fibrosis progression but altered genes involved in lipid metabolism. The adoptive transfer of CD103+ cDC1s to Batf3 deficient animals reversed these observed changes and more importantly could attenuate cellular damage and inflammation in established murine steatohepatitis. CONCLUSION: Here, we have identified the murine CD103+ cDC1s as a protective DC subtype that influences the pro-anti-inflammatory balance and protects the liver from metabolic damage. As guardians of liver integrity, they play a key role in the inflammatory process during the development of steatohepatitis in mice. LAY SUMMARY: Non-alcoholic fatty liver (NAFL) is the hepatic consequence of metabolic syndrome and can lead to non-alcoholic steatohepatitis (NASH). The current study demonstrated that a specific murine dendritic cell subtype possesses a potent regulatory role to influence the inflammatory milieu of the liver in this process.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Integrin alpha Chains/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Adoptive Transfer , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Cytokines/metabolism , Dendritic Cells/classification , Dendritic Cells/metabolism , Disease Progression , Inflammation Mediators/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/immunology , Repressor Proteins/deficiency , Repressor Proteins/genetics
14.
J Hepatol ; 67(2): 282-292, 2017 08.
Article in English | MEDLINE | ID: mdl-28267620

ABSTRACT

BACKGROUND & AIMS: Large extracellular vesicles, specifically AnnexinV+ EpCAM+ CD147+ tumour-associated microparticles (taMPs), facilitate the detection of colorectal carcinoma (CRC), non-small cell lung carcinoma (NSCLC) as well as pancreas carcinoma (PaCa). Here we assess the diagnostic value of taMPs for detection and monitoring of hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). Specifically, the aim of this study was to differentiate liver taMPs from other cancer taMPs, such as CRC and NSCLC. METHODS: Fluorescence-activated cell scanning (FACS) was applied to detect various taMP populations in patients' sera that were associated with the presence of a tumour (AnnexinV+ EpCAM+ CD147+ taMPs) or could discriminate between cirrhosis (due to HCV or HBV) and liver cancers (AnnexinV+ EpCAM+ ASGPR1+ taMPs). In total 172 patients with liver cancer (HCC or CCA), 54 with cirrhosis and no liver neoplasia, and 202 control subjects were enrolled. RESULTS: The results indicate that AnnexinV+ EpCAM+ CD147+ taMPs were elevated in HCC and CCA. Furthermore, AnnexinV+ EpCAM+ ASGPR1+ CD133+ taMPs allowed the distinction of liver malignancies (HCC or CCA) and cirrhosis from tumour-free individuals and, more importantly, from patients carrying other non-liver cancers. In addition, AnnexinV+ EpCAM+ ASGPR1+ taMPs were increased in liver cancer-bearing patients compared to patients with cirrhosis that lacked any detectable liver malignancy. The smallest sizes of successfully detected cancers were ranging between 11-15mm. AnnexinV+ EpCAM+ ASGPR1+ taMPs decreased at 7days after curative R0 tumour resection suggesting close correlations with tumour presence. ROC values, sensitivity/specificity scores and positive/negative predictive values (>78%) indicated a potent diagnostic accuracy of AnnexinV+ EpCAM+ ASGPR1+ taMPs. CONCLUSION: These data provide strong evidence that AnnexinV+ EpCAM+ ASGPR1+ taMPs are a novel biomarker of HCC and CCA liquid biopsy that permit a non-invasive assessment of the presence and possible extent of these cancers in patients with advanced liver diseases. LAY SUMMARY: Microparticles (MPs) are small vesicles that bleb from the membrane of every cell, including cancer cells, and are released to circulate in the bloodstream. Since their surface composition is similar to the surface of their underlying parental cell, MPs from the bloodstream can be isolated and by screening their surface components, the presence of their parental cells can be identified. This way, it was possible to detect and discriminate between patients bearing liver cancer and chronic liver cirrhosis.


Subject(s)
Bile Duct Neoplasms/blood , Carcinoma, Hepatocellular/blood , Cell-Derived Microparticles/pathology , Cholangiocarcinoma/blood , Liver Neoplasms/blood , Adult , Aged , Annexin A5/blood , Asialoglycoprotein Receptor/blood , Basigin/blood , Bile Duct Neoplasms/diagnosis , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/diagnosis , Cell Line, Tumor , Cholangiocarcinoma/diagnosis , Diagnosis, Differential , Epithelial Cell Adhesion Molecule/blood , Female , Hep G2 Cells , Humans , Liver Neoplasms/diagnosis , Male , Middle Aged , Tumor Burden , Young Adult
15.
Am J Physiol Gastrointest Liver Physiol ; 310(1): G1-12, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26564718

ABSTRACT

Podoplanin/gp38(+) stromal cells present in lymphoid organs play a central role in the formation and reorganization of the extracellular matrix and in the functional regulation of immune responses. Gp38(+) cells are present during embryogenesis and in human livers of primary biliary cirrhosis. Since little is known about their function, we studied gp38(+) cells during chronic liver inflammation in models of biliary and parenchymal liver fibrosis and steatohepatitis. Gp38(+) cells were analyzed using flow cytometry and confocal microscopy, and the expression of their steady state and inflammation-associated genes was evaluated from healthy and inflamed livers. Gp38(+) cells significantly expanded in all three models of liver injury and returned to baseline levels during regression of inflammation. Based on CD133 and gp38 expression in the CD45(-)CD31(-)Asgpr1(-) liver cell fraction, numerous subsets could be identified that were negative for CD133 (gp38(hi)CD133(-), gp38(low)CD133(-), and gp38(-)CD133(-)). Moreover, among the CD133(+) cells, previously identified as progenitor population in injured liver, two subpopulations could be distinguished based on their gp38 expression (gp38(-)CD133(+) and CD133(+)gp38(+)). Importantly, the distribution of the identified subsets in inflammation illustrated injury-specific changes. Moreover, the gp38(+)CD133(+) cells exhibited liver progenitor cell characteristics similar to the gp38(-)CD133(+) population, thus representing a novel subset within the classical progenitor cell niche. Additionally, these cells expressed distinct sets of inflammatory genes during liver injury. Our study illuminates a novel classification of the stromal/progenitor cell compartment in the liver and pinpoints a hitherto unrecognized injury-related alteration in progenitor subset composition in chronic liver inflammation and fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver/metabolism , Membrane Glycoproteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Stem Cells/metabolism , Stromal Cells/metabolism , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Separation/methods , Cells, Cultured , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Flow Cytometry , Gene Expression Regulation , Glycoproteins/metabolism , Inflammation Mediators/metabolism , Liver/pathology , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Peptides/metabolism , Phenotype , Stem Cells/pathology , Stromal Cells/pathology , ATP-Binding Cassette Sub-Family B Member 4
16.
Eur J Immunol ; 44(2): 500-10, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24136200

ABSTRACT

The DC-derived chemokine CCL17, a ligand of CCR4, has been shown to promote various inflammatory diseases such as atopic dermatitis, atherosclerosis, and inflammatory bowel disease. Under steady-state conditions, and even after systemic stimulation with LPS, CCL17 is not expressed in resident splenic DCs as opposed to CD8α⁻CD11b⁺ LN DCs, which produce large amounts of CCL17 in particular after maturation. Upon systemic NKT cell activation through α-galactosylceramide stimulation however, CCL17 can be upregulated in both CD8α⁻ and CD8α⁺ splenic DC subsets and enhances cross-presentation of exogenous antigens. Based on genome-wide expression profiling, we now show that splenic CD11b⁺ DCs are susceptible to IFN-γ-mediated suppression of CCL17, whereas LN CD11b⁺CCL17⁺ DCs downregulate the IFN-γR and are much less responsive to IFN-γ. Under inflammatory conditions, particularly in the absence of IFN-γ signaling in IFN-γRKO mice, CCL17 expression is strongly induced in a major proportion of splenic DCs by the action of GM-CSF in concert with IL-4. Our findings demonstrate that the local cytokine milieu and differential cytokine responsiveness of DC subsets regulate lymphoid organ specific immune responses at the level of chemokine expression.


Subject(s)
Cell Differentiation/immunology , Cellular Microenvironment/immunology , Dendritic Cells/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Receptors, Interferon/metabolism , Spleen/metabolism , Animals , CD11b Antigen/immunology , CD11b Antigen/metabolism , Chemokine CCL17/immunology , Chemokine CCL17/metabolism , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interferon-gamma/immunology , Interleukin-4/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Receptors, Interferon/deficiency , Receptors, Interferon/immunology , Spleen/immunology , Interferon gamma Receptor
17.
Liver Int ; 34(3): 447-61, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23998316

ABSTRACT

BACKGROUND: Interleukin 12 (IL-12), one of the most potent Th1-cytokines, has been used to improve dendritic cells (DC)-based immunotherapy of cancer. However, it failed to achieve clinical response in patients with hepatocellular carcinoma (HCC). In this study, improved conditions of immunotherapy with DC engineered to express IL-12 were studied in murine subcutaneous HCC. METHODS: Tumour-lysate pulsed DC were transduced with IL-12-encoding adenoviruses or cultivated with recombinant (r)IL-12. DC were injected intratumourally, subcutaneously or intravenously at different stages of tumour-development. RESULTS: Dendritic cell overexpressing IL-12 by adenoviruses showed enhanced expression of costimulatory molecules and stronger priming of HCC-specific effector cells than DC cultured with rIL-12. Intratumoural but not systemic injections of IL-12-DC induced the strongest antitumoural effects reaching complete regressions in 75% of early-staged tumours and in 33% of advanced tumours. Importantly, antitumoural effects could be further enhanced through combination with sorafenib. Analysing the tumour-environment, IL-12-DC increased the levels of Th1-cytokines/chemokines and of CD4(+) -, CD8(+) -T- and NK-cells. Induced immunity was tumour-specific and sustained since all tumour-free animals were protected towards hepatic tumour-cell rechallenge. However, IL-12-DC also enhanced immunosuppressive cytokines, regulatory T cells and even myeloid-derived suppressor cells within the tumours. CONCLUSIONS: Induced IL-12-overexpression by adenoviral vectors can effectively immunostimulate DC. Intratumoural but not systemic injection of activated IL-12-DC was crucial for effective tumour regression. The mechanism of this approach seems to be the induction of a sufficient Th1 tumour-environment allowing the recruitment of effector cells rather than the inhibition of tumour immunosuppression. Thus, improved immunotherapy with IL-12-DC represents a promising approach towards HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/therapy , Dendritic Cells/immunology , Interleukin-12/genetics , Liver Neoplasms/therapy , Adenoviridae/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Humans , Immunotherapy , Mice , Mice, Inbred C3H , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Sorafenib
18.
Front Immunol ; 15: 1379225, 2024.
Article in English | MEDLINE | ID: mdl-38650949

ABSTRACT

Dendritic cells (DCs) are major antigen-presenting cells that connect innate and adaptive immunity. Hepatic DCs are less activated and contribute to maintain the tolerogenic environment of the liver under steady state. Several studies indicated DCs in metabolic dysfunction-associated steatohepatitis (MASH), representing a substantial burden on healthcare systems due to its association with liver-related morbidity and mortality. Studies highlighted the potential disease-promoting role of liver DCs in the development of MASH while other experimental systems suggested their protective role. This review discusses this controversy and the current understanding of how DCs affect the pathogenesis of MASH.


Subject(s)
Dendritic Cells , Dendritic Cells/immunology , Humans , Animals , Liver/immunology , Fatty Liver/immunology
19.
J Affect Disord ; 360: 126-136, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38815757

ABSTRACT

BACKGROUND: Healthcare professionals are in short supply worldwide, especially in China, which can result in increased stress in the work environment and allostatic load for Chinese hospital staff. This study aimed to investigate the prevalence of anxiety and depressive symptoms and their relationship with total stress, allostatic overload, sleep quality, and episodic memory among Chinese hospital staff. METHOD: In this cross-sectional study, self-assessments including Generalized Anxiety Disorder 7-item (GAD-7), Patient Health Questionnaire-9 (PHQ-9), PsychoSocial Index (PSI), Pittsburgh Sleeping Quality Index (PSQI), and MemTrax test were used to evaluate participants' anxiety symptoms, depressive symptoms, total stress, allostatic load/overload, sleep quality, and episodic memory. RESULTS: A total of 9433 hospital staff from 304 cities participated. Anxiety prevalence was 21.0 % (95 % confidential interval (CI) 20.2 %, 21.8 %), while the prevalence of depressive symptoms was at 21.4 % (95 % CI 20.5 %, 22.2 %). 79.8 % (95 % CI 79.0 %, 80.6 %) of the hospital staff had allostatic overload. Poor sleep quality affected 50.4 % of participants, and 32.1 % experienced poor episodic memory. LIMITATIONS: This study utilized a convenience sampling approach, relying on an online survey as its data collection method. CONCLUSIONS: Hospital staff in China are facing a stressful environment with a high prevalence of anxiety and depressive symptoms, significant allostatic overload, poor sleep quality, and compromised episodic memory. It is imperative that local management and community structures enhance their support and care for these essential workers, enabling them to manage and withstand the stresses of their professional roles effectively.


Subject(s)
Anxiety , Depression , Personnel, Hospital , Humans , Cross-Sectional Studies , Male , Female , Adult , China/epidemiology , Depression/epidemiology , Anxiety/epidemiology , Personnel, Hospital/statistics & numerical data , Personnel, Hospital/psychology , Middle Aged , Prevalence , Sleep Quality , Surveys and Questionnaires , Allostasis/physiology , Anxiety Disorders/epidemiology , Young Adult , Stress, Psychological/epidemiology
20.
Hepatol Commun ; 8(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38358371

ABSTRACT

BACKGROUND: The level of type-I interferons (IFNs) in primary sclerosing cholangitis (PSC) was investigated to evaluate its association with disease activity and progression. METHODS: Bioactive type-I IFNs were evaluated in a murine model of PSC and human patients' sera using a cell-based reporter assay and ELISA techniques. In total, 57 healthy participants, 71 PSC, and 38 patients with primary biliary cholangitis were enrolled in this study. RESULTS: Bioactive type-I IFNs were elevated in the liver and serum of multidrug resistance protein 2-deficient animals and showed a correlation with the presence of CD45+ immune cells and serum alanine transaminase levels. Concordantly, bioactive type-I IFNs were elevated in the sera of patients with PSC as compared to healthy controls (sensitivity of 84.51%, specificity of 63.16%, and AUROC value of 0.8267). Bioactive IFNs highly correlated with alkaline phosphatase (r=0.4179, p<0.001), alanine transaminase (r=0.4704, p<0.0001), and gamma-glutamyl transpeptidase activities (r=0.6629, p<0.0001) but not with serum bilirubin. In addition, patients with PSC with advanced fibrosis demonstrated significantly higher type-I IFN values. Among the type-I IFN subtypes IFNα, ß and IFNω could be detected in patients with PSC with IFNω showing the highest concentration among the subtypes and being the most abundant among patients with PSC. CONCLUSIONS: The selectively elevated bioactive type-I IFNs specifically the dominating IFNω could suggest a novel inflammatory pathway that might also have a hitherto unrecognized role in the pathomechanism of PSC.


Subject(s)
Cholangitis, Sclerosing , Interferon Type I , Liver , Animals , Humans , Mice , Alanine Transaminase , Fibrosis , Interferon Type I/blood , Liver/pathology
SELECTION OF CITATIONS
SEARCH DETAIL