Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Internist (Berl) ; 61(2): 170-174, 2020 Feb.
Article in German | MEDLINE | ID: mdl-31938815

ABSTRACT

Cholangiocellular carcinoma (CCA) is one of the primary liver tumors and overall represents a rare malignancy; however, in recent years the incidence, particularly of intrahepatic CCA (iCCA) has increased worldwide. Due to the high mortality, CCAs cause a significant proportion of cancer-related deaths also in Germany. Because the diagnosis is often made in advanced stages of the disease, in many cases a surgical approach with curative intention is not possible. For locally advanced or metastatic CCA the combination of gemcitabine and cisplatin currently remains the only approved systemic treatment. As the average survival time is only approximately 12 months even under first-line treatment with gemcitabine/cisplatin, research is focused on developing new molecularly targeted and immunological treatment options. Various studies are currently being carried out to investigate approval options for targeted treatment, which could be considered for genetically altered tumors, e.g. in fibroblast growth factor receptor (FGFR) fusion and isocitrate dehydrogenase (IDH) mutations. Additionally, initial clinical data on immune checkpoint inhibitors are available for CCA. Due to the complex selection and partially limited applicability of current treatment options in patients with CCA, an early collaboration with a gastroenterology and oncology center with the possibility of supervision by a tumor board consisting of gastroenterological oncologists, surgeons, radiologists and radio-oncologists or in advanced stages by a molecular tumor board is essential.


Subject(s)
Bile Duct Neoplasms , Bile Ducts, Intrahepatic , Cholangiocarcinoma , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/therapy , Germany , Humans , Molecular Targeted Therapy , Precision Medicine
2.
Mol Imaging Biol ; 20(1): 4-20, 2018 02.
Article in English | MEDLINE | ID: mdl-28971346

ABSTRACT

The 6th annual meeting to address key issues in positron emission tomography (PET)/magnetic resonance imaging (MRI) was held again in Tübingen, Germany, from March 27 to 29, 2017. Over three days of invited plenary lectures, round table discussions and dialogue board deliberations, participants critically assessed the current state of PET/MRI, both clinically and as a research tool, and attempted to chart future directions. The meeting addressed the use of PET/MRI and workflows in oncology, neurosciences, infection, inflammation and chronic pain syndromes, as well as deeper discussions about how best to characterise the tumour microenvironment, optimise the complementary information available from PET and MRI, and how advanced data mining and bioinformatics, as well as information from liquid biomarkers (circulating tumour cells and nucleic acids) and pathology, can be integrated to give a more complete characterisation of disease phenotype. Some issues that have dominated previous meetings, such as the accuracy of MR-based attenuation correction (AC) of the PET scan, were finally put to rest as having been adequately addressed for the majority of clinical situations. Likewise, the ability to standardise PET systems for use in multicentre trials was confirmed, thus removing a perceived barrier to larger clinical imaging trials. The meeting openly questioned whether PET/MRI should, in all cases, be used as a whole-body imaging modality or whether in many circumstances it would best be employed to give an in-depth study of previously identified disease in a single organ or region. The meeting concluded that there is still much work to be done in the integration of data from different fields and in developing a common language for all stakeholders involved. In addition, the participants advocated joint training and education for individuals who engage in routine PET/MRI. It was agreed that PET/MRI can enhance our understanding of normal and disrupted biology, and we are in a position to describe the in vivo nature of disease processes, metabolism, evolution of cancer and the monitoring of response to pharmacological interventions and therapies. As such, PET/MRI is a key to advancing medicine and patient care.


Subject(s)
Magnetic Resonance Imaging , Positron-Emission Tomography , Humans , Liquid Biopsy , Radiotherapy, Image-Guided , Tumor Microenvironment
3.
Oncogene ; 36(14): 1911-1924, 2017 04 06.
Article in English | MEDLINE | ID: mdl-27748763

ABSTRACT

MYC genes have both essential roles during normal development and exert oncogenic functions during tumorigenesis. Expression of a dominant-negative allele of MYC, termed OmoMYC, can induce rapid tumor regression in mouse models with little toxicity for normal tissues. How OmoMYC discriminates between physiological and oncogenic functions of MYC is unclear. We have solved the crystal structure of OmoMYC and show that it forms a stable homodimer and as such recognizes DNA in the same manner as the MYC/MAX heterodimer. OmoMYC attenuates both MYC-dependent activation and repression by competing with MYC/MAX for binding to chromatin, effectively lowering MYC/MAX occupancy at its cognate binding sites. OmoMYC causes the largest decreases in promoter occupancy and changes in expression on genes that are invaded by oncogenic MYC levels. A signature of OmoMYC-regulated genes defines subgroups with high MYC levels in multiple tumor entities and identifies novel targets for the eradication of MYC-driven tumors.


Subject(s)
Neoplasms/genetics , Peptide Fragments/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/physiology , Binding Sites , Cells, Cultured , DNA/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, Dominant , Humans , Models, Molecular , Neoplasms/pathology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Multimerization , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Sequence Homology , Transcriptome , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
4.
Internist (Berl) ; 57(12): 1191-1205, 2016 Dec.
Article in German | MEDLINE | ID: mdl-27822622

ABSTRACT

Cholangiocellular carcinoma (CCA) is the second most frequent primary liver carcinoma and is an aggressive tumor, which is mostly diagnosed in advanced stages. The overall survival is poor. Histpathological analysis of tumor biopsies or cytological analysis of biliary brushings can be used to confirm the diagnosis. A differentiation is made between distal, perihilar and intrahepatic CCA. The anatomical position determines the diagnostic and therapeutic strategy. Before diagnostic or therapeutic measures are undertaken it is essential to resolve biliary obstruction via endoscopic stenting or percutaneous biliary drainage. Depending on the tumor stage curative treatment options comprise radical surgical resection with hepaticojejunostomy or in selected cases liver transplantation. For intrahepatic or distal CCA liver transplantation is not indicated. In the palliative setting systemic chemotherapy with gemcitabine and cisplatin leads to a significant improvement in survival time.


Subject(s)
Antineoplastic Agents/administration & dosage , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/therapy , Cholangiocarcinoma/pathology , Cholangiocarcinoma/therapy , Endoscopy, Digestive System/methods , Combined Modality Therapy/methods , Drainage/methods , Evidence-Based Medicine , Hepatectomy/methods , Humans , Liver Transplantation/methods , Treatment Outcome
5.
Oncogene ; 30(10): 1229-40, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21076472

ABSTRACT

Yes-associated protein (YAP) is a downstream effector of the Hippo signaling pathway, which controls organ expansion and tissue development. We have recently defined the tumorigenic potential and clinical significance of the YAP1 oncogene in human hepatocellular carcinoma (HCC). The present study aims to define the tumorigenic properties of YAP in HCC and elucidate the related downstream signaling mechanism. In a gain-of-function study, we demonstrated that ectopic increased expression of YAP in the immortalized non-tumorigenic hepatocyte cell line MIHA confers tumorigenic and metastatic potentials, as evidenced by (1) enhanced aptitudes in cell viability, anchorage-independent growth, migration and invasion; (2) tumor formation in a xenograft mouse model; and (3) induction of HCC biomarker α-fetoprotein and activation of mitogen-activated protein kinase. Furthermore, we have identified AXL, a receptor tyrosine kinase, as a key downstream target that drives YAP-dependent oncogenic functions. RNAi-mediated knockdown of AXL expression decreased the ability of YAP-expressing MIHA cells and of the primary HCC cell line to proliferate and invade. These results indicate that AXL is a mediator of YAP-dependent oncogenic activities and implicates it as a potential therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins , Cell Line , Cell Movement/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Gene Expression , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Immunoblotting , Liver Neoplasms/genetics , Male , Mice , Mice, Nude , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , RNA, Small Interfering , Receptor Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection , Transplantation, Heterologous , Axl Receptor Tyrosine Kinase
6.
Gene Ther ; 16(7): 849-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19369968

ABSTRACT

Overexpression of myeloid cell leukemia 1 protein (Mcl-1), an anti-apoptotic B-cell lymphoma 2 (Bcl-2) family member, contributes to chemotherapy resistance of tumors. The short half-life of Mcl-1 makes it an interesting target for therapeutic agents that negatively interfere with cellular protein biosynthesis, such as oncolytic viruses. Vesicular Stomatitis Virus (VSV) has been established as the oncolytic virus that efficiently disrupts de novo protein biosynthesis of infected cells. Here, we show that after VSV infection, Mcl-1 protein levels rapidly declined, whereas the expression of other members of the Bcl-2 family remained unchanged. Mcl-1 elimination was a consequence of proteasomal degradation, as overexpression of a degradation-resistant Mcl-1 mutant restored Mcl-1 levels. Mcl-1 rescue inhibited apoptosis and thereby confirmed that Mcl-1 downregulation contributes to VSV-induced apoptosis. In vitro, VSV virotherapy in combination with chemotherapy revealed an enhanced therapeutic effect compared with the single treatments, which could be reverted by Mcl-1 rescue or RNA interference (RNAi)-mediated knockdown of pro-apoptotic Bax and Bak proteins. Finally, in a tumor mouse model, combinations of doxorubicin and VSV showed a superior therapeutic efficacy compared with VSV or doxorubicin alone. In summary, our data indicate that VSV virotherapy is an attractive strategy to overcome tumor resistance against conventional chemotherapy by elimination of Mcl-1.


Subject(s)
Apoptosis/drug effects , Neoplasm Proteins/biosynthesis , Oncolytic Virotherapy , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Vesicular stomatitis Indiana virus/physiology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Autophagy/drug effects , Autophagy/physiology , Caspase 3/metabolism , Cell Line, Tumor , Cytopathogenic Effect, Viral , Down-Regulation/genetics , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Half-Life , Humans , Mice , Mice, Nude , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/growth & development , Virus Replication/drug effects , Xenograft Model Antitumor Assays
7.
Article in English | MEDLINE | ID: mdl-19150958

ABSTRACT

Cellular senescence is characterized by an irreversible cell cycle arrest that, when bypassed by mutation, contributes to cellular immortalization. Activated oncogenes induce a hyperproliferative response, which might be one of the senescence cues. We have found that expression of such an oncogene, Akt, causes senescence in primary mouse hepatoblasts in vitro. Additionally, AKT-driven tumors undergo senescence in vivo following p53 reactivation and show signs of differentiation. In another in vivo system, i.e., liver fibrosis, hyperproliferative signaling through AKT might be a driving force of the senescence in activated hepatic stellate cells. Senescent cells up-regulate and secrete molecules that, on the one hand, can reinforce the arrest and, on the other hand, can signal to an innate immune system to clear the senescent cells. The mechanisms governing senescence and immortalization are overlapping with those regulating self-renewal and differentiation. These respective control mechanisms, or their disregulation, are involved in multiple pathological conditions including fibrosis, wound healing, and cancer. Understanding extracellular cues that regulate these processes may enable new therapies for these conditions.


Subject(s)
Cellular Senescence , Neoplasms/pathology , Stem Cells/cytology , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Cellular Senescence/genetics , Cellular Senescence/physiology , Gene Expression , Genes, p53 , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/metabolism , Humans , Immunity, Innate , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , Neoplasms/genetics , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Oncogenes , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Stem Cells/metabolism , Wound Healing
8.
Gut ; 54(11): 1590-6, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227360

ABSTRACT

BACKGROUND AND AIMS: Tumour necrosis factor related apoptosis inducing ligand (TRAIL) induces apoptosis in transformed cells and is considered as an agent for cancer therapy. As there is evidence that TRAIL is also essential for apoptosis in animal models of liver injury, we investigated the role of TRAIL in viral hepatitis and after alcohol consumption. METHODS: Expression of TRAIL was determined by western blot analysis in the liver of patients with chronic hepatitis C virus (HCV) infection as well as in experimental acute adenoviral hepatitis and after alcohol intake in the liver of mice. To investigate the effect of FasL and TRAIL expression, we used low dose adenoviral gene transfer. Apoptosis and steatosis were assessed by TUNEL and fat red staining, and by caspase assays. RESULTS: TRAIL was overexpressed in the liver of patients with HCV associated steatosis while acute adenoviral hepatitis resulted in upregulation of TRAIL-DR5. In contrast with FasL, TRAIL expression was harmless to healthy livers. However, in virally infected livers, TRAIL expression induced apoptosis and steatosis whereas expression of FasL only resulted in apoptosis of hepatocytes without steatosis. After alcohol intake, TRAIL expression led to hepatic steatosis, without apoptosis of hepatocytes, indicating that TRAIL mediated apoptosis and steatosis may be independently modulated after viral infection and alcohol intake. In viral hepatitis and after alcohol intake, Ad-TRAIL mediated steatosis can be inhibited by injection of a neutralising TRAIL antibody. CONCLUSIONS: We identified TRAIL as a new mediator of hepatic steatosis in viral hepatitis and after alcohol intake. Consequently, TRAIL mediated hepatotoxicity has to be considered in patients with viral hepatitis and alcoholic liver disease.


Subject(s)
Alcohol Drinking/adverse effects , Fatty Liver/etiology , Hepatitis C, Chronic/complications , Membrane Glycoproteins/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis , Apoptosis Regulatory Proteins , Fatty Liver/metabolism , Fatty Liver/virology , Hepatitis C, Chronic/metabolism , Hepatocytes/pathology , Humans , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
9.
Article in English | MEDLINE | ID: mdl-16869761

ABSTRACT

Hepatocellular carcinoma is a chemoresistant cancer and a leading cause of cancer mortality; however, the molecular mechanisms responsible for the aggressive nature of this disease are poorly understood. In this study, we developed a new liver cancer mouse model that is based on the ex vivo genetic manipulation of embryonic liver progenitor cells (hepatoblasts). After retroviral gene transfer of oncogenes or short hairpin RNAs targeting tumor suppressor genes, genetically altered liver progenitor cells are seeded into the liver of otherwise normal recipient mice. We show that histopathology of the engineered liver carcinomas reveals features of the human disease. Furthermore, representational oligonucleotide microarray analysis (ROMA) of murine liver tumors initiated by two defined genetic hits revealed spontaneously acquired genetic alterations that are characteristic for human hepatocellular carcinoma. This model provides a powerful platform for applications like cancer gene discovery or high-throughput preclinical drug testing.


Subject(s)
Hepatocytes/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Multipotent Stem Cells/pathology , Neoplastic Stem Cells/pathology , Animals , Disease Models, Animal , Female , Gene Targeting , Genes, Reporter , Genes, Tumor Suppressor , Green Fluorescent Proteins/genetics , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Oncogenes , RNA Interference , Recombinant Proteins/genetics , Transduction, Genetic
10.
Apoptosis ; 9(1): 51-4, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14739598

ABSTRACT

Apoptosis is a genetically controlled mechanism of cell death which is important for embryogenesis, metamorphosis, tissue homeostasis and tumor regression of multicellular organisms. In normal cells as well as in transformed cells signals released from the cytoplasm and/or the cell membrane can trigger the activation of caspases which in turn cleave many cellular substrates, leading to the characteristic morphology of apoptosis. Systematic analysis and dissection of apoptotic pathways was obtained by the use of knock out or transgenic organisms, expressing dominant active or negative proteins. The use of antisense oligonucleotides (ASO) for analysis of apoptotic pathways was commonly restricted to cultured cells, because of the low efficacy of ASO in in vivo experiments. In order to investigate the contribution of specific apoptotic pathways in the onset and maintenance of disease in vivo experiments are needed. This approach allows the analysis of apoptotic pathways within their physiological/pathophysiological environment. The combination of recent advances in in vivo gene delivery with siRNA technique for efficient gene silencing provides new, unique possibilities to study apoptotic pathways thereby evaluating new molecular therapeutic strategies in vivo. In this minireview we will focus on the use of RNA interference for analytical and therapeutical suppression of apoptotic pathways in vivo with special consideration of the liver.


Subject(s)
Apoptosis , Genetic Techniques , RNA, Small Interfering/metabolism , Animals , Genes, Dominant , Humans , Liver/metabolism , Liver Failure/prevention & control , Mice , Oligonucleotides, Antisense/chemistry , RNA/chemistry
12.
J Mol Biol ; 309(4): 855-68, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11399064

ABSTRACT

CCAAT/enhancer binding protein beta (C/EBP beta) also named liver-enriched transcriptional activating protein (LAP) is a member of the C/EBP family of transcription factors and is involved in hepatocyte-specific gene expression and in the process of tissue differentiation. The activity of LAP/C/EBP beta can be regulated at the transcriptional and posttranslational level or by protein-protein interaction with other transcription factors. In this study we show that LAP/C/EBP beta can stimulate its own transcription. Deletion analysis of the rat LAP/C/EBP beta promoter in luciferase reporter gene experiments demonstrated that the region located between nucleotide -121 to -71, comprising two recently characterized cAMP responsive element (CRE)-like elements, is important for autoregulation. Gel shift experiments using oligonucleotides with overlapping point mutations identified the sequence GCAATGA (beta-site) adjacent to and partially overlapping the first CRE-like site as core motif for LAP/C/EBP beta binding. Analysis of a mutated beta-site in reporter gene experiments showed the functional relevance of this site for autoregulation. The composite C/EBP beta-CRE-element in the promoter enables synergistic activation of transcription by LAP/C/EBP beta and the protein kinase A (PKA)/cAMP responsive element binding protein (CREB) pathway in a cell-type specific manner. In hepatoma cells nuclear factor kappa B (NF-kappa B) increased autoregulation and therefore could mediate enhanced activation during inflammatory responses. In summary, our results demonstrated that the assembly of the three binding sites in the promoter and thus the interaction between LAP/C/EBP beta and members of the CREB or NF-kappa B family allows the control of LAP/C/EBP beta gene transcription as a response to different stimuli in a tissue specific manner.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Signal Transduction , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Animals , Binding Sites , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Footprinting , Deoxyribonuclease I/metabolism , Genes, Reporter/genetics , Humans , Mice , NF-kappa B/metabolism , Organ Specificity , Promoter Regions, Genetic/genetics , Rats , Response Elements/genetics , Sequence Deletion/genetics , Tumor Cells, Cultured
13.
Cancer Gene Ther ; 7(10): 1315-23, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11059688

ABSTRACT

The transcription factor nuclear factor kappaB (NFkappaB) is an essential antagonist of apoptosis during liver regeneration and embryonal development of hepatocytes. Several reports have indicated that NFkappaB may also inhibit the programmed cell death induced by cytokines, ionizing radiation, or cytotoxic drugs in some cancer cell lines. Because hepatocellular carcinomas (HCCs) are one of the most resistant tumors to systemic chemotherapy, we investigated the activation of NFkappaB and the consequence of its inhibition by an IkappaBalpha-super repressor during tumor necrosis factor alpha (TNFalpha)- and chemotherapy-induced apoptosis in HCC cell lines. We demonstrate that both TNFalpha and adriamycin activate NFkappaB in hepatoma cells. Activation of NFkappaB could be blocked through an adenoviral vector expressing the IkappaBalpha super repressor, regardless of the activating agent. Inhibition of NFkappaB enhanced the apoptosis induced by TNFalpha, whereas IkappaBalpha had an anti-apoptotic effect on chemotherapy-induced programmed cell death. A strong inhibition of chemotherapy- and TNFalpha-induced apoptosis by dominant-negative Fas-associated death domain indicated an essential contribution of death receptor-mediated apoptosis. To elucidate the different role of NFkappaB in chemotherapy-induced apoptosis, we investigated the expression of Fas (CD95) and Fas ligand (CD95 ligand), which have been described as important mediators of chemotherapy-induced cell death and as target genes of NFkappaB. However, our investigations demonstrated that in hepatoma cells, the chemotherapy-induced up-regulation of Fas (CD95) and Fas ligand (CD95 ligand) is not transcriptionally mediated through NFkappaB. Thus, other molecular mechanisms must account for the anti-apoptotic effect of IkappaBalpha in adriamycin-induced death of hepatoma cells. In summary, our investigations indicate that the activation of NFkappaB in response to cytotoxic drugs, in contrast to TNFalpha, exerts a pro-apoptotic stimulus rather than an anti-apoptotic function, which has implications for therapy of HCCs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/therapy , DNA-Binding Proteins/genetics , Doxorubicin/pharmacology , Genetic Therapy/methods , I-kappa B Proteins , Liver Neoplasms/therapy , NF-kappa B/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Adenoviridae/genetics , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Caspases/metabolism , DNA-Binding Proteins/metabolism , Drug Resistance , Electrophoresis, Agar Gel , Fas Ligand Protein , Flow Cytometry , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Glycoproteins/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Structure-Activity Relationship , Transfection , Tumor Cells, Cultured , fas Receptor/metabolism , fas Receptor/physiology
14.
J Biol Chem ; 275(9): 6421-7, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10692445

ABSTRACT

NFkappaB is an essential survival factor in several physiological conditions such as embryonal liver development and liver regeneration. However, NFkappaB is also a main mediator of the cellular response to a variety of extracellular stress stimuli, and it has been shown that some viral-induced host cell apoptosis appears to be dependent on NFkappaB activation. The activation of NFkappaB upon viral infection may be a rapid way of initiating an innate immune response against the viral particles. We have assessed the role of NFkB during the early phase of adenoviral hepatitis in a nude mouse model using an adenoviral vector expressing a mutant form of IkappaBalpha. Administration of a LacZ-expressing adenoviral vector induces NFkB DNA and correlates with the up-regulation of Fas (CD95) mRNA, but not FasL (CD95L) mRNA, during the early phase of adenoviral hepatitis. The rapid increase in NFkappaB DNA binding after adenoviral infection of the liver could be very effectively inhibited by IkappaBalpha. Compared with the LacZ control virus, the IkappaBalpha-expressing adenoviral vector inhibits the increase of Fas (CD95) mRNA expression, in particular in the very early phase of the hepatitis. Reporter gene experiments in hepatoma cell lines with a Fas promoter-luciferase construct indicated that the repression of Fas (CD95) mRNA by IkappaBalpha was transcriptionally mediated. The functional relevance of the NFkappaB-dependent increase in Fas (CD95) transcription was assessed by caspase 3 assays and terminal dUTP nick-end labeling tests. Compared with the control, IkappaBalpha adenoviral infection resulted in reduced caspase 3 activity during the early phase of viral hepatitis and in a prevention of liver cell apoptosis 24 h after adenoviral administration. Therefore our study demonstrates a new pro-apoptotic function of NFkappaB in Fas (CD95)-mediated apoptosis of hepatocytes. Interestingly, NFkappaB mediates liver cell apoptosis upon viral infection even in a phase where tumor necrosis factor-alpha is already induced, as shown by the time curves of tumor necrosis factor-alpha serum levels. Therefore, the pro- or anti-apoptotic role of NFkappaB appears to be more determined by the nature of the death stimulus than by the origin of the tissue.


Subject(s)
Adenoviridae Infections/genetics , Apoptosis , I-kappa B Proteins , NF-kappa B/metabolism , Transcriptional Activation/genetics , fas Receptor/genetics , Animals , Caspase 3 , Caspases/metabolism , Cell Line , DNA-Binding Proteins/genetics , Genes, Reporter , Hepatitis, Animal/virology , Humans , In Situ Nick-End Labeling , Liver/pathology , Liver/virology , Mice , Mice, Nude , NF-KappaB Inhibitor alpha , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
J Biol Chem ; 274(45): 32137-44, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542249

ABSTRACT

p53 is a transcription factor that is activated by genotoxic stress and mediates cell cycle arrest and apoptosis. Here we demonstrate that infection of mouse liver with recombinant E1/E3-deleted adenovirus leads to p53 activation and simultaneously to the down-regulation of albumin gene expression. In vitro transcription assays indicate that transcriptional mechanisms mediated through the albumin promoter are responsible for reduced albumin mRNA levels during viral infection. Albumin expression is maintained in the liver by a combination of liver-enriched transcription factors such as CAAT enhancer-binding protein (C/EBP)alpha and C/EBPbeta. We show that p53 wild type and tumor-derived p53 mutations repress C/EBP-mediated transactivation of the albumin promoter. The binding of C/EBPalpha or -beta to its cognate sequence in the albumin promoter is not inhibited by p53 expression. Deletion analysis and domain swapping experiments show that repression of C/EBPbeta-mediated transactivation is dependent on the N-terminal domain of p53 and the transactivation domain, leucine zipper domain, and the inhibitory domain II (amino acids 163-191) of C/EBPbeta. Our results provide a molecular explanation for the p53-mediated down-regulation of liver-specific gene expression after viral infection. Additionally, as overexpression of p53 mutants is frequently found in undifferentiated hepatocellular carcinomas, the same mechanisms may contribute to the lack of liver-specific gene transcription in these tumors.


Subject(s)
Albumins/genetics , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Hepatitis, Viral, Animal/physiopathology , Hepatitis, Viral, Human/physiopathology , Liver Neoplasms/etiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , CCAAT-Enhancer-Binding Proteins , Humans , Liver Neoplasms/virology , Mice , Mutagenesis , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...