Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biol Res ; 57(1): 21, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704600

ABSTRACT

BACKGROUND: Research on prostate cancer is mostly performed using cell lines derived from metastatic disease, not reflecting stages of tumor initiation or early progression. Establishment of cancer cell lines derived from the primary tumor site has not been described so far. By definition, cancer cells are able to be cultured indefinitely, whereas normal epithelial cells undergo senescence in vitro. Epithelial cells can be immortalized, accomplished by using viral integration of immortalization factors. Viral approaches, however, might be impaired by regulatory and safety issues as well as random integration into regulatory genetic elements, modifying precise gene expression. We intend to use surgical specimen of prostate cancer patients to (i) prove for establishment of cancer cell lines, and (ii) perform non-viral, Sleeping Beauty (SB) transposase-based immortalization of prostate epithelial cells. METHODS: Radical prostatectomy samples of prostate cancer patients (n = 4) were dissociated and cultured in vitro. Cells were cultivated either without or after non-viral, Sleeping-Beauty transposase-based stable transfection with immortalization factors SV40LT and hTERT. Established cell lines were analyzed in vitro and in vivo for characteristics of prostate (cancer) cells. RESULTS: Initial cell cultures without genetic manipulation underwent senescence within ≤ 15 passages, demonstrating inability to successfully derive primary prostate cancer cell lines. By using SB transposase-based integration of immortalization factors, we were able to establish primary prostate cell lines. Three out of four cell lines displayed epithelial characteristics, however without expression of prostate (cancer) characteristics, e.g., androgen receptor. In vivo, one cell line exhibited tumorigenic potential, yet characteristics of prostate adenocarcinoma were absent. CONCLUSION: Whereas no primary prostate cancer cell line could be established, we provide for the first-time immortalization of primary prostate cells using the SB transposase system, thereby preventing regulatory and molecular issues based on viral immortalization approaches. Although, none of the newly derived cell lines demonstrated prostate cancer characteristics, tumor formation was observed in one cell line. Given the non-prostate adenocarcinoma properties of the tumor, cells have presumably undergone oncogenic transformation rather than prostate cancer differentiation. Still, these cell lines might be used as a tool for research on prostate cancer initiation and early cancer progression.


Subject(s)
Epithelial Cells , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/pathology , Cell Line, Tumor , Animals , Prostate/pathology , Carcinogenesis , Telomerase/genetics , Cell Transformation, Neoplastic
2.
Plant J ; 111(5): 1486-1500, 2022 09.
Article in English | MEDLINE | ID: mdl-35819300

ABSTRACT

Quantification of reaction fluxes of metabolic networks can help us understand how the integration of different metabolic pathways determines cellular functions. Yet, intracellular fluxes cannot be measured directly but are estimated with metabolic flux analysis (MFA), which relies on the patterns of isotope labeling of metabolites in the network. The application of MFA also requires a stoichiometric model with atom mappings that are currently not available for the majority of large-scale metabolic network models, particularly of plants. While automated approaches such as the Reaction Decoder Toolkit (RDT) can produce atom mappings for individual reactions, tracing the flow of individual atoms of the entire reactions across a metabolic model remains challenging. Here we establish an automated workflow to obtain reliable atom mappings for large-scale metabolic models by refining the outcome of RDT, and apply the workflow to metabolic models of Arabidopsis thaliana. We demonstrate the accuracy of RDT through a comparative analysis with atom mappings from a large database of biochemical reactions, MetaCyc. We further show the utility of our automated workflow by simulating 15 N isotope enrichment and identifying nitrogen (N)-containing metabolites which show enrichment patterns that are informative for flux estimation in future 15 N-MFA studies of A. thaliana. The automated workflow established in this study can be readily expanded to other species for which metabolic models have been established and the resulting atom mappings will facilitate MFA and graph-theoretic structural analyses with large-scale metabolic networks.


Subject(s)
Arabidopsis , Arabidopsis/metabolism , Carbon Isotopes/metabolism , Isotope Labeling/methods , Metabolic Flux Analysis , Metabolic Networks and Pathways , Models, Biological , Workflow
3.
Br J Cancer ; 129(9): 1462-1476, 2023 10.
Article in English | MEDLINE | ID: mdl-37740039

ABSTRACT

BACKGROUND: Androgen signalling remains the seminal therapeutic approach for the management of advanced prostate cancer. However, most tumours eventually shift towards an aggressive phenotype, characterised by androgen independence and treatment resistance. The cyclic adenosine monophosphate (cAMP) pathway plays a crucial role in regulating various cellular processes, with the phosphodiesterase PDE4D7 being a vital modulator of cAMP signalling in prostate cancer cells. METHODS: Using shRNA-mediated PDE4D7 knockdown in LNCaP cells and downstream analysis via RNA sequencing and phenotypic assays, we replicate clinical observations that diminished PDE4D7 expression promotes an aggressive prostate cancer phenotype. RESULTS: Our study provides evidence that loss of PDE4D7 expression represents a pivotal switch driving the transition from an androgen-sensitive state to hormone unresponsiveness and neuroendocrine differentiation. In addition, we demonstrate that PDE4D7 loss affects DNA repair pathways, conferring resistance to poly ADP ribose polymerase (PARP) inhibitors. CONCLUSION: Reinstating PDE4D7 expression sensitises prostate cancer cells to anti-androgens, DNA damage response inhibitors, and cytotoxic therapies. These findings provide significant insight into the regulatory role of PDE4D7 in the development of lethal prostate cancer and the potential of its modulation as a novel therapeutic strategy.


Subject(s)
Androgens , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/pathology , Prostate/metabolism , DNA Repair , Cell Differentiation , Receptors, Androgen/genetics , Cell Line, Tumor
4.
Int J Cancer ; 144(3): 607-614, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30265376

ABSTRACT

BRCA1 is a pivotal tumor suppressor. Its dysfunction is known to play a role in different tumors. Among others, BRCA1 germline mutations account for higher risk and more aggressive course of prostate cancer (PCa). In addition, somatic BRCA1 gene loss was demonstrated to be a signature of PCa dissemination to lymph nodes and peripheral blood, and indicate worse clinical outcome. In order to substantiate the data for BRCA1 gene loss in PCa and reveal its phenotypical background, BRCA1 gene status was assessed in a large cohort of PCa patients and compared to different molecular factors. BRCA1 gene dosage was assessed in 2398 tumor samples from 1,199 PCa patients using fluorescent in situ hybridization. It was compared to clinico-pathological parameters, patients' outcome as well as selected proteins (Ki-67, apoptosis marker, cytokeratins, vimentin, E- and N-cadherin, ALDH1 and EGFR) examined immunohistochemically. BRCA1 losses were found in 10%, whereas gains appeared in 7% of 603 informative PCa patients. BRCA1 losses correlated to higher T stage (p = 0.027), Gleason score (p = 0.039), shorter time to biochemical recurrence in patients with Gleason score > 7 independently of other factors (multivariate analysis, p = 0.005) as well as expression of proteins regulating stemness and epithelial-mesenchymal transition, that is, ALDH1 (p = 0.021) and EGFR (p = 0.011), respectively. BRCA1 gains correlated to shorter time to metastasis (p = 0.012) and expression of ALDH1 (p = 0.014). These results support the assumption that BRCA1 gene losses contribute to a progressive and stem cell-like phenotype of PCa. Furthermore, they reveal that also BRCA1 gain conceivably representing loss-of-function might mark more invasive tumors.


Subject(s)
Genes, BRCA1 , Germ-Line Mutation , Isoenzymes/metabolism , Prostatic Neoplasms/genetics , Retinal Dehydrogenase/metabolism , Aged , Aged, 80 and over , Aldehyde Dehydrogenase 1 Family , BRCA1 Protein/genetics , Disease Progression , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Immunohistochemistry , Isoenzymes/biosynthesis , Isoenzymes/genetics , Male , Middle Aged , Prostatic Neoplasms/metabolism , Retinal Dehydrogenase/biosynthesis , Retinal Dehydrogenase/genetics
5.
J Card Surg ; 34(10): 1097-1099, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31374577

ABSTRACT

The noninvasive characterization of cardiac tumors is of clinical importance for surgical resection planning. Conventional radiological examinations like cardiac computed tomography (CT) or magnetic resonance imaging (MRI) may be misleading as benign cardiac lesions can present features suspicious for malignancy. Moreover, the low prevalence of cardiac tumors may additionally hamper a sound diagnosis. However, fluorodeoxyglucose-positron emission tomography (FDG-PET) has proven to be a reliable tool for cardiac tumor characterization. Here, FDG-PET/CT imaging of a 50-year-old man suffering from a cardiac tumor is presented. Despite CT and MRI signs of malignancy, FDG-PET characterized the tumor as benign. Histology confirmed the FDG-PET prediction and revealed a pericardial capillary hemangioma. Thereby, it seems important to integrate FDG-PET in the diagnostic workup of cardiac tumors.


Subject(s)
Fluorodeoxyglucose F18/pharmacology , Heart Neoplasms/diagnosis , Hemangioma/diagnosis , Positron Emission Tomography Computed Tomography/methods , Diagnosis, Differential , Heart Atria , Humans , Male , Middle Aged , Radiopharmaceuticals/pharmacology , Rare Diseases
6.
Eur J Nucl Med Mol Imaging ; 45(5): 860-877, 2018 05.
Article in English | MEDLINE | ID: mdl-29335762

ABSTRACT

BACKGROUND: Prostate-specific membrane antigen (PSMA) is the up-and-coming target for molecular imaging of prostate cancer. Despite its name, non-prostate-related PSMA expression in physiologic tissue as well as in benign and malignant disease has been reported in various publications. Unlike in prostate cancer, PSMA expression is only rarely observed in non-prostate tumor cells. Instead, expression occurs in endothelial cells of tumor-associated neovasculature, although no endothelial expression is observed under physiologic conditions. The resulting potential for tumor staging in non-prostate malignant tumors has been demonstrated in first patient studies. This review summarizes the first clinical studies and deduces future perspectives in staging, molecular characterization, and PSMA-targeted radionuclide therapy based on histopathologic examinations of PSMA expression. CONCLUSIONS: The non-exclusivity of PSMA in prostate cancer opens a window to utilize the spectrum of available radioactive PSMA ligands for imaging and molecular characterization and maybe even therapy of non-prostate disease.


Subject(s)
Gene Expression Profiling , Neoplasms/diagnostic imaging , Prostate-Specific Antigen/analysis , Gene Expression Regulation, Neoplastic , Humans , Ligands , Magnetic Resonance Imaging , Male , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms , Radiopharmaceuticals , Whole Body Imaging
7.
Prostate ; 77(4): 396-405, 2017 03.
Article in English | MEDLINE | ID: mdl-27862105

ABSTRACT

BACKGROUND: Fresh tissue is mandatory to perform high-quality translation studies. Several models for tissue extraction from prostatectomy specimens without guidance by frozen sections are already introduced. However, little is known about the sampling efficacy of these models, which should provide representative tissue in adequate volumes, account for multifocality and heterogeneity of tumor, not violate the routine final pathological examination, and perform quickly without frozen section-based histological control. The aim of the study was to evaluate the sampling efficacy of the existing tissue extraction models without guidance by frozen sections ("blind") and to develop an optimized model for tissue extraction. METHODS: Five hundred thirty-three electronic maps of the tumor distribution in prostates from a single-center cohort of the patients subjected to radical prostatectomy were used for analysis. Six available models were evaluated in silico for their sampling efficacy. Additionally, a novel model achieving the best sampling efficacy was developed. RESULTS: The available models showed high efficacies for sampling "any part" from the tumor (up to 100%), but were uniformly low in efficacy to sample all tumor foci from the specimens (with the best technique sampling only 51.6% of the all tumor foci). The novel 4-level extraction model achieved a sampling efficacy of 93.1% for all tumor foci. CONCLUSIONS: The existing "blind" tissue extraction models from prostatectomy specimens without frozen sections control are suitable to target tumor tissues but these tissues do not represent the whole tumor. The novel 4-level model provides the highest sampling efficacy and a promising potential for integration into routine. Prostate 77: 396-405, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Biological Specimen Banks , Prostatectomy/methods , Prostatic Neoplasms/pathology , Specimen Handling/methods , Biological Specimen Banks/standards , Cohort Studies , Frozen Sections/methods , Frozen Sections/standards , Humans , Male , Prostatectomy/standards , Single-Blind Method , Specimen Handling/standards
8.
Am J Pathol ; 185(11): 3025-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26506472

ABSTRACT

The immunoregulatory cytokine IL-10 suppresses T-cell immunity. The complementary question, whether IL-10 is also involved in limiting the collateral damage of vigorous T cell responses, has not been addressed in detail. Here, we report that the particularly strong virus-specific immune response during acute primary infection with the lymphocytic choriomeningitis virus (LCMV) in mice is significantly further increased in Il10-deficient mice, particularly regarding frequencies and cytotoxic activity of CD8(+) T cells. This increase results in exacerbating immunopathology in select organs, ranging from transient local swelling to an increased risk for mortality. Remarkably, LCMV-induced, T cell-mediated hepatitis is not affected by endogenous Il10. The alleviating effect of Il10 on LCMV-induced immunopathology was found to be operative in delayed-type hypersensitivity footpad-swelling reaction and in debilitating meningitis in mice of both the C57BL/6 and BALB/c strains. These strains are prototypic counterpoles for genetically imprinted type 1-biased versus type 2-biased T cell-mediated immune responses against various infectious pathogens. However, during acute LCMV infection, neither systemic cytokine patterns nor the impact of Il10 on LCMV-induced immunopathology differed conspicuously between these two strains of mice. This study documents a physiological role of Il10 in the regulation of a balanced T-cell response limiting immunopathological damage.


Subject(s)
Antiviral Agents/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Interleukin-10/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antiviral Agents/metabolism , CD8-Positive T-Lymphocytes/physiology , Cytokines/blood , Cytokines/immunology , Female , Hypersensitivity, Delayed , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocytic Choriomeningitis/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
9.
Int J Cancer ; 137(11): 2578-88, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26084847

ABSTRACT

Liposarcomas (LS) are the most common malignant mesenchymal tumors, with an overall long-term mortality rate of 60%. LS comprise three major subtypes, i.e., well-differentiated/dedifferentiated liposarcoma (WDLS/DDLS), myxoid/round cell liposarcoma (MLS) and pleomorphic liposarcoma (PLS). Aiming at the preclinical identification of novel therapeutic options, we here investigate the functional significance of SRC in primary human LS and in LS-derived cell lines. Immunohistochemical and Western blot analyses reveal relevant levels of activated p-(Tyr416)-SRC in LS of the different subtypes with particular activation in MLS and PLS. Dysregulation of the SRC modifiers CSK and PTP1B was excluded as major reason for the activation of the kinase. Consistent siRNA-mediated knockdown of SRC or inhibition by the SRC inhibitor Dasatinib led to decreased proliferation of LS cell lines of the different subtypes, with MLS cells reacting particularly sensitive in MTT assays. Flow cytometric analyses revealed that this effect was due to a significant decrease in mitotic activity and an induction of apoptosis. SRC inhibition by Dasatinib resulted in dephosphorylation of SRC itself, its interacting partners FAK and IGF-IR as well as its downstream target AKT. Consistent with a particular role of SRC in cell motility, Dasatinib reduced the migratory and invasive potential of MLS cells in Boyden chamber and Matrigel chamber assays. In summary, we provide evidence that SRC activation plays an important role in LS biology and therefore represents a potential therapeutic target, particularly in MLS and PLS.


Subject(s)
Liposarcoma, Myxoid/drug therapy , Liposarcoma/drug therapy , Protein Kinase Inhibitors/pharmacology , src-Family Kinases/antagonists & inhibitors , Apoptosis/drug effects , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Focal Adhesion Kinase 1/metabolism , Humans , Mitosis/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism
10.
Hepatology ; 59(2): 630-42, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23959575

ABSTRACT

UNLABELLED: Chronic liver injury promotes hepatic inflammation, representing a prerequisite for organ fibrosis. We hypothesized a contribution of chemokine receptor CCR6 and its ligand, CCL20, which may regulate migration of T-helper (Th)17, regulatory, and gamma-delta (γδ) T cells. CCR6 and CCL20 expression was intrahepatically up-regulated in patients with chronic liver diseases (n = 50), compared to control liver (n = 5). Immunohistochemistry revealed the periportal accumulation of CCR6(+) mononuclear cells and CCL20 induction by hepatic parenchymal cells in liver disease patients. Similarly, in murine livers, CCR6 was expressed by macrophages, CD4 and γδ T-cells, and up-regulated in fibrosis, whereas primary hepatocytes induced CCL20 upon experimental injury. In two murine models of chronic liver injury (CCl4 and methionine-choline-deficient diet), Ccr6(-/-) mice developed more severe fibrosis with strongly enhanced hepatic immune cell infiltration, compared to wild-type (WT) mice. Although CCR6 did not affect hepatic Th-cell subtype composition, CCR6 was explicitly required by the subset of interleukin (IL)-17- and IL-22-expressing γδ T cells for accumulation in injured liver. The adoptive transfer of WT γδ, but not CD4 T cells, into Ccr6(-/-) mice reduced hepatic inflammation and fibrosis in chronic injury to WT level. The anti-inflammatory function of hepatic γδ T cells was independent of IL-17, as evidenced by transfer of Il-17(-/-) cells. Instead, hepatic γδ T cells colocalized with hepatic stellate cells (HSCs) in vivo and promoted apoptosis of primary murine HSCs in a cell-cell contact-dependent manner, involving Fas-ligand (CD95L). Consistent with γδ T-cell-induced HSC apoptosis, activated myofibroblasts were more frequent in fibrotic livers of Ccr6(-/-) than in WT mice. CONCLUSION: γδ T cells are recruited to the liver by CCR6 upon chronic injury and protect the liver from excessive inflammation and fibrosis by inhibiting HSCs.


Subject(s)
Cell Movement , Hepatitis/prevention & control , Liver Cirrhosis/prevention & control , Liver Diseases/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR6/metabolism , T-Lymphocytes/metabolism , Animals , Apoptosis , Case-Control Studies , Chemokine CCL20/metabolism , Disease Models, Animal , Female , Hepatitis/metabolism , Hepatitis/pathology , Humans , Interleukin-17/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Diseases/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR6/deficiency , Receptors, CCR6/genetics , T-Lymphocytes/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th17 Cells/metabolism , Th17 Cells/pathology , Up-Regulation
11.
Hepatology ; 60(1): 334-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24619965

ABSTRACT

UNLABELLED: Activation of the renin angiotensin system resulting in stimulation of angiotensin-II (AngII) type I receptor (AT1R) is an important factor in the development of liver fibrosis. Here, we investigated the role of Janus kinase 2 (JAK2) as a newly described intracellular effector of AT1R in mediating liver fibrosis. Fibrotic liver samples from rodents and humans were compared to respective controls. Transcription, protein expression, activation, and localization of JAK2 and downstream effectors were analyzed by real-time polymerase chain reaction, western blotting, immunohistochemistry, and confocal microscopy. Experimental fibrosis was induced by bile duct ligation (BDL), CCl4 intoxication, thioacetamide intoxication or continuous AngII infusion. JAK2 was inhibited by AG490. In vitro experiments were performed with primary rodent hepatic stellate cells (HSCs), Kupffer cells (KCs), and hepatocytes as well as primary human and human-derived LX2 cells. JAK2 expression and activity were increased in experimental rodent and human liver fibrosis, specifically in myofibroblastic HSCs. AT1R stimulation in wild-type animals led to activation of HSCs and fibrosis in vivo through phosphorylation of JAK2 and subsequent RhoA/Rho-kinase activation. These effects were prevented in AT1R(-/-) mice. Pharmacological inhibition of JAK2 attenuated liver fibrosis in rodent fibrosis models. In vitro, JAK2 and downstream effectors showed increased expression and activation in activated HSCs, when compared to quiescent HSCs, KCs, and hepatocytes isolated from rodents. In primary human and LX2 cells, AG490 blocked AngII-induced profibrotic gene expression. Overexpression of JAK2 led to increased profibrotic gene expression in LX2 cells, which was blocked by AG490. CONCLUSION: Our study substantiates the important cell-intrinsic role of JAK2 in HSCs for development of liver fibrosis. Inhibition of JAK2 might therefore offer a promising therapy for liver fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Janus Kinase 2/metabolism , Liver Cirrhosis/metabolism , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/toxicity , Animals , Bile Ducts , Carbon Tetrachloride/toxicity , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Humans , Ligation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/metabolism , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Thioacetamide/toxicity
12.
Mod Pathol ; 26(7): 1004-12, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23599150

ABSTRACT

About 10-15% of gastrointestinal stromal tumors (GISTs) carry wild-type sequences in all hot spots of KIT and platelet-derived growth factor receptor alpha (PDGFRA) (wt-GISTs). These tumors are currently defined by having no mutations in exons 9, 11, 13, and 17 of the KIT gene and exons 12, 14, and 18 of the PDGFRA gene. Until now, the analysis of further exons is not recommended. However, we have previously published a report on a KIT exon 8 germline mutation, which was associated with familial GIST and mastocytosis. We therefore investigated whether KIT exon 8 mutations might also occur in sporadic GIST. We screened a cohort of 145 wt-GISTs from a total of 1351 cases from our registry for somatic mutations in KIT exon 8. Two primary GISTs with an identical exon 8 mutation (p.D419del) were detected, representing 1.4% of all the cases analyzed. Based on all GISTs from our registry, the overall frequency of KIT exon 8 mutations was 0.15%. The first tumor originating in the small bowel of a 53-year-old male patient had mostly a biphasic spindled-epithelioid pattern with a high proliferative activity (14 mitoses/50 HPF) combined with a second low proliferative spindle cell pattern (4/50 HPF). The patient developed multiple peritoneal metastases 29 months later. The second case represented a jejunal GIST in a 67-year old woman who is relapse-free under adjuvant imatinib treatment. We conclude that about 1-2% of GISTs being classified as 'wild type' so far might, in fact, carry KIT mutations in exon 8. Moreover, this mutational subtype was shown to be activating and imatinib sensitive in vitro. We therefore propose that screening for KIT exon 8 mutations should become a routine in the diagnostic work-up of GIST and that patients with an exon 8 mutation and a significant risk for tumor progression should be treated with imatinib.


Subject(s)
Gastrointestinal Stromal Tumors/genetics , Proto-Oncogene Proteins c-kit/genetics , Adolescent , Aged , Aged, 80 and over , Child , DNA Mutational Analysis , Exons/genetics , Female , Gastrointestinal Stromal Tumors/pathology , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Mutation , Young Adult
13.
Hepatology ; 55(3): 898-909, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22031018

ABSTRACT

UNLABELLED: Chemokines critically control the infiltration of immune cells upon liver injury, thereby promoting hepatic inflammation and fibrosis. The chemokine receptor CCR8 can affect trafficking of monocytes/macrophages, monocyte-derived dendritic cells (DCs) and T-helper cell (Th) subsets, but its role in liver diseases is currently unknown. To investigate the functional role of CCR8 in liver diseases, ccr8(-/-) and wild-type (WT) mice were subjected to chronic experimental injury models of carbon tetrachloride (CCl(4) ) administration and surgical bile duct ligation (BDL). CCR8 was strongly up-regulated in the injured liver. Ccr8(-/-) mice displayed attenuated liver damage (e.g., ALT, histology, and TUNEL) compared to WT mice and were also protected from liver fibrosis in two independent injury models. Flow cytometry revealed reduced infiltrates of liver macrophages, neutrophils and natural killer cells, whereas hepatic CD4(+) T cells increased. The main CCR8-expressing cells in the liver were hepatic macrophages, and CCR8 was functionally necessary for CCL1-directed migration of inflammatory but not for nonclassical monocytes into the liver. Moreover, the phenotype of liver macrophages from injured ccr8(-/-) animals was altered with increased expression of DC markers and enhanced expression of T-cell-attracting chemokine macrophage inflammatory protein 1-alpha (MIP-1α/CCL3). Correspondingly, hepatic CD4(+) T cells showed increased Th1 polarization and reduced Th2 cells in CCR8-deficient animals. Liver fibrosis progression, but also subsequent T-cell alterations, could be restored by adoptively transferring CCR8-expressing monocytes/macrophages into ccr8(-/-) mice during experimental injury. CONCLUSIONS: CCR8 critically mediates hepatic macrophage recruitment upon injury, which subsequently shapes the inflammatory response in the injured liver, affecting macrophage/DC and Th differentiation. CCR8 deficiency protects the liver against injury, ameliorating initial inflammatory responses and hepatic fibrogenesis. Inhibition of CCR8 or its ligand, CCL1, might represent a successful therapeutic target to limit liver inflammation and fibrosis progression.


Subject(s)
Cell Differentiation/physiology , Cell Movement/physiology , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Liver/pathology , Macrophages/pathology , Receptors, CCR8/physiology , Animals , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Female , Immunity, Innate/physiology , Killer Cells, Natural/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/pathology , Phenotype , Receptors, CCR8/deficiency , Receptors, CCR8/genetics , Up-Regulation/physiology
14.
Hepatology ; 56(3): 1140-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22454377

ABSTRACT

UNLABELLED: Liver fibrogenesis is associated with the transition of quiescent hepatocytes and hepatic stellate cells (HSCs) into the cell cycle. Exit from quiescence is controlled by E-type cyclins (cyclin E1 [CcnE1] and cyclin E2 [CcnE2]). Thus, the aim of the current study was to investigate the contribution of E-type cyclins for liver fibrosis in man and mice. Expression of CcnE1, but not of its homolog, CcnE2, was induced in fibrotic and cirrhotic livers from human patients with different etiologies and in murine wild-type (WT) livers after periodical administration of the profibrotic toxin, CCl(4). To further evaluate the potential function of E-type cyclins for liver fibrogenesis, we repetitively treated constitutive CcnE1(-/-) and CcnE2(-/-) knock-out mice with CCl(4) to induce liver fibrosis. Interestingly, CcnE1(-/-) mice were protected against CCl(4)-mediated liver fibrogenesis, as evidenced by reduced collagen type I α1 expression and the lack of septum formation. In contrast, CcnE2(-/-) mice showed accelerated fibrogenesis after CCl(4) treatment. We isolated primary HSCs from WT, CcnE1(-/-), and CcnE2(-/-) mice and analyzed their activation, proliferation, and survival in vitro. CcnE1 expression in WT HSCs was maximal when they started to proliferate, but decreased after the cells transdifferentiated into myofibroblasts. CcnE1(-/-) HSCs showed dramatically impaired survival, cell-cycle arrest, and strongly reduced expression of alpha smooth muscle actin, indicating deficient HSC activation. In contrast, CcnE2-deficient HSCs expressed an elevated level of CcnE1 and showed enhanced cell-cycle activity and proliferation, compared to WT cells. CONCLUSIONS: CcnE1 and CcnE2 have antagonistic roles in liver fibrosis. CcnE1 is indispensable for the activation, proliferation, and survival of HSCs and thus promotes the synthesis of extracellular matrix and liver fibrogenesis.


Subject(s)
Cell Proliferation , Cyclin E/physiology , Hepatic Stellate Cells/physiology , Liver Cirrhosis/pathology , Oncogene Proteins/physiology , Animals , Humans , Male , Mice
15.
Hepatology ; 56(5): 1924-33, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22610745

ABSTRACT

UNLABELLED: Immunity against cancer is impeded by local mechanisms promoting development of tumor-specific T cell tolerance, such as regulatory T cells, myeloid-derived suppressor cells, or immunosuppressive factors in the tumor microenvironment. The release of soluble antigens, such as carcinoembryonic antigen (CEA) from colorectal carcinoma (CRC) cells, has been investigated for diagnostic purposes, but not for its immunological consequences. Here, we address the question of whether soluble CEA influences tumor-specific immunity. Mice were injected with soluble CEA protein, and CEA-specific CD8 T cells were analyzed for their phenotype and functionality by means of restimulation ex vivo or antitumor efficacy in vivo. We furthermore characterized the CD8 T cell population in peripheral blood mononuclear cell (PBMCs) from healthy donors and colorectal carcinoma patients. In mice, circulating CEA was preferentially taken up in a mannose receptor-dependent manner and cross-presented by liver sinusoidal endothelial cells, but not dendritic cells, to CD8 T cells. Such systemically circulating CEA promoted tolerization of CEA-specific CD8 T cells in the endogenous T cell repertoire through the coinhibitory molecule B7H1. These CD8 T cells were not deleted but were rendered nonresponsive to antigen-specific stimulation and failed to control growth of CEA-expressing tumor cells. These nonresponsive CD8 T cells were phenotypically similar to central memory T cells being CD44(high) CD62L(high) CD25(neg) . We found T cells with a similar phenotype in PBMCs of healthy donors and at increased frequency also in patients with colorectal carcinoma. CONCLUSION: Our results provide evidence for the existence of an unrecognized tumor immune escape involving cross-presentation of systemically circulating tumor antigens that may influence immunotherapy of cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoembryonic Antigen/immunology , Carcinoma/immunology , Colorectal Neoplasms/immunology , Endothelial Cells/immunology , Immune Tolerance , Liver/immunology , Animals , Antigen-Presenting Cells/immunology , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Carcinoembryonic Antigen/blood , Carcinoma/blood , Colorectal Neoplasms/blood , Humans , Hyaluronan Receptors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , L-Selectin/metabolism , Leukocytes, Mononuclear/immunology , Lymphocyte Count , Mice , Mice, Transgenic , Phenotype
16.
Histopathology ; 62(2): 294-304, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23020601

ABSTRACT

AIMS AND METHODS: Desmoid-type fibromatosis (desmoid) is a fibroblastic tumour that shows locally aggressive growth. Mesenteric desmoid is a rare lesion that shares morphological and biological features with fibromatoses occurring in the abdominal wall or in extraabdominal sites, but differs in terms of gross appearance and clinical presentation. We report on a series of 56 cases of mesenteric desmoids from our consultation files and compare them with cases of non-mesenteric desmoids and retroperitoneal fibrosis. RESULTS: Primary diagnosis of desmoid-type fibromatosis was correct in 42%, and gastrointestinal stromal tumour was a common misdiagnosis. Nuclear expression of ß-catenin was detected in 91.6% of all desmoids. Mutational analysis of exon 3 of the ß-catenin gene (CTNNB1) revealed that mesenteric desmoids carried mutations significantly more often (51/56, 91.1%) than non-mesenteric tumours (20/28; 71.4%; P = 0.027). p.T41A occurred significantly more frequently in mesenteric fibromatoses (80.4%) than in abdominal wall and extra-abdominal fibromatoses (46.4%; P = 0.002). Two novel mutations (p.S45C and p.D32G) were found. In retroperitoneal fibrosis, mutations and nuclear ß-catenin expression were absent. ß-Catenin-negative desmoids either carried a CTNNB1 mutation or were associated with Gardner syndrome. CONCLUSIONS: Our study provides evidence that some clinical and genetic features of mesenteric desmoids differ from those of non-mesenteric fibromatosis, and corroborates the usefulness of mutational analysis, especially in diagnosing ß-catenin-negative mesenteric desmoids.


Subject(s)
Diagnostic Errors , Fibromatosis, Aggressive/diagnosis , Gastrointestinal Stromal Tumors/diagnosis , Mutation , Peritoneal Neoplasms/diagnosis , beta Catenin/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cell Nucleus/metabolism , Cell Nucleus/pathology , Child , Female , Fibromatosis, Aggressive/genetics , Gardner Syndrome/genetics , Gardner Syndrome/pathology , Humans , Male , Mesentery/pathology , Middle Aged , Peritoneal Neoplasms/genetics , Retroperitoneal Fibrosis/diagnosis , Retroperitoneal Fibrosis/genetics , Young Adult
17.
BMC Gastroenterol ; 13: 8, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23311569

ABSTRACT

BACKGROUND: Four and one half LIM domain protein 2 (FHL2) has been reported to be a key regulator in many cellular processes being associated with fibrogenesis such as cell migration and contraction. Moreover, hepatic FHL2 is involved in regulation pathways mediating proliferation and cell death machineries. We here investigated the role of FHL2 in the setting of experimental and clinical liver fibrosis. METHODS: FHL2(-/-) and wild type (wt) mice were challenged with CCl(4). Fibrotic response was assessed by quantitative real time PCR (qRT-PCR) of fibrotic marker genes, measurement of hydroxyproline content and histological methods. Murine FHL2(-/-) and hepatic stellate cells (HSC) were isolated and investigated via immunofluorescence. Human fibrotic and normal liver samples were analysed immunohistochemically using antibodies directed against FHL2. RESULTS: FHL2(-/-) mice displayed aggravated liver fibrosis compared to wt mice. However, immunofluorescence revealed no significant morphological changes in cultured FHL2(-/-) and wt myofibroblasts (MFB). In human liver samples, FHL2 was strongly expressed both in the nucleus and cytoplasm in MFB of fibrotic livers. In contrast, FHL2 expression was absent in normal liver tissue. CONCLUSIONS: Deficiency of FHL2 results in aggravation of murine liver fibrosis. In human liver samples, FHL2 is expressed in activated HSCs and portal fibroblasts in human fibrotic livers, pointing to a central role of FHL2 for human hepatic fibrogenesis as well.


Subject(s)
Disease Progression , LIM-Homeodomain Proteins/deficiency , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Muscle Proteins/deficiency , Transcription Factors/deficiency , Animals , Apoptosis , Carbon Tetrachloride/adverse effects , Cell Movement , Cells, Cultured , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Gut ; 61(3): 416-26, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21813474

ABSTRACT

OBJECTIVE: Monocyte chemoattractant protein-1 (MCP-1, CCL2), the primary ligand for chemokine receptor C-C chemokine receptor 2 (CCR2), is increased in livers of patients with non-alcoholic steatohepatitis (NASH) and murine models of steatohepatitis and fibrosis. It was recently shown that monocyte/macrophage infiltration into the liver upon injury is critically regulated by the CCL2/CCR2 axis and is functionally important for perpetuating hepatic inflammation and fibrogenesis. The structured L-enantiomeric RNA oligonucleotide mNOX-E36 (a so-called Spiegelmer) potently binds and inhibits murine MCP-1. Pharmacological inhibition of MCP-1 with mNOX-E36 was investigated in two murine models of chronic liver diseases. METHODS: Pharmacological inhibition of MCP-1 by thrice-weekly mNOX-E36 subcutaneously was tested in murine models of acute or chronic carbon tetrachloride (CCl(4))- and methionine-choline-deficient (MCD) diet-induced chronic hepatic injury in vivo. RESULTS: Antagonising MCP-1 by mNOX-E36 efficiently inhibited murine monocyte chemotaxis in vitro as well as migration of Gr1(+) (Ly6C(+)) blood monocytes into the liver upon acute toxic injury in vivo. In murine models of CCl(4)- and MCD diet-induced hepatic injury, the infiltration of macrophages into the liver was significantly decreased in anti-MCP-1-treated mice as found by fluorescence-activated cell sorting (FACS) analysis and immunohistochemistry. In line with lower levels of intrahepatic macrophages, proinflammatory cytokines (tumour necrosis factor α, interferon γ and interleukin 6) were significantly reduced in liver tissue. Overall fibrosis progression over 6 (CCl(4)) or 8 weeks (MCD diet) was not significantly altered by anti-MCP-1 treatment. However, upon MCD diet challenge a lower level of fatty liver degeneration (histology score, Oil red O staining, hepatic triglyceride content, lipogenesis genes) was detected in mNOX-E36-treated animals. mNOX-E36 also ameliorated hepatic steatosis upon therapeutic administration. CONCLUSIONS: These results demonstrate the successful pharmacological inhibition of hepatic monocyte/macrophage infiltration by blocking MCP-1 during chronic liver damage in two in vivo models. The associated ameliorated steatosis development suggests that inhibition of MCP-1 is an interesting novel approach for pharmacological treatment in liver inflammation and steatohepatitis.


Subject(s)
Aptamers, Nucleotide/pharmacology , Chemical and Drug Induced Liver Injury, Chronic/complications , Chemokine CCL2/antagonists & inhibitors , Fatty Liver/prevention & control , Macrophages/drug effects , Acute Disease , Animals , Aptamers, Nucleotide/therapeutic use , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Carbon Tetrachloride , Cells, Cultured , Chemical and Drug Induced Liver Injury/complications , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury, Chronic/drug therapy , Chemical and Drug Induced Liver Injury, Chronic/pathology , Chemokine CCL2/physiology , Chemotaxis/drug effects , Cytokines/metabolism , Disease Progression , Drug Evaluation, Preclinical/methods , Fatty Liver/drug therapy , Fatty Liver/etiology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/prevention & control , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease
19.
Gut ; 61(12): 1733-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22267597

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a typical inflammation-associated cancer, but may also provoke antitumour immune responses whose significance and underlying mechanisms are incompletely understood. OBJECTIVE: To characterise immune responses in the diethylnitrosamine (DEN)-liver cancer mouse model. DESIGN: Tumour development and immune cell functions upon DEN treatment were compared between C57BL/6 wild-type (WT), chemokine scavenging receptor D6-deficient, B cell- (Igh6), CD4 T cell- (MHC-II) and T-/B cell-deficient (Rag1) mice. Relevance for human HCC was tested by comparing gene array results from 139 HCC tissues. RESULTS: The induction of premalignant lesions after 24 weeks and of HCC-like tumours after 42 weeks by DEN in mice was accompanied by significant leucocyte infiltration in the liver and upregulation of distinct intrahepatic chemokines (CCL2, CCL5, CXCL9). Macrophages and CD8 (cytotoxic) T cells were most prominently enriched in tumour-bearing livers, similar to samples from human HCC. Myeloid-derived suppressor cells (MDSC) increased in extrahepatic compartments of DEN-treated mice (bone marrow, spleen). The contribution of immune cell subsets for DEN-induced hepatocarcinogenesis was functionally dissected. In D6(-/-) mice, which lack the chemokine scavenging receptor D6, hepatic macrophage infiltration was significantly increased, but tumour formation and progression did not differ from that of WT mice. In contrast, progression of hepatic tumours (numbers, diameters, tumour load) was strikingly enhanced in T-/B cell-deficient Rag1(-/-) mice upon DEN treatment. When mice deficient for B cells (Igh6(-/-), µMT) or major histocompatibility complex II were used, the data indicated that T cells prevent initial tumour formation, while B cells critically limit growth of established tumours. Accordingly, in tumour-bearing mice antibody production against liver-related model antigen was enhanced, indicating tumour-associated B cell activation. In agreement, T and B cell pathways were differentially regulated in gene array analyses from 139 human HCC tissues and significantly associated with patients' survival. CONCLUSIONS: Distinct axes of the adaptive immune system, which are also prognostic in human HCC, actively suppress DEN-induced hepatocarcinogenesis by controlling tumour formation and progression.


Subject(s)
Adaptive Immunity , Carcinoma, Hepatocellular/immunology , Liver Neoplasms, Experimental/immunology , Animals , B-Lymphocytes/metabolism , Biomarkers/metabolism , Carcinogens , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Chemokine CXCL9/metabolism , Diethylnitrosamine , Disease Progression , Humans , Leukocytes/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/chemically induced , Precancerous Conditions/immunology , Precancerous Conditions/pathology , Survival Analysis , T-Lymphocytes, Cytotoxic/metabolism
20.
Front Plant Sci ; 14: 1178239, 2023.
Article in English | MEDLINE | ID: mdl-37346134

ABSTRACT

Quantification of reaction fluxes of metabolic networks can help us understand how the integration of different metabolic pathways determine cellular functions. Yet, intracellular fluxes cannot be measured directly but are estimated with metabolic flux analysis (MFA) that relies on the patterns of isotope labeling of metabolites in the network. For metabolic systems, typical for plants, where all potentially labeled atoms effectively have only one source atom pool, only isotopically nonstationary MFA can provide information about intracellular fluxes. There are several global approaches that implement MFA for an entire metabolic network and estimate, at once, a steady-state flux distribution for all reactions with identifiable fluxes in the network. In contrast, local approaches deal with estimation of fluxes for a subset of reactions, with smaller data demand for flux estimation. Here we present a systematic comparative review and benchmarking of the existing local approaches for isotopically nonstationary MFA. The comparison is conducted with respect to the required data and underlying computational problems solved on a synthetic network example. Furthermore, we benchmark the performance of these approaches in estimating fluxes for a subset of reactions using data obtained from the simulation of nitrogen fluxes in the Arabidopsis thaliana core metabolism. The findings pinpoint practical aspects that need to be considered when applying local approaches for flux estimation in large-scale plant metabolic networks.

SELECTION OF CITATIONS
SEARCH DETAIL