Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 11(10): 875, 2020 10 17.
Article in English | MEDLINE | ID: mdl-33070156

ABSTRACT

Since metastatic colorectal cancer (CRC) is a leading cause of cancer-related death, therapeutic approaches overcoming primary and acquired therapy resistance are an urgent medical need. In this study, the efficacy and toxicity of high-affinity inhibitors targeting antiapoptotic BCL-2 proteins (BCL-2, BCL-XL, and MCL-1) were evaluated. By RNA sequencing analysis of a pan-cancer cohort comprising >1500 patients and subsequent prediction of protein activity, BCL-XL was identified as the only antiapoptotic BCL-2 protein that is overactivated in CRC. Consistently, pharmacologic and genetic inhibition of BCL-XL induced apoptosis in human CRC cell lines. In a combined treatment approach, targeting BCL-XL augmented the efficacy of chemotherapy in vitro, in a murine CRC model, and in human ex vivo derived CRC tissue cultures. Collectively, these data show that targeting of BCL-XL is efficient and safe in preclinical CRC models, observations that pave the way for clinical translation.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Humans , Myeloid Cell Leukemia Sequence 1 Protein/drug effects , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/drug effects
2.
Int J Mol Sci ; 21(3)2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32046105

ABSTRACT

Autophagy is a catabolic process that enables cells to degrade obsolete content and refuel energy depots. In colorectal cancer (CRC) autophagy has been shown to promote tumorigenesis through energy delivery in the condition of uncontrolled proliferation. With this study, we aimed at evaluating whether autophagy sustains CRC cell viability and if it impacts therapy resistance. Initially, a colorectal cancer tissue micro array, containing mucosa (n = 10), adenoma (n = 18) and adenocarcinoma (n = 49) spots, was stained for expression of essential autophagy proteins LC3b, Atg7, p62 and Beclin-1. Subsequently, central autophagy proteins were downregulated in CRC cells using siRNA technology. Viability assays, flow cytometry and immunoblotting were performed and three-dimensional cell culture was utilized to study autophagy in a tissue mimicking environment. In our study we found an upregulation of Atg7 in CRC. Furthermore, we identified Atg7 as crucial factor within the autophagy network for CRC cell viability. Its disruption induced cell death via triggering apoptosis and in combination with conventional chemotherapy it exerted synergistic effects in inducing CRC cell death. Cell death was strictly dependent on nuclear LC3b, since simultaneous knockdown of Atg7 and LC3b completely restored viability. This study unravels a novel cell death preventing function of Atg7 in interaction with LC3b, thereby unmasking a promising therapeutic target in CRC.


Subject(s)
Adenocarcinoma/metabolism , Apoptosis , Autophagy-Related Protein 7/metabolism , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Adenocarcinoma/genetics , Antineoplastic Agents/pharmacology , Autophagy , Autophagy-Related Protein 7/genetics , Beclin-1/genetics , Beclin-1/metabolism , Cell Survival/drug effects , Cells, Cultured , Colorectal Neoplasms/genetics , Fluorouracil/pharmacology , HT29 Cells , Humans , Irinotecan/pharmacology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism
3.
Gastroenterology ; 156(4): 1190-1205.e14, 2019 03.
Article in English | MEDLINE | ID: mdl-30445013

ABSTRACT

BACKGROUND & AIMS: Cholangiocyte proliferation and ductular reaction contribute to the onset and progression of liver diseases. Little is known about the role of the transcription factor nuclear factor-κB (NF-κB) in this process. We investigated the activities of the RELB proto-oncogene NF-κB subunit in human cholangiocytes and in mouse models of liver disease characterized by a ductular reaction. METHODS: We obtained liver tissue samples from patients with primary sclerosing cholangitis, primary biliary cholangitis, hepatitis B or C virus infection, autoimmune hepatitis, alcoholic liver disease, or without these diseases (controls) from a tissue bank in Germany. Tissues were analyzed by immunohistochemistry for levels of RELB and lymphotoxin ß (LTB). We studied mice with liver parenchymal cell (LPC)-specific disruption of the cylindromatosis (CYLD) lysine 63 deubiquitinase gene (Cyld), with or without disruption of Relb (CyldΔLPC mice and Cyld/RelbΔLPC mice) and compared them with C57BL/6 mice (controls). Mice were fed 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) or standard chow diets to induce biliary injury or were given injections of CCl4 to induce non-cholestatic liver fibrosis. Liver tissues were analyzed by histology, immunohistochemistry, immunoblots, in situ hybridization, and quantitative real-time polymerase chain reaction. Cholangiocytes were isolated from normal human liver, incubated with LTB receptor agonist, and transfected with small interfering RNAs to knock down RELB. RESULTS: In liver tissues from patients with primary sclerosing cholangitis, primary biliary cholangitis, chronic infection with hepatitis B or C virus, autoimmune hepatitis, or alcoholic liver disease, we detected increased nuclear translocation of RELB and increased levels of LTB in cholangiocytes that formed reactive bile ducts compared with control liver tissues. Human cholangiocytes, but not those with RELB knockdown, proliferated with exposure to LTB. The phenotype of CyldΔLPC mice, which included ductular reaction, oval cell activation, and biliary fibrosis, was completely lost from Cyld/RelbΔLPC mice. Compared with livers from control mice, livers from CyldΔLPC mice (but not Cyld/RelbΔLPC mice) had increased levels of mRNAs encoding cytokines (LTB; CD40; and tumor necrosis factor superfamily [TNFSF] members TNFSF11 [RANKL], TNFSF13B [BAFF], and TNFSF14 [LIGHT]) produced by reactive cholangiocytes. However, these strains of mice developed similar levels of liver fibrosis in response to CCl4 exposure. CyldΔLPC mice and Cyld/RelbΔLPC mice had improved liver function on the DDC diet compared with control mice fed the DDC diet. CONCLUSION: Reactive bile ducts in patients with chronic liver diseases have increased levels of LTB and nuclear translocation of RELB. RELB is required for the ductular reaction and development of biliary fibrosis in CyldΔLPC mice. Deletion of RELB and CYLD from LPCs protects mice from DDC-induced cholestatic liver fibrosis.


Subject(s)
Bile Ducts/metabolism , Bile Ducts/pathology , Cholangitis, Sclerosing/metabolism , Cytokines/genetics , Liver Diseases/metabolism , Transcription Factor RelB/metabolism , Adolescent , Adult , Aged , Animals , Carbon Tetrachloride , Cell Nucleus , Cell Proliferation , Cells, Cultured , Cysteine Endopeptidases/genetics , Deubiquitinating Enzyme CYLD , Dicarbethoxydihydrocollidine , Epithelial Cells/metabolism , Female , Fibrosis , Gene Knockdown Techniques , Humans , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Lymphotoxin beta Receptor/agonists , Lymphotoxin-beta/metabolism , Male , Mice , Middle Aged , Parenchymal Tissue/pathology , Protein Transport , Proto-Oncogene Mas , RNA, Messenger/metabolism , Transcription Factor RelB/genetics , Young Adult
4.
EBioMedicine ; 32: 125-133, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29884457

ABSTRACT

A higher capacity to grow under hypoxic conditions can lead to a more aggressive behavior of tumor cells. Determining tumor activity under hypoxia may identify chronic lymphocytic leukemia (CLL) with aggressive clinical course and predict response to chemo-immunotherapy (CIT). A metabolic score was generated by determining pyruvate kinase and lactate dehydrogenase, key enzymes of glycolysis, ex vivo in primary CLL samples under normoxic and hypoxic conditions. This score was further correlated with clinical endpoints and response to CIT in 96 CLL patients. 45 patients were classified as metabolic high risk (HR), 51 as low risk (LR). Treatment-free survival (TFS) was significantly shorter in HR patients (median 394 vs 723 days, p = .021). 15 HR patients and 14 LR patients received CIT after sample acquisition. HR patients had a significantly shorter progression-free survival after treatment compared to LR patients (median 216 days vs not reached, p = .008). Multivariate analysis evaluating age, IGHV, TP53 deletion or mutation and 11q22-23 deletion besides the capacity of tumor cells to grow under severe hypoxic conditions identified the metabolic profile as the strongest independent risk factor for shorter TFS (hazard ratio 2.37, p = .011). The metabolic risk can provide prognostic and predictive information complementary to genetic biomarkers and identify patients who might benefit from alternative treatment approaches.


Subject(s)
Biomarkers, Tumor/genetics , Immunotherapy , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Prognosis , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Glycolysis/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Mutation , Proportional Hazards Models , Risk Factors , Tumor Hypoxia/genetics , Tumor Hypoxia/immunology
6.
Mol Cell Oncol ; 3(4): e1175538, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27652323

ABSTRACT

Targeting tumor glycolysis would hit the main energy source of cancer. We show that natural killer (NK) cells pursue this strategy by employing high mobility group box 1 (HMGB1) protein-a well-known proinflammatory cytokine-to specifically target glycolysis in cancer cells. This opens up new perspectives for cancer immunotherapy.

7.
Alcohol Clin Exp Res ; 40(10): 2094-2101, 2016 10.
Article in English | MEDLINE | ID: mdl-27581253

ABSTRACT

BACKGROUND: Chronic alcohol consumption is a risk factor for colorectal cancer. The mechanisms by which ethanol (EtOH) exerts its carcinogenic effect on the colorectal mucosa are not clear and may include oxidative stress with the action of reactive oxygen species (ROS) generated through EtOH metabolism via cytochrome P4502E1 (CYP2E1) leading to carcinogenic etheno-DNA adducts. ROS may also induce apoptosis. However, the effect of chronic EtOH consumption on CYP2E1, etheno-DNA adducts as well as anti-apoptotic proteins in the colorectal mucosa of heavy drinkers without colorectal inflammation is still not known. METHODS: Rectal biopsies from 32 alcoholics (>60 g EtOH/d) and from 12 controls (<20 g EtOH/d) were histologically examined, and immunohistochemistry for CYP2E1 and etheno-DNA adducts was performed. Apoptosis (cleaved PARP) as well as anti-apoptotic proteins including Bcl-xL , Bcl-2, and Mcl-1 were immunohistochemically determined. RESULTS: No significant difference in mucosal CYP2E1 or etheno-DNA adducts was observed between alcoholics and control patients. However, CYP2E1 and etheno-DNA adducts correlated significantly when both groups were combined (p < 0.001). In addition, although apoptosis was found not to be significantly affected by EtOH, the anti-apoptotic protein Mcl-1, but neither Bcl-xL nor Bcl-2, was found to be significantly increased in heavy drinkers as compared to controls (p = 0.014). CONCLUSIONS: Although colorectal CYP2E1 was not found to be significantly increased in alcoholics, CYP2E1 correlated overall with the level of etheno-DNA adducts in the colorectal mucosa, which identifies CYP2E1 as an important factor in colorectal carcinogenesis. Most importantly, however, is the up-regulation of the anti-apoptotic protein Mcl-1 in heavy drinkers counteracting apoptosis and possibly stimulating cancer development.


Subject(s)
Alcoholism/metabolism , Carcinogenesis/chemically induced , Cytochrome P-450 CYP2E1/metabolism , DNA Adducts/metabolism , Ethanol/toxicity , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Rectum/metabolism , Aged , Alcoholism/complications , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Case-Control Studies , Female , Humans , Male , Middle Aged , Rectum/drug effects
8.
Cell Death Dis ; 7(8): e2342, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27537525

ABSTRACT

Colorectal cancer (CRC) is the second most common malignant neoplasia in women and men worldwide. The B-cell lymphoma 2 (Bcl-2) protein family is mainly known for its pivotal role in the regulation of the mitochondrial death pathway. Anti-apoptotic Bcl-2 proteins may provide survival benefits and induce therapy resistance in cancer cells. Among anti-apoptotic Bcl-2 proteins, we found solely Bcl-xL strongly upregulated in human CRC specimens. In order to study protein function in the context of tumor initiation and progression in vivo, we generated a mouse model lacking Bcl-xL in intestinal epithelial cells (Bcl-xL(IEC-KO)). If challenged in an inflammation-driven tumor model, Bcl-xL(IEC-KO) mice showed a significantly reduced tumor burden with lower tumor numbers per animal and decreased tumor sizes. Analysis of cell death events by immunohistochemistry and immunoblotting revealed a striking increase of apoptosis in Bcl-xL-negative tumors. qRT-PCR and immunohistochemistry excluded changes in proliferative capacity and immune cell infiltration as reasons for the reduced tumor load and thereby identify apoptosis as key mechanism. Human CRC tissue was cultured ex vivo and treated with the small molecule compound ABT-737, which inhibits Bcl-xL and Bcl-2. Under ABT-737 treatment, the amount of apoptotic tumor cells significantly increased compared with controls, whereas proliferation levels remained unaltered. In summary, our findings identify Bcl-xL as a driver in colorectal tumorigenesis and cancer progression, making it a valuable target for clinical application.


Subject(s)
Colorectal Neoplasms/genetics , Oncogenes , bcl-X Protein/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biphenyl Compounds/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Disease Models, Animal , Enterocytes/drug effects , Enterocytes/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Inflammation/genetics , Inflammation/pathology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Knockout , Nitrophenols/pharmacology , Organ Specificity , Phenotype , Piperazines/pharmacology , Sulfonamides/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics , bcl-X Protein/metabolism
9.
Am J Med Genet A ; 170(6): 1502-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27016154

ABSTRACT

Intellectual disability (ID) with cerebellar ataxia comprises a genetically heterogeneous group of neurodevelopmental disorders. We identified a homozygous frameshift mutation in CWF19L1 (c.467delC; p.(P156Hfs*33)) by a combination of linkage analysis and Whole Exome Sequencing in a consanguineous Turkish family with a 9-year-old boy affected by early onset cerebellar ataxia and mild ID. Serial MRI showed mildly progressive cerebellar atrophy. Absent C19L1 protein expression in lymphoblastoid cell lines strongly suggested that c.467delC is a disease-causing alteration. One further pregnancy of the mother had been terminated at 22 weeks of gestation because of a small cerebellum and agenesis of corpus callosum. The homozygous CWF19L1 variant was also present in the fetus. Postmortem examination of the fetus in addition showed unilateral hexadactyly and vertebral malformations. These features have not been reported and may represent an expansion of the CWF19L1-related phenotypic spectrum, but could also be due to another, possibly autosomal recessive disorder. The exact function of the evolutionarily highly conserved C19L1 protein is unknown. So far, homozygous or compound heterozygous mutations in CWF19L1 have been identified in two Turkish siblings and a Dutch girl, respectively, affected by cerebellar ataxia and ID. A zebrafish model showed that CWF19L1 loss-of-function mutations result in abnormal cerebellar morphology and movement disorders. Our report corroborates that loss-of-function mutations in CWF19Ll lead to early onset cerebellar ataxia and (progressive) cerebellar atrophy. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Cycle Proteins/genetics , Cerebellum/abnormalities , Exome , High-Throughput Nucleotide Sequencing , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Mutation , Nervous System Malformations/diagnosis , Nervous System Malformations/genetics , Brain/abnormalities , Child , Comparative Genomic Hybridization , Consanguinity , Developmental Disabilities/diagnosis , Developmental Disabilities/genetics , Genetic Association Studies , Genetic Linkage , Humans , Magnetic Resonance Imaging , Male , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Radiography
10.
Nat Commun ; 7: 10764, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26948869

ABSTRACT

The high-mobility group box 1 (HMGB1) protein has a central role in immunological antitumour defense. Here we show that natural killer cell-derived HMGB1 directly eliminates cancer cells by triggering metabolic cell death. HMGB1 allosterically inhibits the tetrameric pyruvate kinase isoform M2, thus blocking glucose-driven aerobic respiration. This results in a rapid metabolic shift forcing cells to rely solely on glycolysis for the maintenance of energy production. Cancer cells can acquire resistance to HMGB1 by increasing glycolysis using the dimeric form of PKM2, and employing glutaminolysis. Consistently, we observe an increase in the expression of a key enzyme of glutaminolysis, malic enzyme 1, in advanced colon cancer. Moreover, pharmaceutical inhibition of glutaminolysis sensitizes tumour cells to HMGB1 providing a basis for a therapeutic strategy for treating cancer.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/physiopathology , HMGB1 Protein/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Death , Cell Line, Tumor , Cell Respiration , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Glucose/metabolism , Glycolysis , HMGB1 Protein/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
11.
Int J Oncol ; 46(2): 667-76, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25434832

ABSTRACT

The HMGB1 protein has multiple functions in tumor biology and can act both as a transcription factor and as a cytokine. HMGB1 is released during cell death, and in our previous studies we demonstrated that HMGB1 induces a distinct, necrosis-like cell death in glioblastoma. In epithelial malignant tumors such as colorectal cancer (CRC), the HMGB1-dependent effects show cross-talk with apoptotic signal transduction. Treatment of CRC cells with low concentrations of recombinant HMGB1 results in dose-dependent cytotoxicity which is morphologically characterized by the formation of giant mitochondria and does not share features of apoptosis. HMGB1-triggered cell death is associated with intracellular ROS release, and overexpression of Bcl-2 blocks both the increase of ROS as well as HMGB1-dependent cell death. Importantly, treatment with recombinant HMGB1 or overexpression of endogenous HMGB1 strongly sensitizes CRC cells to the cytotoxic activity of the pro-apoptotic death ligand TRAIL as well as the small molecule Bcl-2 family inhibitor ABT­737. Moreover, treatment of CRC cells with TRAIL or ABT­737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT­737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. Finally, we investigated the expression of HMGB1 in human CRC tumor samples and found that loss of HMGB1 expression is associated with a more aggressive phenotype and a more advanced stage of disease in patients with CRC. Altogether, our findings demonstrate a functional link between cytotoxic signaling cascades triggered by HMGB1 and pro-apoptotic agents leading to an HMGB1-dependent sensitization to CRC cell death. Thus, a further evaluation of recombinant HMGB1 as part of an experimental combination treatment of CRC seems warranted.


Subject(s)
Colonic Neoplasms/genetics , HMGB1 Protein/genetics , Recombinant Proteins/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Apoptosis/genetics , Apoptosis Regulatory Proteins/administration & dosage , Biphenyl Compounds/administration & dosage , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Glycyrrhizic Acid/administration & dosage , HCT116 Cells , HMGB1 Protein/administration & dosage , HMGB1 Protein/metabolism , Humans , Nitrophenols/administration & dosage , Piperazines/administration & dosage , Proto-Oncogene Proteins c-bcl-2 , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Sulfonamides/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/metabolism
12.
Neuro Oncol ; 14(1): 64-78, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22015597

ABSTRACT

The role of microglia, the brain-resident macrophages, in glioma biology is still a matter of debate. Clinical observations and in vitro studies in the mouse model indicate that microglia and macrophages that infiltrate the brain tumor tissue in high numbers play a tumor-supportive role. Here, we provide evidence that human microglia isolated from brain tumors indeed support tumor cell growth, migration, and invasion. However, after stimulation with the Toll-like receptor 3 agonist poly (I:C), microglia secrete factors that exerted toxic and suppressive effects on different glioblastoma cell lines, as assessed in cytotoxicity, migration, and tumor cell spheroid invasion assays. Remarkably, these effects were tumor-specific because the microglial factors impaired neither growth nor viability of astrocytes and neurons. Culture supernatants of tumor cells inhibited the poly (I:C) induction of this microglial M1-like, oncotoxic profile. Microglia stimulation before coculture with tumor cells circumvented the tumor-mediated suppression, as demonstrated by the ability to kill and phagocytose glioma cells. Our results show, for the first time to our knowledge, that human microglia exert tumor-supporting functions that are overridden by tumor-suppressing activities gained after poly (I:C) stimulation.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioma/metabolism , Microglia/physiology , Poly I-C/pharmacology , Toll-Like Receptor 3/agonists , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cells, Cultured , Coculture Techniques , Humans , Microglia/drug effects , Neoplasm Invasiveness
13.
Virchows Arch ; 458(3): 371-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21234762

ABSTRACT

The main cause of death from novel (swine origin) influenza A/H1N1 infection is acute respiratory distress syndrome. Most fatal cases are immunocompromised patients or patients with a severe underlying disease. Here, we report a fatal case of acute interstitial myocarditis associated with novel influenza A/H1N1 infection in an immunocompetent young woman. A previously healthy 18-year-old woman experienced malaise, diarrhea, and fever for several days prior to a sudden collapse at home. Autopsy revealed a predominantly lymphocytic myocarditis in the absence of a significant respiratory tract infection. Infection with novel (swine origin) influenza A/H1N1 was confirmed by PCR analysis of blood as well as myocardial tissue. Influenza-caused diarrhea with consecutive hypokalemia potentially contributed to the fatal outcome of the myocarditis, characterized by ventricular fibrillation. In conclusion, sudden death by myocarditis may be a rare complication of novel influenza A/H1N1 infection in otherwise healthy individuals, even in the absence of significant respiratory tract infection.


Subject(s)
Death, Sudden, Cardiac/etiology , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/virology , Myocarditis/pathology , Adolescent , DNA, Viral/analysis , Death, Sudden, Cardiac/pathology , Fatal Outcome , Female , Humans , Immunocompetence , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/pathology , Myocarditis/virology , Polymerase Chain Reaction
14.
Cancer Res ; 70(21): 8558-68, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20959471

ABSTRACT

Cells dying by necrosis release the high-mobility group box 1 (HMGB1) protein, which has immunostimulatory effects. However, little is known about the direct actions of extracellular HMGB1 protein on cancer cells. Here, we show that recombinant human HMGB1 (rhHMGB1) exerts strong cytotoxic effects on malignant tumor cells. The rhHMGB1-induced cytotoxicity depends on the presence of mitochondria and leads to fast depletion of mitochondrial DNA, severe damage of the mitochondrial proteome by toxic malondialdehyde adducts, and formation of giant mitochondria. The formation of giant mitochondria is independent of direct nuclear signaling events, because giant mitochondria are also observed in cytoplasts lacking nuclei. Further, the reactive oxygen species scavenger N-acetylcysteine as well as c-Jun NH(2)-terminal kinase blockade inhibited the cytotoxic effect of rhHMGB1. Importantly, glioblastoma cells, but not normal astrocytes, were highly susceptible to rhHMGB1-induced cell death. Systemic treatment with rhHMGB1 results in significant growth inhibition of xenografted tumors in vivo. In summary, rhHMGB1 induces a distinct form of cell death in cancer cells, which differs from the known forms of apoptosis, autophagy, and senescence, possibly representing an important novel mechanism of specialized necrosis. Further, our findings suggest that rhHMGB1 may offer therapeutic applications in treatment of patients with malignant brain tumors.


Subject(s)
Apoptosis , Glioblastoma/pathology , HMGB1 Protein/metabolism , Mitochondria/pathology , Acetylcysteine/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Electrophoresis, Gel, Two-Dimensional , Female , Fluorescent Antibody Technique , Free Radical Scavengers/pharmacology , Glioblastoma/drug therapy , Glioblastoma/metabolism , HMGB1 Protein/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Nude , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Necrosis , Proteome/analysis , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Cells, Cultured
15.
Hepatology ; 52(6): 2023-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20979053

ABSTRACT

UNLABELLED: The A kinase anchor protein 12 (AKAP12) is a central mediator of protein kinase A and protein kinase C signaling. Although AKAP12 has been described to act as a tumor suppressor and its expression is frequently down-regulated in several human malignancies, the underlying molecular mechanisms responsible for the AKAP12 reduction are poorly understood. We therefore analyzed the expression of AKAP12 and its genetic and epigenetic regulatory mechanisms in human hepatocarcinogenesis. Based on tissue microarray analyses (n = 388) and western immunoblotting, we observed a significant reduction of AKAP12 in cirrhotic liver (CL), premalignant lesions (DN), and hepatocellular carcinomas (HCCs) compared to histologically normal liver specimens (NL). Analyses of array comparative genomic hybridization data (aCGH) from human HCCs revealed chromosomal losses of AKAP12 in 36% of cases but suggested additional mechanisms underlying the observed reduction of AKAP12 expression in hepatocarcinogenesis. Quantitative methylation analysis by MassARRAY of NL, CL, DN, and HCC tissues, as well as of various tumorigenic and nontumorigenic liver cell lines revealed specific hypermethylation of the AKAP12α promoter but not of the AKAP12ß promoter in HCC specimens and in HCC cell lines. Consequently, restoration experiments performed with 5-aza-2'deoxycytidine drastically increased AKAP12α mRNA levels in a HCC cell line (AKN1) paralleled by AKAP12α promoter demethylation. As hypermethylation is not observed in CL and DN, we investigated microRNA-mediated posttranscriptional regulation as an additional mechanism to explain reduced AKAP12 expression. We found that miR-183 and miR-186 are up-regulated in CL and DN and are able to target AKAP12. CONCLUSION: In addition to genetic alterations, epigenetic mechanisms are responsible for the reduction of the tumor suppressor gene AKAP12 in human hepatocarcinogenesis.


Subject(s)
A Kinase Anchor Proteins/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins/genetics , Epigenesis, Genetic , Liver Neoplasms/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Decitabine , Down-Regulation , Humans , Liver Neoplasms/genetics , MicroRNAs/metabolism , Promoter Regions, Genetic/physiology , Protein Array Analysis
16.
Clin Cancer Res ; 16(10): 2715-28, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20442299

ABSTRACT

PURPOSE: Stem-like tumor cells comprise a highly tumorigenic and therapy-resistant tumor subpopulation, which is believed to substantially influence tumor initiation and therapy resistance in glioma. Currently, therapeutic, drug-induced differentiation is considered as a promising approach to eradicate this tumor-driving cell population; retinoic acid is well known as a potent modulator of differentiation and proliferation in normal stem cells. In glioma, knowledge about the efficacy of retinoic acid-induced differentiation to target the stem-like tumor cell pool could have therapeutic implications. EXPERIMENTAL DESIGN: Stem-like glioma cells (SLGC) were differentiated with all-trans retinoic acid-containing medium to study the effect of differentiation on angiogenesis, invasive growth, as well as radioresistance and chemoresistance of SLGCs. In vivo effects were studied using live microscopy in a cranial window model. RESULTS: Our data suggest that in vitro differentiation of SLGCs induces therapy-sensitizing effects, impairs the secretion of angiogenic cytokines, and disrupts SLGCs motility. Further, ex vivo differentiation reduces tumorigenicity of SLGCs. Finally, we show that all-trans retinoic acid treatment alone can induce antitumor effects in vivo. CONCLUSIONS: Altogether, these results highlight the potential of differentiation treatment to target the stem-like cell population in glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Differentiation/drug effects , Glioma/drug therapy , Neoplastic Stem Cells/drug effects , Tretinoin/pharmacology , Animals , Blotting, Western , Brain Neoplasms/pathology , Cell Separation , Flow Cytometry , Glioma/pathology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred NOD , Neoplastic Stem Cells/pathology , Polymerase Chain Reaction , Xenograft Model Antitumor Assays
17.
Mol Carcinog ; 49(2): 175-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19852062

ABSTRACT

Approximately 15% of small intestinal adenocarcinomas show inactivation of DNA-mismatch repair (MMR) and display high-level microsatellite instability (MSI-H). MSI-H tumors progress as a result of mutations affecting coding microsatellites (coding microsatellite instability, cMSI) that may result in a functional inactivation of the encoded proteins and provide a selective growth advantage for the affected cell. To investigate the cMSI selection in small intestinal carcinogenesis 56 adenocarcinomas were tested for MSI. Eleven MSI-H carcinomas (19.6%) were identified and subjected to cMSI analysis in 24 potentially tumor relevant genes. Mutation frequencies were similar to those observed in colorectal cancer (CRC). Beside high frequencies of cMSI in TGFbetaR2, ACVR2, and AIM2 we detected MARCKS mutations in 10 out of 11 (91%) tumors with a 30% share of biallelic mutations. Since little is known about MARCKS expression in the intestine, we analyzed MARCKS protein expression in 31 carcinomas. In non-neoplastic mucosa, MARCKS was found to be expressed with a concentration gradient along the crypt-villus axis. In line with cMSI induced functional inactivation of MARCKS, 8 out of 11 MSI-H adenocarcinomas showed regional or complete loss of the protein. In microsatellite stable (MSS) small bowel adenocarcinoma, loss of MARCKS expression was seen in 2 out of 20 tumors (10%). In conclusion, we herein present a cMSI profile of MSI-H small intestinal adenocarcinomas identifying MARCKS as a frequent target of mutation. Loss of MARCKS protein expression suggests a significant role of MARCKS inactivation in the pathogenesis of small intestinal adenocarcinomas.


Subject(s)
Adenocarcinoma/genetics , Intestinal Neoplasms/genetics , Intestine, Small/pathology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Microsatellite Repeats/genetics , DNA Methylation , Humans , Immunohistochemistry , Mutation , Myristoylated Alanine-Rich C Kinase Substrate , Promoter Regions, Genetic
18.
Int J Oncol ; 35(1): 149-58, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19513562

ABSTRACT

Chemotherapy of non-Hodgkin's lymphoma is frequently hampered by drug resistance. The monoclonal antibody rituximab specifically targets the CD20 antigen and sensitizes B-cell lymphoma cells to standard anticancer drugs. In the present investigation, we analyzed, whether a combination of rituximab and artesunate may act in a complementary manner and eventually synergize in tumor cell killing. Artesunate is an anti-malarial drug, which also exerts profound activity towards cancer cells. While rituximab alone was minimally cytotoxic, rituximab increased cytotoxicity to artesunate in Ramos cells. Artesunate induced apoptosis, induced Fas/CD95 expression and the formation of reactive oxygen species (ROS) and resulted in a breakdown of mitochondrial membrane potential. This argues for the involvement of both receptor-driven extrinsic and mitochondrial intrinsic routes of apoptosis. Rituximab increased Fas/CD95 expression and ROS formation and decreased mitochondrial membrane potential ultimately leading to increased apoptosis induced by artesunate. The transcription factors YY1 and Sp1 are upstream regulators of apoptosis by controlling the expression of apoptosis-regulating genes. YY1 and Sp1 were down-regulated and Fas/CD95 was up-regulated by rituximab and artesunate indicating that artesunate activated the Fas/CD95 pathway and that rituximab increased the susceptibility of tumor cells to artesunate-induced apoptosis. Furthermore, rituximab affected the expression of antioxidant genes. The antibody decreased artesunate-induced up-regulation of catalase expression and increased artesunate-induced down-regulation of glutathione S-transferase-phi expression. Manganese-dependent superoxide dismutase expression was not changed by artesunate. Antioxidant proteins may help to detoxify artesunate-induced ROS. Rituximab reversed the artesunate-induced expression changes of antioxidant genes and, hence, reduced the detoxification capacity of Ramos cells. The effects of rituximab on antioxidant genes represent a novel mechanism of rituximab for chemosensitization.


Subject(s)
Antigens, CD20/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Lymphoma, B-Cell/pathology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Antimalarials/administration & dosage , Artemisinins/administration & dosage , Artesunate , BH3 Interacting Domain Death Agonist Protein/metabolism , Catalase/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glutathione S-Transferase pi/metabolism , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Rituximab , Sp1 Transcription Factor/metabolism , Superoxide Dismutase/metabolism , YY1 Transcription Factor/metabolism , fas Receptor/metabolism
19.
Cancer Res ; 69(6): 2234-43, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19258502

ABSTRACT

Dynamic instability of the microtubule network modulates processes such as cell division and motility, as well as cellular morphology. Overexpression of the microtubule-destabilizing phosphoprotein stathmin is frequent in human malignancies and represents a promising therapeutic target. Although stathmin inhibition gives rise to antineoplastic effects, additional and functionally redundant microtubule-interacting proteins may attenuate the efficiency of this therapeutic approach. We have systematically analyzed the expression and potential protumorigenic effects of stathmin family members in human non-small cell lung cancer (NSCLC). Both stathmin and stathmin-like 3 (SCLIP) were overexpressed in adenocarcinoma as well as squamous cell carcinoma (SCC) tissues and induced tumor cell proliferation, migration, and matrix invasion in respective cell lines. Accordingly, reduced stathmin and SCLIP levels affected cell morphology and were associated with a less malignant phenotype. Combined inhibition of both factors caused additive effects on tumor cell motility, indicating partial functional redundancy. Because stathmin and SCLIP expression significantly correlated in NSCLC tissues, we searched for common upstream regulators and identified the far upstream sequence element-binding protein-1 (FBP-1) as a pivotal inducer of several stathmin family members. Our results indicate that the coordinated overexpression of microtubule-destabilizing factors by FBP-1 is a critical step to facilitate microtubule dynamics and subsequently increases proliferation and motility of tumor cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Lung Neoplasms/metabolism , Stathmin/biosynthesis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Growth Processes/physiology , Cell Movement/physiology , Cell Survival/physiology , DNA Helicases/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Invasiveness , RNA, Small Interfering/genetics , RNA-Binding Proteins , Stathmin/genetics , Transfection
20.
Apoptosis ; 13(3): 437-47, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18188704

ABSTRACT

The HIPPI (HIP-1 protein interactor) protein is a multifunctional protein that is involved in the regulation of apoptosis. The interaction partners of HIPPI include HIP-1 (Huntingtin-interacting protein-1), Apoptin, Homer1c, Rybp/DEDAF, and BAR (bifunctional apoptosis regulator). In search for other binding partners of HIPPI, we performed a yeast two hybrid screen and identified BLOC1S2 (Biogenesis of lysosome-related organelles complex-1 subunit 2) as a novel HIPPI-interacting protein. In co-immunoprecipitation assays, BLOC1S2 specifically associates with HIPPI, but not with HIP-1. To study the expression of BLOC1S2 on the protein level, we generated a mouse monoclonal antibody specific for BLOC1S2 and a multiple tissue array comprising 70 normal and cancer tissue samples of diverse origin. BLOC1S2 protein is widely expressed in normal tissue as well as in malignant tumors with a tendency towards lower expression levels in certain subtypes of tumors. On the subcellular level, BLOC1S2 is expressed in an organellar-like pattern and co-localizes with mitochondria. Over-expression of BLOC1S2 in the presence or absence of HIPPI does not induce apoptosis. However, BLOC1S2 and HIPPI sensitize NCH89 glioblastoma cells to the pro-apoptotic actions of staurosporine and the death ligand TRAIL by enhancing caspase activation, cytochrome c release, and disruption of the mitochondrial membrane potential. Given its interaction with HIPPI and its pro-apoptotic activity, BLOC1S2 might play an important functional role in cancer and neurodegenerative diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis/drug effects , Glioblastoma/pathology , Proteins/physiology , Adult , Amino Acid Sequence , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Female , HeLa Cells , Humans , Immunohistochemistry , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Molecular Sequence Data , Neoplasms/pathology , Protein Binding , Sequence Alignment , Staurosporine/pharmacology , TNF-Related Apoptosis-Inducing Ligand/physiology , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...